Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Circulation ; 104(21): 2588-94, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11714655

ABSTRACT

BACKGROUND: Diabetes is associated with increased risk of mortality as a consequence of acute myocardial infarction. This study determined whether rosiglitazone (ROSI) could reduce myocardial infarction after ischemia/reperfusion injury. METHODS AND RESULTS: Male Lewis rats were anesthetized, and the left anterior descending coronary artery was ligated for 30 minutes. After reperfusion for 24 hours, the ischemic and infarct sizes were determined. ROSI at 1 and 3 mg/kg IV reduced infarct size by 30% and 37%, respectively (P<0.01 versus vehicle). Pretreatment with ROSI (3 mg. kg(-1). d(-1) PO) for 7 days also reduced infarct size by 24% (P<0.01). ROSI also improved ischemia/reperfusion-induced myocardial contractile dysfunction. Left ventricular systolic pressure and positive and negative maximal values of the first derivative of left ventricular pressure (dP/dt) were significantly improved in ROSI-treated rats. ROSI reduced the accumulation of neutrophils and macrophages in the ischemic heart by 40% and 43%, respectively (P<0.01). Ischemia/reperfusion induced upregulation of CD11b/CD18 and downregulation of L-selectin on neutrophils and monocytes; these effects were significantly attenuated in ROSI-treated animals. Likewise, intercellular adhesion molecule-1 expression in ischemic hearts was markedly diminished by ROSI, as was the ischemia/reperfusion-stimulated upregulation of monocyte chemoattractant protein-1. CONCLUSIONS: ROSI reduced myocardial infarction and improved contractile dysfunction caused by ischemia/reperfusion injury. The cardioprotective effect of ROSI was most likely due to inhibition of the inflammatory response.


Subject(s)
Hypoglycemic Agents/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/complications , Receptors, Cytoplasmic and Nuclear/agonists , Thiazoles/therapeutic use , Thiazolidinediones , Transcription Factors/agonists , Animals , CD18 Antigens/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Diabetes Complications , Hypoglycemic Agents/pharmacology , Macrophage-1 Antigen/metabolism , Macrophages/immunology , Male , Monocytes/immunology , Myocardial Contraction/drug effects , Myocardial Infarction/etiology , Myocardial Infarction/immunology , Neutrophil Infiltration/drug effects , Neutrophils/immunology , RNA, Messenger/biosynthesis , Rats , Rats, Inbred Lew , Rosiglitazone , Thiazoles/pharmacology
2.
J Cardiovasc Pharmacol ; 36(2): 277-81, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10942172

ABSTRACT

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


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Antioxidants/pharmacology , Carbazoles/pharmacology , Metoprolol/pharmacology , Propanolamines/pharmacology , Animals , Carvedilol , Cell Membrane/drug effects , Cells, Cultured , Free Radicals/chemistry , Heart/drug effects , Lipid Peroxidation/drug effects , Neurons/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Thiobarbituric Acid Reactive Substances/metabolism
3.
Eur J Biochem ; 267(3): 919-26, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10651831

ABSTRACT

Murine MARCO has been identified recently in subsets of macrophages located in the peritoneum, marginal zone of the spleen, and the medullary cord of lymph nodes, where it has been proposed that it serves as a bacteria-binding receptor. A scavenger receptor family member with an extended collagenous domain, murine MARCO has also been demonstrated in atherosclerotic lesions of susceptible mice. We report here the identification, tissue and chromosomal localization, and pharmacological characterization of human (h)MARCO. hMARCO was identified from a macrophage cDNA library by electronic screening with the murine MARCO sequence. Nucleotide sequence analysis confirmed that the full-length hMARCO clone encoded a 519-amino acid protein sharing 68.5% identity with murine MARCO. RNA blot analysis indicated that the hMARCO transcript is 2.0 kb in length and is predominantly expressed in human lung, liver, and lymph nodes. Radiation hybrid mapping localized hMARCO to chromosome 2q14. Ligand-binding studies of COS cells expressing hMARCO demonstrated significant specific binding of both Escherichia coli and Staphylococcus aureus. In contrast, the hMARCO receptor expressed in COS cells did not specifically bind the scavenger receptor ligand acetylated low-density lipoprotein (LDL), despite its similarity to the elongated collagen-like binding domain of the macrophage scavenger receptor. In addition, acetylated (Ac)LDL and oxidized (Ox)LDL did not inhibit E. coli binding to hMARCO. These data suggest that hMARCO may play an important role in host defense, but it has no obvious role in the accumulation of modified lipoproteins during atherogenesis.


Subject(s)
Macrophages/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Amino Acid Sequence , Animals , Bacterial Adhesion , Base Sequence , COS Cells , Chromosome Mapping , Chromosomes, Human, Pair 2/genetics , Cloning, Molecular , DNA Primers/genetics , Escherichia coli/immunology , Female , Humans , Macrophages/immunology , Macrophages/microbiology , Male , Mice , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Species Specificity , Tissue Distribution
4.
FEBS Lett ; 462(1-2): 145-50, 1999 Nov 26.
Article in English | MEDLINE | ID: mdl-10580108

ABSTRACT

Oxidized low-density lipoprotein (OxLDL) plays a critical role in foam cell formation and atherosclerogenesis. A cDNA encoding adipophilin was identified in cultured human macrophages stimulated with OxLDL using mRNA differential display. Adipophilin is a 50 kDa protein known to be a specific marker for adipocyte cell differentiation and lipid accumulation in a variety of cells. The time-dependent induction of adipophilin mRNA in macrophages was specific to OxLDL but not native LDL, and not to various cytokines and serum. In human atherosclerotic lesions, adipophilin mRNA expression was localized in a subset of lipid-rich macrophages. These data suggest that adipophilin-expressing macrophages may represent foam cells and this gene expression is likely to be associated with the lipid accumulation in foam cells of the atherosclerotic lesions.


Subject(s)
Arteriosclerosis/metabolism , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Peptides/metabolism , Arteriosclerosis/pathology , Blood Proteins/metabolism , Cells, Cultured , Cytokines/metabolism , Humans , In Situ Hybridization , Membrane Proteins , Peptides/genetics , Perilipin-2 , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Up-Regulation
5.
Mol Cell Biochem ; 197(1-2): 179-85, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10485337

ABSTRACT

Calcitonin gene-related peptide (CGRP) is a neuropeptide with diverse biological properties including potent vasodilating activity. Recently, we reported the cloning of complementary DNAs (cDNAs) encoding the human and porcine CGRP receptors which share significant amino acid sequence homology with the human calcitonin receptor, a member of the recently described novel subfamily of G-protein-coupled 7TM receptors. Activation of this family of receptors has been shown to result in an increase in intracellular cAMP accumulation and calcium release. In this study, we demonstrate that HEK-293 cells expressing recombinant CGRP receptors (HEK-293HR or PR) respond to CGRP with increased intracellular calcium release (EC50 = 1.6 nM) in addition to the activation of adenylyl cyclase (EC50 = 1.4 nM). The effect of CGRP on adenylyl cyclase activation and calcium release was inhibited by CGRP (8-37), a CGRP receptor antagonist. Both effects were mediated by cholera toxin-sensitive G-proteins, but these two signal transduction pathways were independent of each other. While cholera toxin pretreatment of HEK-293PR cells resulted in permanent activation of adenylyl cyclase, the same pretreatment resulted in an inhibition of CGRP-mediated [Ca2+]i release. Pertussis toxin was without effect on CGRP-mediated responses. In addition, CGRP-mediated calcium release appears to be due to release from a thapsigargin-sensitive intracellular calcium pool. These results show that the recombinant human as well as porcine CGRP receptor can independently increase both cAMP production and intracellular calcium release when stably expressed in the HEK-293 cell line.


Subject(s)
Calcium Signaling/physiology , Cyclic AMP/metabolism , Receptors, Calcitonin Gene-Related Peptide/physiology , Adenylyl Cyclases/metabolism , Animals , Calcium Channel Blockers/pharmacology , Cell Line , Cholera Toxin/pharmacology , Enzyme Activation , GTP-Binding Protein alpha Subunits, Gs/metabolism , Humans , Inositol Phosphates/biosynthesis , Nifedipine/pharmacology , Recombinant Proteins/metabolism , Swine , Thapsigargin/pharmacology , Type C Phospholipases/metabolism
6.
J Leukoc Biol ; 66(1): 135-43, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10411001

ABSTRACT

Chemoattraction of monocytes by the CXC chemokine stromal cell-derived factor-1alpha (SDF-1alpha) and its receptor CXCR4 may be involved in vascular diseases like atherosclerosis. We studied the regulation of CXCR4 transcription and SDF-1-induced functional responses in human monocytes during their differentiation in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF), oxidized low-density lipoprotein (Ox-LDL), and unmodified LDL. Our results reveal that the rapid decline of SDF-1-mediated [Ca2+]i influx after monocyte isolation is followed by a gradual functional restoration and a concomitant reexpression of CXCR4 mRNA over time. A further three- to fourfold induction of CXCR4 mRNA occurred in macrophage-derived foam cells on treatment with Ox-LDL. HL-60 cells induced with phorbol myristate acetate (PMA) showed a rapid fourfold stimulation of CXCR4 mRNA within 1 h, declining to barely detectable levels at 3 h, with eventual restoration over time, mirroring the expression pattern in monocytes. Surface expression of CXCR4 is maintained in HL-60 cells during PMA-induced differentiation, as demonstrated by flow cytometry. GM-CSF had no effect on CXCR4 mRNA in HL-60 cells and does not cause its down-regulation in human macrophages.


Subject(s)
Chemokines, CXC/metabolism , Macrophages/cytology , Monocytes/cytology , Receptors, CXCR4/biosynthesis , Cell Differentiation , Cell Line , Chemokine CXCL12 , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , HL-60 Cells , Humans , K562 Cells , Leukemia, Myeloid , Lipoproteins, LDL/metabolism , Mitogens/pharmacology , Receptors, CXCR4/genetics , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Transcription, Genetic , U937 Cells , Up-Regulation
7.
Brain Res Mol Brain Res ; 68(1-2): 29-41, 1999 May 07.
Article in English | MEDLINE | ID: mdl-10320781

ABSTRACT

Active neuronal-glial interaction is important in the maintenance of brain homeostasis and is vital for neuronal survival following brain injury. The time course of post-ischemic astroglial dysfunction and neuronal death was studied in the spontaneously hypertensive rat (SHR) brain following permanent middle cerebral artery occlusion (MCAO). In situ hybridization with 35S-labeled riboprobes for GFAP and GLUT3 was used to monitor mRNA expression in glia and neurons. Astrocytic proteins GFAP, vimentin, S100, Glutathione-S-Transferase Yb (GST Yb) and neuronal protein TG2 were detected by immunofluorescence. Cells were co-stained with in situ end labeling (ISEL) to detect DNA fragmentation, a hallmark of cell death. GFAP mRNA expression declined rapidly in the ischemic region of the cortex and was almost absent by 12 h. Immunohistochemical studies revealed a parallel decline in the corresponding protein: a reduction in GFAP staining was apparent in the infarct after 3 h and by 24 h, there was essentially no remaining GFAP. Three other glial proteins (vimentin, S100 and GST Yb) disappeared from infarct over a similar time course. A few ISEL positive cells were observed in the infarct at 6 h, but maximal detection was not seen until 24-48 h. Most of the ISEL-positive cells were neurons, identified by co-staining with the neuronal marker TG2. Few cells expressing GFAP or other glial markers were positive at any time point. Neuronal GLUT3 mRNA declined more slowly than GFAP mRNA in the ischemic core and disappeared during the period of neuronal death. Concurrent with the loss of GFAP mRNA and protein expression in the infarct, there was a rapid rise in GFAP mRNA in the peri-infarct region of ipsilateral hemisphere and proximal region of the contralateral hemisphere. This was followed by the enhanced GFAP protein expression characteristic of reactive astrocytes, but over a significantly slower time course. These studies show that MCAO leads to a rapid decline of GFAP mRNA and glial proteins, which appears to precede the decline in neuronal mRNA and neuronal death within the infarct. Early astroglial dysfunction may play a critical role in determining the outcome of acute hypoxic-ischemic injury by compromising neuronal-glial interactions.


Subject(s)
Astrocytes/pathology , Hypertension/pathology , Ischemic Attack, Transient/pathology , Neurons/pathology , Animals , Cell Death/physiology , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/analysis , Glutathione Transferase/analysis , Immunohistochemistry , In Situ Hybridization , Male , Protein Glutamine gamma Glutamyltransferase 2 , Rats , Rats, Inbred SHR , S100 Proteins/analysis , Time Factors , Vimentin/analysis
8.
J Pharmacol Exp Ther ; 289(3): 1277-85, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10336517

ABSTRACT

Class A scavenger receptor (SR-A) antagonists may prevent the initiation of atherosclerosis, because a recent report found that SR-A/apolipoprotein E (apoE) double-knockout mice had 60% smaller lesions than apoE single-knockout littermates. We transfected human embryonic kidney (HEK) 293 cells with SR-A type I or II receptors to find small-molecule antagonists. Uptake of 1,1'-dioctadecyl-3,3,3', 3'-tetramethylindocarbocyanine perchlorate-labeled acetylated low-density lipoprotein (DiI-AcLDL) showed that among common polyanionic ligands, polyinosine was the most potent, with an IC50 of 0.74 microgram/ml, whereas the novel compound (E)-methyl 4-chloro-alpha-[4-(4-chlorophenyl)-1, 5-dihydro-3-hydroxy-5-oxo-1-(2-thiazolyl)-2H-pyrrol-2-ylidene]benzene acetate gave an IC50 of 6.1 microgram/ml (13 microM). The novel antagonist also inhibited DiI-AcLDL uptake in cultured human peripheral and rat peritoneal macrophages with IC50 values of 21 microM and 17 microM, respectively. With [125I]AcLDL as ligand for transfected HEK 293 cells, binding/uptake and degradation at 37 degrees C for 5 h was saturable and selective. In a comparison of both types of receptor, we found no difference between the capacity of SR-AI or SR-AII for either binding or degradation. Polyinosine competed both [125I]AcLDL binding and degradation with a Ki of 1 microgram/ml, whereas the novel antagonist competed with a Ki of 19 microgram/ml (40 microM) and 8.6 microgram/ml (18 microM), respectively, for binding and degradation. Saturation binding in the presence of the ionophore monensin indicated that the novel compound behaved as a noncompetitive antagonist and perhaps as an allosteric effector. This is the first report to describe a small-molecule macrophage scavenger receptor antagonist. Utilization of this permanently transfected HEK 293 cell line will allow the identification of more potent macrophage scavenger receptor antagonists, so that their utility as therapeutics for atherosclerosis can be determined.


Subject(s)
Macrophages, Peritoneal/physiology , Macrophages/physiology , Receptors, Immunologic/antagonists & inhibitors , Animals , Binding, Competitive , Biological Transport/drug effects , Cell Line , Cells, Cultured , Cloning, Molecular , Dextran Sulfate/pharmacology , Humans , Kinetics , Ligands , Lipoproteins, LDL/pharmacokinetics , Macrophages/drug effects , Macrophages, Peritoneal/drug effects , Mice , Mice, Knockout , Phenylacetates/pharmacology , Poly I/pharmacology , Rats , Receptors, Immunologic/genetics , Receptors, Immunologic/physiology , Receptors, Scavenger , Recombinant Proteins/antagonists & inhibitors , Scavenger Receptors, Class A , Thiazoles/pharmacology , Transfection
9.
J Vasc Res ; 36(2): 120-5, 1999.
Article in English | MEDLINE | ID: mdl-10213907

ABSTRACT

Bone morphogenetic proteins (BMPs) and their serine/threonine kinase receptors have been identified in atherosclerotic arteries and vascular smooth muscle cells, respectively. Thus, BMPs (the largest subfamily of the TGF-beta superfamily) have been implicated in the pathogenesis of atherosclerosis. However, the origins of BMP biosynthesis and the functional roles of BMP in blood vessels are unclear. The present study explored BMP-2 gene expression in various human blood vessels and vascular cell types. Functional in vitro studies were also performed to determine the effects of recombinant human BMP-2 on migration (transwell assay) and proliferation ([3H]-thymidine incorporation) of human aortic vascular smooth muscle cells (HASMC). RT-PCR experiments revealed BMP-2 gene expression in normal and atherosclerotic human arteries as well as cultured human aortic and coronary vascular smooth muscle cells, human umbilical vein endothelial cells (HUVECs) and human macrophages. In cellular migration studies, incubation with BMP-2 produced efficacious (

Subject(s)
Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/pharmacology , Gene Expression/physiology , Muscle, Smooth, Vascular/drug effects , Transforming Growth Factor beta , Aorta/cytology , Aorta/drug effects , Aorta/physiology , Bone Morphogenetic Protein 2 , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Drug Synergism , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Platelet-Derived Growth Factor/pharmacology , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/physiology
10.
J Neurochem ; 71(3): 1194-204, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9721745

ABSTRACT

Focal cerebral ischemia elicits local inflammatory reaction as demonstrated by the accumulation of inflammatory cells and mediators in the ischemic brain. Interferon-inducible protein-10 (IP-10) is a member of the C-X-C chemokine family that possesses potent chemoattractant actions for monocytes, T cells, and smooth muscle cells. To investigate a potential role of IP-10 in focal stroke, we studied the temporal expression of IP-10 mRNA after occlusion of the middle cerebral artery in rat by means of northern analysis. IP-10 mRNA expression after focal stroke demonstrated a unique biphasic profile, with a marked increase early at 3 h (4.9-fold over control; p < 0.01), a peak level at 6 h (14.5-fold; p < 0.001) after occlusion of the middle cerebral artery, and a second wave induction 10-15 days after ischemic injury (7.2- and 9.3-fold increase for 10 and 15 days, respectively; p < 0.001). In situ hybridization confirmed the induced expression of IP-10 mRNA and revealed its spatial distribution after focal stroke. Immunohistochemical studies demonstrated the expression of IP-10 peptide in neurons (3-12 h) and astroglial cells (6 h to 15 days) of the ischemic zone. To explore further the potential role of IP-10 in focal stroke, we demonstrated a dose-dependent chemotactic action of IP-10 on C6 glial cells and enhanced attachment of rat cerebellar granule neurons. Taken together, the data suggest that ischemia induces IP-10, which may play a pleiotropic role in prolonged leukocyte recruitment, astrocyte migration/activation, and neuron attachment/sprouting after focal stroke.


Subject(s)
Arterial Occlusive Diseases/complications , Brain Ischemia/etiology , Brain Ischemia/metabolism , Cerebral Arteries , Cerebral Cortex/metabolism , Chemokines, CXC/metabolism , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , Cerebellum/cytology , Cerebellum/drug effects , Chemokine CXCL10 , Chemokines, CXC/genetics , Immunohistochemistry , Interleukin-1/genetics , Neurons/drug effects , Neurons/physiology , RNA, Messenger/metabolism , Rats , Tissue Distribution , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics
11.
J Biol Chem ; 273(7): 4282-7, 1998 Feb 13.
Article in English | MEDLINE | ID: mdl-9461627

ABSTRACT

Chemokines play an important role in the regulation of endothelial cell (EC) function, including proliferation, migration and differentiation during angiogenesis, and re-endothelialization after injury. In this study, reverse transcriptase-polymerase chain reaction was used to reveal expression of various CXC and CC chemokine receptors in human umbilical vein EC. Northern analysis showed that CXCR4 was selectively expressed in vascular EC, but not in smooth muscle cells. Compared with other chemokines, stromal cell-derived factor-1alpha (SDF-1alpha), the known CXCR4 ligand, was an efficacious chemoattractant for EC, causing the migration of approximately 40% input cells with an EC50 of 10-20 nM. Of the chemokines tested, only SDF-1alpha induced a rapid, though variable mobilization of intracellular Ca2+ in EC. Experiments with actinomycin D demonstrated that CXCR4 transcripts were short-lived, indicating a rapid mRNA turnover. Interferon-gamma (IFN-gamma) caused a pronounced down-regulation of CXCR4 mRNA in a concentration- and time-dependent manner. In a striking functional correlation, IFN-gamma treatment also attenuated the chemotactic response of EC to SDF-1alpha. IL-1beta, tumor necrosis factor-alpha, and lipopolysaccharide produced a time course-dependent biphasic effect on CXCR4 transcription. Expression of CXCR4 in EC is significant, more so as it and several CC chemokine receptors have been shown to serve as fusion co-receptors along with CD4 during human immunodeficiency virus infection. Taken together, these findings provide evidence of chemokine receptor expression in EC and offer an explanation for the action of chemokines like SDF-1alpha on the vascular endothelium.


Subject(s)
Chemokines/pharmacology , Endothelium, Vascular/drug effects , Receptors, Chemokine/physiology , Calcium/metabolism , Cell Movement/drug effects , Cells, Cultured , Chemokine CXCL12 , Chemokines, CXC/pharmacology , Dactinomycin/pharmacology , Flow Cytometry , Gene Expression Regulation/genetics , Humans , Interferon-gamma/pharmacology , Muscle, Smooth/drug effects , RNA, Messenger/analysis , Receptors, CXCR4/metabolism , Transcription, Genetic/genetics , Tumor Necrosis Factor-alpha/pharmacology , Umbilical Veins
12.
Biochem Pharmacol ; 56(12): 1645-56, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9973186

ABSTRACT

Carvedilol is a vasodilating beta-blocker and antioxidant approved for treatment of mild to moderate hypertension, angina, and congestive heart failure. SB 211475 (4-[2-hydroxyl-3-[[2-(2-methoxyphenoxy)ethyl]amino]propoxyl]-9H-++ +carbazol-3-ol), a hydroxylated carvedilol analogue, is an even more potent antioxidant in several assay systems. Carvedilol also has neuroprotective capacity with modulatory actions at N-methyl-D-aspartate (NMDA) receptors and Na+ channels. In the present study, we demonstrated that in cultured rat cerebellar neurons, SB 211475 has 28-fold greater antioxidant activity than carvedilol, but is 2- to 6-fold less potent, respectively, at inhibiting neurotoxic activities at Na+ channels and at NMDA receptor channels. To determine a biophysical rationale for these differential activities, small angle x-ray scattering data were obtained from model lipid and brain membrane bilayers containing either carvedilol, SB 211475, or dihydropyridine calcium channel blockers. Electron density profiles revealed that the location of SB 211475 was restricted to the glycerol backbone/hydrocarbon interface and significantly reduced membrane width by 5%, whereas the time-averaged location for carvedilol and flunarizine also extended to the hydrated surface of the bilayer. Comparison of carvedilol with several dihydropyridines showed a correlation between high ClogP values (lipophilicity), Na+ channel inhibitory potency, and bilayer localization. The antioxidant activity of SB 211475 could be explained by restricted intercalation into the glycerol phosphate/hydrocarbon interface, creating an increase in volume associated with the phospholipid acyl chains, which would then become resistant to lipid peroxidation. Differential channel modulation may also be explained by these membrane structural results, which indicate that carvedilol and the less spatially restricted dihydropyridine molecules are more likely to inhibit transmembrane receptor channels.


Subject(s)
Carbazoles/pharmacology , Cell Membrane/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Propanolamines/pharmacology , Animals , Antioxidants/pharmacology , Calcium Channel Blockers/chemistry , Carbazoles/chemistry , Carvedilol , Cell Membrane/chemistry , Cells, Cultured , Cerebellum/chemistry , Dihydropyridines/antagonists & inhibitors , Free Radical Scavengers/pharmacology , Lipid Bilayers/chemistry , Models, Molecular , Neurons/chemistry , Propanolamines/chemistry , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/drug effects , Sodium Channel Blockers , Veratridine/toxicity , X-Ray Diffraction
13.
Mol Pharmacol ; 51(6): 951-62, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9187261

ABSTRACT

2-Methoxyestradiol (2-ME) is an endogenous metabolite of estradiol-17beta and the oral contraceptive agent 17-ethylestradiol. 2-ME was recently reported to inhibit endothelial cell proliferation. The current study was undertaken to explore the mechanism of 2-ME effects on endothelial cells, especially whether 2-ME induces apoptosis, a prime mechanism in tissue remodeling and angiogenesis. Cultured bovine pulmonary artery endothelial cells (BPAEC) exposed to 2-ME showed morphological (including ultrastructural) features characteristic of apoptosis: cell shrinkage, cytoplasmic and nuclear condensation, and cell blebbing. 2-ME-induced apoptosis in BPAEC was a time- and concentration-dependent process (EC50 = 0.45 +/- 0.09 microM, n = 8). Nucleosomal DNA fragmentation in BPAEC treated with 2-ME was identified by agarose gel electrophoresis (DNA ladder) as well as in situ nick end labeling. Under the same experimental conditions, estradiol-17beta and two of its other metabolites, estriol and 2-methoxyestriol (< or =10 microM), did not have an apoptotic effect on BPAEC. 2-ME activated stress-activated protein kinase (SAPK)/c-Jun amino-terminal protein kinase in BPAEC in a concentration-dependent manner. The activity of SAPK was increased by 170 +/- 27% and 314 +/- 22% over the basal level in the presence of 0.4 and 2 microM 2-ME (n = 3-6), respectively. The activation of SAPK was detected at 10 min, peaked at 20 min, and returned to basal levels at 60 min after exposure to 2-ME. Inhibition of SAPK/c-Jun amino-terminal protein kinase activation by basic fibroblast growth factor, insulin-like growth factor, or forskolin reduced 2-ME-induced apoptosis. Immunohistochemical analysis of BPAEC indicated that 2-ME up-regulated expression of both Fas and Bcl-2. In addition, 2-ME inhibited BPAEC migration (IC50 = 0.71 +/- 0.11 microM, n = 4) and basic fibroblast growth factor-induced angiogenesis in the chick chorioallantoic membrane model. Taken together, these results suggest that promotion of endothelial cell apoptosis, thereby inhibiting endothelial cell proliferation and migration, may be a major mechanism by which 2-ME inhibits angiogenesis.


Subject(s)
Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Estradiol/analogs & derivatives , Mitogen-Activated Protein Kinases , Neovascularization, Physiologic/drug effects , Signal Transduction/physiology , fas Receptor/biosynthesis , 2-Methoxyestradiol , Allantois/blood supply , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cattle , Cell Movement/drug effects , Cells, Cultured , Chick Embryo , Chorion/blood supply , Colforsin/pharmacology , DNA/drug effects , DNA/metabolism , Drug Interactions , Endothelium, Vascular/cytology , Enzyme Activation/drug effects , Estradiol/pharmacology , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/pharmacology , JNK Mitogen-Activated Protein Kinases , Neovascularization, Physiologic/physiology , Nucleosomes/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Somatomedins/pharmacology , Up-Regulation/drug effects , Vitronectin/pharmacology
14.
Stroke ; 28(6): 1233-44, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9183357

ABSTRACT

BACKGROUND AND PURPOSE: Tumor necrosis factor-alpha (TNF-alpha) is a pleiotropic cytokine that rapidly upregulates in the brain after injury. The present study was designed to explore the pathophysiological significance of brain TNF-alpha in the ischemic brain by systematically evaluating the effects of lateral cerebroventricular administration of exogenous TNF-alpha and agents that block the effects of TNF-alpha on focal stroke and by examining the potential direct toxic effects of TNF-alpha on cultured neurons to better understand how TNF-alpha might mediate stroke injury. METHODS: TNF-alpha (2.5 or 25 pmol) was administered intracerebroventricularly to spontaneously hypertensive rats 24 hours before permanent or transient (80 minutes and 160 minutes) middle cerebral artery occlusion (MCAO). Animals were examined 24 hours later for neurological deficits and ischemic hemisphere necrosis and swelling. In some of these studies, neutralizing anti-TNF-alpha monoclonal antibody (mAb) (60 pmol) was injected intracerebroventricularly 30 minutes before exogenous TNF-alpha (25 pmol). In addition, the effects of blocking endogenous TNF-alpha on permanent focal ischemic injury were determined with the use of either mAb (60 pmol) or soluble TNF receptor I (sTNF-RI) (0.3 or 0.7 nmol) administered intracerebroventricularly 30 minutes before and 3 and 6 hours after MCAO. Finally, the direct neurotoxic effects of TNF-alpha were studied in cultured rat cerebellar granule cells exposed to TNF-alpha (10 to 2000 U/mL for 6 to 24 hours), and neurotransmitter release, glutamate toxicity, and oxygen radical toxicity were studied. RESULTS: TNF-alpha increased the percent hemispheric infarct induced by permanent MCAO in a dose-related manner from 13.1 +/- 1.3% (vehicle) to 18.9 +/- 1.7% at 2.5 pmol (P < .05) and 27.1 +/- 1.3% at 25 pmol (P < .0001). The high dose of TNF-alpha increased ischemia-induced forelimb deficits from 1.6 +/- 0.2 to 2.3 +/- 0.2 (P < 0.1). TNF-alpha (2.5 pmol) also increased the infarction induced by 80 or 160 minutes of transient MCAO from 1.9 +/- 0.9% to 4.3 +/- 0.4% (P < .01) and from 14.2 +/- 1.3% to 21.6 +/- 2.2% (P < .05), respectively. The exacerbation of infarct size, swelling, and neurological deficit after exogenous TNF-alpha was reversed by preinjection of 60 pmol mAb. Blocking endogenous TNF-alpha also significantly reduced focal ischemic brain injury. Treatment with 60 pmol mAb before and after permanent MCAO significantly reduced infarct size compared with control (nonimmune) antibody treatment by 20.2% (P < .05). Reduced brain infarction also was produced by brain administration of 0.3 nmol (decreased 18.2%) or 0.7 nmol (decreased 26.1%, P < .05) sTNF-RI before and after focal stroke. The intracerebroventricular administration of TNF-alpha or sTNF-RI did not alter brain or body temperature, blood gases or pH, blood pressure, blood glucose, or general blood chemistry. In cultured cerebellar granule cells, the application of TNF-alpha did not directly affect neurotransmitter release or glutamate or oxygen free radical toxicity. CONCLUSIONS: These studies demonstrate that exogenous TNF-alpha exacerbates focal ischemic injury and that blocking endogenous TNF-alpha is neuroprotective. The specificity of the action(s) of TNF-alpha was demonstrated by antagonism of its effects with specific anti-TNF-alpha tools (ie, mAb and sTNF-RI). TNF-alpha toxicity does not appear to be due to a direct effect on neurons or modulation of neuronal sensitivity to glutamate or oxygen radicals and apparently is mediated through nonneuronal cells. These data suggest that inhibiting TNF-alpha may represent a novel pharmacological strategy to treat ischemic stroke.


Subject(s)
Brain Ischemia/physiopathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antibodies, Monoclonal , Cells, Cultured , Cerebellum/drug effects , Cerebral Infarction/physiopathology , Dose-Response Relationship, Drug , Male , Neurons/drug effects , Rats , Rats, Inbred SHR , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/physiology
15.
Exp Cell Res ; 237(1): 46-54, 1997 Nov 25.
Article in English | MEDLINE | ID: mdl-9417865

ABSTRACT

Human cartilage glycoprotein 39 (HC gp-39) has been described as a major secreted product of cultured articular chondrocytes, synovial fibroblasts, and the osteosarcoma line MG63. However, its expression in these cells types has not been directly linked to corresponding cell types in vivo. In this report, expression of HC gp-39 is demonstrated from peripheral blood-derived macrophages in association with their differentiation from monocytes to macrophages. Consistent with macrophage specificity, HC gp-39 expression is also induced upon selective stimulation of the pluripotent promyelocytic leukemia cell line HL-60 toward the monocyte/macrophage lineage with vitamin D3 or phorbol 12-myristate 13-acetate (PMA), while treatments stimulating granulocyte and eosinophilic pathways do not induce expression. Furthermore, HC gp-39 expression levels correlate with the degree of morphological differentiation induced by PMA and vitamin D3 treatments. PMA-induced mRNA expression occurs by 36 h and is a secondary transcriptional response since its synthesis is inhibited by cycloheximide. Apparently, HC gp-39 expression is tied to later events in the differentiation of monocytes into macrophages. The in vivo significance of these results is validated by the in situ detection of HC gp-39 mRNA in inflammatory macrophages associated with rheumatoid synovium. Thus, macrophages appear to be an important source of HC gp-39, which has been shown to be present at elevated levels in the blood and synovium of rheumatoid arthritis patients. The implications of this extend well beyond the previously restricted observations in cell types associated with the joint and suggest a potential involvement of macrophage-derived HC gp-39 in other aspects of inflammation, tissue remodeling, and host defense.


Subject(s)
Arthritis, Rheumatoid/metabolism , Cartilage, Articular/metabolism , Glycoproteins/biosynthesis , Macrophages/metabolism , Monocytes/metabolism , Adipokines , Arthritis, Rheumatoid/pathology , Cartilage, Articular/pathology , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Chitinase-3-Like Protein 1 , Cholecalciferol/pharmacology , Cycloheximide/pharmacology , HL-60 Cells/metabolism , Humans , Inflammation , Jurkat Cells/metabolism , Kinetics , Lectins , Macrophages/cytology , Macrophages/pathology , Monocytes/cytology , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Recombinant Proteins/biosynthesis , Reference Values , Synovial Membrane/metabolism , Synovial Membrane/pathology , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic/drug effects
16.
J Neurosci ; 16(8): 2546-52, 1996 Apr 15.
Article in English | MEDLINE | ID: mdl-8786430

ABSTRACT

Recombinant human immunodeficiency virus (HIV-1) Tat protein added to the culture medium of rat cerebellar neurons promoted aggregation and formation of spoke-like neurites in a dose-dependent manner. Tat proteins containing mutations in the Arg-Gly-Asp (RGD) cell adhesion motif or a deletion of the cysteine-rich domain had no effect on neuronal morphology. In contrast, a Tat protein that contained a deletion of the proline-rich domain promoted neuronal aggregation. Aggregation of neurons was inhibited by the addition of monoclonal antibodies directed against the RGD and basic domains of Tat, but not against the proline-rich domain. The same domains of Tat required to induce aggregation also mediated adhesion of neurons to Tat-coated substrates. The HIV-2 Tat protein, which lacks an RGD sequence but contains cysteine-rich and basic domains similar to HIV-1 Tat, induced aggregation and acted as a substrate for adhesion when added at higher concentrations than HIV-1 Tat. Vitronectin, fibronectin, and RGD-containing peptides did not induce morphological changes in neurons or act as substrates for adhesion. The ability of Tat to induce morphological changes and promote adhesion was independent of the ability of Tat to transactivate HIV gene expression. Our results suggest that extracellular Tat protein most likely alters neuronal morphology and mediates adhesion by acting in a manner similar to an extracellular matrix protein.


Subject(s)
Cell Adhesion/drug effects , Cerebellum/metabolism , Gene Products, tat/metabolism , HIV-1/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Molecular Sequence Data , Rats , tat Gene Products, Human Immunodeficiency Virus
17.
Stroke ; 26(9): 1683-9; discussion 1689-90, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7660415

ABSTRACT

BACKGROUND AND PURPOSE: Excessive calcium entry into depolarized neurons contributes significantly to cerebral tissue damage after ischemia. We evaluated the ability of a novel neuronal calcium channel blocker, SB 201823-A, to block central neuronal calcium influx in vitro and to reduce ischemic injury in two rodent models of focal stroke. METHODS: Patch-clamp electrophysiology and intracellular Ca2+ imaging in rat hippocampal and cerebellar neurons were used to determine effects on neuronal calcium channel activity. Middle cerebral artery occlusion was performed in Fisher 344 rats and CD-1 mice to determine the effects on rodent focal ischemic injury and neurological deficits. Cardiovascular monitoring in conscious rats was conducted to determine cardiovascular liabilities of the compound. RESULTS: In cultured rat hippocampal cells, calcium current measured at plateau was reduced by 36 +/- 8% and 89 +/- 4% after 5 and 20 mumol/L SB 201823-A, respectively. In cerebellar granule cells in culture, pretreatment with 2.5 mumol/L SB 201823-A totally prevented initial calcium influx and reduced later calcium influx by 50 +/- 2.5% after N-methyl-D-aspartate/glycine stimulation (P < .01). KCl depolarization-induced calcium influx also was reduced by more than 95%. In rats, a single treatment with 10 mg/kg IV SB 201823-A beginning 30 minutes after focal ischemia decreased (P < .05) hemispheric infarct by 30.4% and infarct volume by 29.3% and reduced (P < .05) forelimb deficits by 47.8% and hindlimb deficits by 36.3%. In mice, treatments with 10 mg/kg IP SB 201823-A beginning 30 minutes after focal ischemia significantly reduced infarct volume by 41.5% (P < .01). No blood pressure effects were observed with the therapeutic dose of the compound. CONCLUSIONS: These results indicate that the new neuronal calcium channel blocker SB 201823-A can block stimulated calcium influx into central neurons and can provide neuroprotection in two models of focal cerebral ischemia without affecting blood pressure. Data from several different studies now indicate that the neuronal calcium channel antagonists are a promising therapy for the postischemic treatment of stroke.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium/antagonists & inhibitors , Ischemic Attack, Transient/metabolism , Neurons/drug effects , Piperidines/pharmacology , Animals , Blood Pressure/drug effects , Calcium Channel Blockers/therapeutic use , Calcium Channels/drug effects , Cells, Cultured , Cerebellum/drug effects , Cerebellum/metabolism , Cerebral Infarction/prevention & control , Disease Models, Animal , Glycine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Ischemic Attack, Transient/drug therapy , Male , Mice , Mice, Inbred Strains , N-Methylaspartate/pharmacology , Neuroprotective Agents/pharmacology , Patch-Clamp Techniques , Piperidines/therapeutic use , Potassium Chloride/pharmacology , Rats , Rats, Inbred F344 , Rats, Inbred Strains , Rats, Sprague-Dawley
18.
Brain Res Dev Brain Res ; 88(1): 96-101, 1995 Aug 28.
Article in English | MEDLINE | ID: mdl-7493411

ABSTRACT

We describe the identification and expression of endothelin (ET) receptor subtypes in differentiating cultured cerebellar neurons. Using [125I]ET-1 and the subtype-selective ligands BQ-123 and sarafotoxin 6c as selective ligands for the ETA and ETB receptors, respectively, we found that cerebellum from 8-day-old rats displayed only the ETB receptor subtype. We next cultivated cerebellar granule cell neurons to study ET receptor differentiation between 2 and 22 days in vitro. Using the above reagents, we found that while unlabeled ET-1 displayed monophasic competition curves, BQ-123 and sarafotoxin 6c displayed partial displacement curves, indicating the presence of both ETA and ETB receptors on these neurons. The proportion of ETB receptors gradually decreased from day 2 onwards the proportion of ETA receptors gradually increased. On days 2, 3, 4, and 5 of culture, the ETB:ETA receptor ratios were 90:10, 70:30, 60:40, and 40:60, respectively. There was no further change in receptor subtype ratio beyond day 5 and up to day 22. Northern blot analysis showed that ETB receptor message expression was 6.9-fold higher than that of ETA receptor expression on day 2, but steadily decreased with time, whereas ETA receptor message expression was minimal on day 2 and maximal by day 3 and 4. By day 7, receptor message was of equal abundance, which was in good agreement with the binding studies. This novel, developmentally regulated process predicts the existence of endogenous mediators of neuronal ET receptor expression.


Subject(s)
Cerebellum/cytology , Cerebellum/metabolism , Neurons/metabolism , Receptors, Endothelin/metabolism , Animals , Blotting, Northern , Cell Differentiation , Cells, Cultured , Cerebellum/growth & development , Endothelin Receptor Antagonists , Endothelins/metabolism , Iodine Radioisotopes , Kinetics , Neurons/drug effects , Peptides, Cyclic/pharmacology , RNA, Messenger/biosynthesis , Radioligand Assay , Rats , Receptors, Endothelin/drug effects , Vasoconstrictor Agents/pharmacology , Viper Venoms/pharmacology
19.
Atherosclerosis ; 115(1): 89-98, 1995 May.
Article in English | MEDLINE | ID: mdl-7545398

ABSTRACT

Increased expression of cell adhesion molecules is an important pathological event during the development of atherosclerosis. The smooth muscle cell (SMC) is one of the cell types present in the atherosclerotic lesion. To evaluate the regulation of adhesion molecules in human vascular SMCs and its possible role, we studied the expression of adhesion molecules in SMCs stimulated with interleukin 1-beta (IL-1 beta), a pleiotropic cytokine that is involved in the pathological development of vascular diseases including atherosclerosis and restenosis. Our data demonstrated that IL-1 beta markedly induced the adhesiveness of human vascular SMCs for monocytes and neutrophils in a concentration (10 pM - 10 nM)- and time (0.5-24 h)-dependent manner. The maximal induced adhesion by IL-1 beta (1 nM) was reached at 4 h, with 4.6-fold and 3.3-fold for monocytes and neutrophils, respectively. This induction was dose-dependently inhibited by the IL-1 receptor antagonist (IL-1 ra). The IL-1 beta-induced expression of intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1) and E-selectin 1 (ELAM-1) on SMCs was examined by reverse transcription/polymerase chain reaction (RT/PCR). Unstimulated, serum-deprived SMCs expressed a low or undetectable level of mRNA for these adhesion molecules. The expression of ICAM-1 and VCAM-1 but not ELAM-1 mRNA was significantly induced with IL-1 beta in a concentration (1 fM - 1 nM)- and time (0.5 - 24 h)-dependent manner. The maximal increase in ICAM-1 and VCAM-1 mRNAs was reached at 4 h after IL-1 beta stimulation. The IL-1 beta-induced adhesion of SMCs for monocytes was partially inhibited by monoclonal anti-human ICAM-1 and anti-human VCAM-1 antibody, but not by anti-human ELAM-1 antibody. Pretreatment of monocytes with anti-human integrin beta 2 antibody significantly reduced the adhesion of monocytes to IL-1 beta-stimulated SMCs. These results suggest that IL-1 beta is a potent inducer for ICAM-1 and VCAM-1 expression in human vascular SMC, and could play a role in the pathogenesis of atherosclerosis by recruitment and retention of inflammatory cells such as monocytes and neutrophils in the lesions.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Cell Adhesion/drug effects , Gene Expression/drug effects , Interleukin-1/pharmacology , Leukocytes/physiology , Muscle, Smooth, Vascular/physiology , Base Sequence , Cells, Cultured , DNA Primers , Dose-Response Relationship, Drug , E-Selectin , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Interleukin 1 Receptor Antagonist Protein , Leukocytes/drug effects , Molecular Sequence Data , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Polymerase Chain Reaction , Receptors, Interleukin-1/antagonists & inhibitors , Recombinant Proteins/pharmacology , Sialoglycoproteins/pharmacology , Vascular Cell Adhesion Molecule-1
20.
Peptides ; 16(7): 1279-82, 1995.
Article in English | MEDLINE | ID: mdl-8545251

ABSTRACT

We studied the binding characteristics of a novel, nonpeptide endothelin antagonist, SB 209670, to two subtypes of endothelin (ET) receptor in cultured rat cerebellar granule cell neurons. Displacement binding studies of [125I]ET-1 performed in the presence of the ETB receptor-selective agonist, sarafotoxin 6c (S6c), allowed us to measure a Ki of 4.0 +/- 1.5 nM for (+/-)SB 209670 at the ETA receptor (n = 4). Similarly, binding studies in the presence of the ETA receptor-selective antagonist, BQ123, allowed us to measure a Ki of 46 +/- 14 nM for (+/-)SB 209670 at the ETB receptor (n = 4). These studies indicate that the novel endothelin antagonist, SB 209670, has high affinity for both types of neuronal endothelin receptor.


Subject(s)
Cerebellum/metabolism , Endothelin Receptor Antagonists , Indans/metabolism , Neurons/metabolism , Receptors, Endothelin/metabolism , Animals , Binding, Competitive , Cells, Cultured , Cerebellum/cytology , Dose-Response Relationship, Drug , Endothelins/metabolism , Neurons/cytology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...