Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 447
Filter
1.
Br J Cancer ; 105(2): 246-54, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21712828

ABSTRACT

BACKGROUND: The epidermal growth factor receptor-targeted monoclonal antibody cetuximab (Erbitux) was recently introduced for the treatment of metastatic colorectal cancer. Treatment response is dependent on Kirsten-Ras (K-Ras) mutation status, in which the majority of patients with tumour-specific K-Ras mutations fail to respond to treatment. Mutations in the oncogenes B-Raf and PIK3CA (phosphoinositide-3-kinase) may also influence cetuximab response, highlighting the need for a sensitive, accurate and quantitative assessment of tumour mutation burden. METHODS: Mutations in K-Ras, B-Raf and PIK3CA were identified by both dideoxy and quantitative pyrosequencing-based methods in a cohort of unselected colorectal tumours (n=102), and pyrosequencing-based mutation calls correlated with various clinico-pathological parameters. RESULTS: The use of quantitative pyrosequencing-based methods allowed us to report a 13.7% increase in mutation burden, and to identify low-frequency (<30% mutation burden) mutations not routinely detected by dideoxy sequencing. K-Ras and B-Raf mutations were mutually exclusive and independently associated with a more advanced tumour phenotype. CONCLUSION: Pyrosequencing-based methods facilitate the identification of low-frequency tumour mutations and allow more accurate assessment of tumour mutation burden. Quantitative assessment of mutation burden may permit a more detailed evaluation of the role of specific tumour mutations in the pathogenesis and progression of colorectal cancer and may improve future patient selection for targeted drug therapies.


Subject(s)
Carcinoma/genetics , Colorectal Neoplasms/genetics , DNA Mutational Analysis/methods , Individuality , Mutation , Oncogenes/genetics , Adult , Aged , Aged, 80 and over , Carcinoma/pathology , Carcinoma/surgery , Cohort Studies , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Mutation/physiology , Sequence Analysis, DNA/methods
2.
Science ; 228(4699): 606-8, 1985 May 03.
Article in English | MEDLINE | ID: mdl-2858918

ABSTRACT

Specific radioimmunoassays were used to measure the effects of hypertonic saline (salt loading), water deprivation, and trichothecene mycotoxin (T2 toxin) on the content of methionine enkephalin (ME), leucine enkephalin (LE), alpha-neoendorphin, dynorphin A, dynorphin B, vasopressin, and oxytocin in the rat posterior pituitary. Concentrations of vasopressin and oxytocin decreased in response to both osmotic stimuli and treatment with T2 toxin, but the decrease was greater with osmotic stimulations. Similarly, concentrations of LE and dynorphin-related peptides declined after salt loading and water deprivation; LE concentrations also decreased after treatment with T2 toxin. The concentration of ME decreased after water deprivation, did not change after salt loading, and increased after T2 toxin treatment. The differentiating effects of these stimuli on the content of immunoreactive LE and ME are consistent with the hypothesis that LE and ME may be localized in separate populations of nerve endings with different roles in the posterior pituitary.


Subject(s)
Enkephalin, Leucine/analysis , Enkephalin, Methionine/analysis , Pituitary Gland, Posterior/analysis , Sesquiterpenes/pharmacology , T-2 Toxin/pharmacology , Animals , Dynorphins/analogs & derivatives , Dynorphins/analysis , Endorphins/analysis , Male , Osmosis , Oxytocin/analysis , Pituitary Gland, Posterior/drug effects , Protein Precursors/analysis , Rats , Rats, Inbred Strains , Saline Solution, Hypertonic , Vasopressins/analysis , Water Deprivation
3.
J Clin Invest ; 76(6): 2134-8, 1985 Dec.
Article in English | MEDLINE | ID: mdl-4077976

ABSTRACT

A murine monoclonal antibody (15H6) against the trichothecene mycotoxin T-2 was capable of neutralizing the in vitro protein synthesis inhibitory effect of T-2 toxin in human B lymphoblastoid cultures. It was further shown that 15H6 given to rats (250 mg/kg) 30 min before or 15 min after a lethal dose (1 mg/kg) of T-2 toxin conferred 100% survival. A lower dose of 15H6 (125 mg/kg), given 15 min after the lethal dose of T-2 toxin, protected 25% of the rats. An increased time to death and 45% survival was seen in rats given the full dose of 15H6 antibody 60 min after lethal toxin. These data are the first demonstration of effective prophylaxis and therapy for T-2 toxemia.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antitoxins/therapeutic use , Sesquiterpenes/antagonists & inhibitors , Sesquiterpenes/immunology , T-2 Toxin/immunology , Trichothecenes/antagonists & inhibitors , Animals , Blood Pressure/drug effects , Cells, Cultured , Heart Rate/drug effects , Neutralization Tests , Protein Biosynthesis , Rats , T-2 Toxin/toxicity , Trichothecenes/immunology
4.
J Clin Invest ; 89(5): 1669-73, 1992 May.
Article in English | MEDLINE | ID: mdl-1314853

ABSTRACT

Interleukin-2 was recently shown to cause acute lung injury characterized by microvascular permeability defect, interstitial edema, and leukosequestration. Similar responses can also be produced by platelet activating factor (PAF). Thus, the present study aimed to examine whether PAF plays a key role in the development of IL-2-induced lung injury in the anesthetized rat. Intravenous infusion (60 min) of recombinant human IL-2 at 10(5)-10(6) U/rat (n = 7-9) dose-dependently elevated lung water content (27 +/- 1%, P less than 0.01), myeloperoxidase activity (+84 +/- 23%, P less than 0.05), and serum thromboxane B2 (990 +/- 70%, P less than 0.01), but failed to alter blood pressure, hematocrit, serum tumor necrosis factor-alpha, and circulating leukocytes and platelets. Pretreatment (-30 min) with a potent and specific PAF antagonist, BN 50739 (10 mg/kg, intraperitoneally, n = 6) prevented the pulmonary edema (P less than 0.05) and thromboxane B2 production (P less than 0.01), and attenuated the elevation of lung myeloperoxidase activity (+18 +/- 16%, P less than 0.05) induced by IL-2. These data suggest that PAF is involved in the pathophysiological processes leading to IL-2-induced lung injury, and point to the potential therapeutic capacity of PAF antagonists in preventing pulmonary edema during IL-2 therapy.


Subject(s)
Azepines/pharmacology , Interleukin-2/toxicity , Lung Diseases/chemically induced , Platelet Activating Factor/physiology , Triazoles/pharmacology , Animals , Blood Pressure/drug effects , Leukocyte Count , Male , Peroxidase/metabolism , Platelet Count/drug effects , Rats , Rats, Inbred Strains , Thromboxane B2/blood , Tumor Necrosis Factor-alpha/metabolism
5.
J Clin Invest ; 67(4): 1118-25, 1981 Apr.
Article in English | MEDLINE | ID: mdl-7009654

ABSTRACT

Severe endotoxemia, a condition where microembolization and intravascular coagulation are thought to play important roles, was treated experimentally with prostacyclin (PGI(2)). In a study of 24 dogs, 8 control animals injected with 1.75 mg.kg(-1) of endotoxin died within 24 h. Six animals given intravenous aspirin 100 mg/kg, 30 min after endotoxin died. 9 of 10 dogs infused with 100 ng PGI(2).kg(-1).min(-1) for 3 h, given 30 min after the injection of endotoxin survived 24 h (P < 0.025). Injection of endotoxin resulted in a: (a) maximal 62% fall in mean arterial pressure (P < 0.001); (b) transient doubling of mean pulmonary arterial pressure (P < 0.001); (c) initial 70% drop in cardiac index (P < 0.001); (d) decline in blood platelets from 213,700 to 13,700/mm(3) (P < 0.001), and leukocytes from 7,719 to < 750/mm(3) (P < 0.001); (e) depressed urine output (P < 0.001); (f) 34% decrease in blood fibrinogen (P < 0.01) and an increase in fibrin degradation products > 50 mug/ml (P < 0.001); (g) fivefold increase in circulating cathepsin D titer (P < 0.005) and (h) increase in blood norepinephrine (P < 0.005), dopamine (P < 0.005), and epinephrine (P < 0.001). Aspirin treatment led to an increase in mean arterial pressure (P < 0.001) and mean pulmonary arterial pressure (P < 0.005), but cardiac index, urine flow, platelets, leukocytes, fibrin degradation products, and cathepsin D levels remained similar to untreated controls. After infusion of PGI(2) there was a: (a) prompt increase of cardiac index to base-line levels; (b) late increase in mean arterial pressure (P < 0.005) after the discontinuation of PGI(2) treatment (c) restoration of urine output; (d) increase in circulating platelets to levels still below base line but above untreated control animals (P < 0.05); (e) no effect on circulating leukocyte levels; (f) fall in fibrin degradation products to 11.2 mug/ml (P < 0.05); (g) decline in cathepsin D levels to values 60% lower than the untreated controls (P < 0.025); and (h) reduction in plasma norepinephrine levels to base line at 4 h (P < 0.005). Although the mode of PGI(2) action is not clear, it is effective in the treatment of experimental endotoxemia.


Subject(s)
Endotoxins/blood , Epoprostenol/therapeutic use , Prostaglandins/therapeutic use , Animals , Catecholamines/blood , Cathepsin D , Cathepsins/blood , Dogs , Endotoxins/antagonists & inhibitors , Fibrin Fibrinogen Degradation Products/metabolism , Fibrinogen/metabolism , Hemodynamics/drug effects , Leukocyte Count , Lung/drug effects , Platelet Count
6.
J Neurosci ; 19(14): 5932-41, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10407032

ABSTRACT

A number of studies have provided evidence that neuronal cell loss after stroke involves programmed cell death or apoptosis. In particular, recent biochemical and immunohistochemical studies have demonstrated the expression and activation of intracellular proteases, notably caspase-3, which act as both initiators and executors of the apoptotic process. To further elucidate the involvement of caspases in neuronal cell death induced by focal stroke we developed a panel of antibodies and investigated the spatial and temporal pattern of both caspase-8 and caspase-3 expression. Our efforts focused on caspase-8 because its "apical" position within the enzymatic cascade of caspases makes it a potentially important therapeutic target. Constitutive expression of procaspase-8 was detectable in most cortical neurons, and proteolytic processing yielding the active form of caspase-8 was found as early as 6 hr after focal stroke induced in rats by permanent middle cerebral artery occlusion. This active form of caspase-8 was predominantly seen in the large pyramidal neurons of lamina V. Active caspase-3 was evident only in neurons located within lamina II/III starting at 24 hr after injury and in microglia throughout the core infarct at all times examined. Terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling, gel electrophoresis of DNA, and neuronal cell quantitation indicated that there was an early nonapoptotic loss of cortical neurons followed by a progressive elimination of neurons with features of apoptosis. These data indicate that the pattern of caspase expression occurring during delayed neuronal cell death after focal stroke will vary depending on the neuronal phenotype.


Subject(s)
Brain Ischemia/enzymology , Caspases/genetics , Cerebral Cortex/enzymology , Neurons/enzymology , Animals , Apoptosis , Brain Ischemia/pathology , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/biosynthesis , Cell Death , Cerebral Cortex/pathology , Functional Laterality , Gene Expression Regulation, Enzymologic , Immunohistochemistry , Ischemic Attack, Transient/enzymology , Ischemic Attack, Transient/pathology , Male , Neurons/pathology , Rats , Rats, Inbred SHR , Reperfusion , Time Factors
7.
Biochim Biophys Acta ; 1284(1): 20-8, 1996 Oct 02.
Article in English | MEDLINE | ID: mdl-8865810

ABSTRACT

Carvedilol (Kredex, Coreg) is a multiple action antihypertensive drug that has been shown to protect cell membranes from lipid peroxidative damages. In this study the physical and structural effects of carvedilol on lipid bilayers are investigated by fluorescence techniques, differential scanning calorimetry and other physical methods. Carvedilol binds to liposomal membranes (9:1 DMPC:DMPG) strongly with an apparent binding constant on the order of 10(4) M-1 in PBS (pH 7.4). The characteristic changes in its intrinsic fluorescence properties when bound to liposomes suggest that this compound is situated in a non-polar environment. The Stern-Volmer and bimolecular quenching constants, determined using nitrate as the fluorescence quencher, for the free and bound carvedilol indicate that the carbazole moiety is at a depth of > 11 A in the lipid bilayer. Fluorescence anisotropy measurements show that, unlike the membrane probes DPH and TMA-DPH, carvedilol is relatively mobile, and does not have a rigidly-defined molecular orientation in the bilayers. Differential scanning calorimetry results indicate that carvedilol is an effective membrane "fluidizer' as it dose-dependently lowers the gel to liquid crystalline transition temperature and broadens the endothermic transition. Comparative studies of interactions of carbazole, 4-OH carbazole and carvedilol with the model liposomal membranes reveal a possible role of membrane-partitioning in their antioxidant efficacy. These findings are discussed in perspective with the membrane biophysical properties of different classes of therapeutic significant lipid antioxidants in mind.


Subject(s)
Antihypertensive Agents/chemistry , Carbazoles/chemistry , Lipid Bilayers/chemistry , Liposomes/chemistry , Propanolamines/chemistry , Binding Sites , Calorimetry , Carvedilol , Fluorescence
8.
Circulation ; 102(11): 1315-22, 2000 Sep 12.
Article in English | MEDLINE | ID: mdl-10982549

ABSTRACT

BACKGROUND: Chemokines are potent proinflammatory and immune modulators. Increased expression of chemokines, eg, monocyte chemoattractant protein-1 (MCP-1), has recently been described in clinical and experimental heart failure. The present report is aimed at exploring the expression, localization, and binding site regulation of MCP-1, a member of the C-C chemokine family, in a rat model of volume-overload congestive heart failure (CHF). METHODS AND RESULTS: An aortocaval fistula was surgically created between the abdominal aorta and inferior vena cava. Rats with CHF were further subdivided into compensated and decompensated subgroups. Northern blot analysis and real-time quantitative polymerase chain reaction demonstrated upregulation of MCP-1 mRNA expression correlating with the severity of CHF (288+/-22, 502+/-62, and 826+/-138 copies/ng total RNA for sham, compensated, and decompensated animals, respectively; n=5, P:<0.05). MCP-1 protein was localized by immunohistochemistry in cardiomyocytes, vascular endothelium and smooth muscle cells, infiltrating leukocytes, and interstitial fibroblasts, and its intensity increased with severity of CHF. In addition, rats with CHF displayed a significant decrease of (125)I-labeled MCP-1 binding sites to myocardium-derived membranes (384.3+/-57.0, 181.3+/-8.8, and 123.3+/-14.1 fmol/mg protein for sham, compensated, and decompensated animals, respectively). CONCLUSIONS: Volume-overload CHF in rats is associated with alterations in the expression, immunohistochemical localization, and receptor binding of the MCP-1 chemokine in the myocardium. These changes were more pronounced in rats with decompensated CHF. The data suggest that activation of the MCP-1 system may contribute to the progressive cardiac decompensation and development of CHF in rats with aortocaval fistula.


Subject(s)
Chemokine CCL2/metabolism , Heart Failure/metabolism , Animals , Binding, Competitive , Blotting, Northern , Heart Failure/diagnostic imaging , Immunohistochemistry , In Vitro Techniques , Male , Myocardium/metabolism , Polymerase Chain Reaction , RNA, Messenger/metabolism , Radioligand Assay , Rats , Rats, Wistar , Ultrasonography , Up-Regulation
9.
J Thromb Haemost ; 3(4): 695-702, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15733058

ABSTRACT

Factor XI (FXI) and factor IX (FIX) are zymogens of plasma serine proteases required for normal hemostasis. The purpose of this work was to evaluate FXI and FIX as potential therapeutic targets by means of a refined ferric chloride (FeCl(3))-induced arterial injury model in factor-deficient mice. Various concentrations of FeCl(3) were used to establish the arterial thrombosis model in C57BL/6 mice. Carotid artery blood flow was completely blocked within 10 min in C57BL/6 mice by application of 3.5% FeCl(3). In contrast, FXI- and FIX-deficient mice were fully protected from occlusion induced by 5% FeCl(3), and were partially protected against the effect of 7.5% FeCl(3). The protective effect was comparable to very high doses of heparin (1000 units kg(-1)) and substantially more effective than aspirin. While FXI and FIX deficiencies were indistinguishable in the carotid artery injury model, there was a marked difference in a tail-bleeding-time assay. FXI-deficient and wild-type mice have similar bleeding times, while FIX deficiency was associated with severely prolonged bleeding times (>5.8-fold increase, P < 0.01). Given the relatively mild bleeding diathesis associated with FXI deficiency, therapeutic inhibition of FXI may be a reasonable strategy for treating or preventing thrombus formation.


Subject(s)
Carotid Arteries/drug effects , Factor IX/physiology , Factor XI Deficiency/pathology , Factor XI/physiology , Ferric Compounds/pharmacology , Hemophilia B/pathology , Animals , Arteries/drug effects , Arteries/injuries , Aspirin/pharmacology , Bleeding Time , Blood Flow Velocity , Carotid Artery Diseases/pathology , Chlorides , Dose-Response Relationship, Drug , Genotype , Heparin/chemistry , Heparin/pharmacology , Homozygote , Mice , Mice, Inbred C57BL , Platelet Aggregation , Regional Blood Flow/drug effects , Thrombosis/pathology , Thrombosis/therapy , Time Factors
10.
Trends Cardiovasc Med ; 7(7): 249-55, 1997 Oct.
Article in English | MEDLINE | ID: mdl-21235893

ABSTRACT

Congestive heart failure (CHF) is the final clinical manifestation of a variety of cardiac (myopathies), coronary (atherosclerosis), and systemic diseases (diabetes, hypertension). Regardless of the origin of the cardiac insult, left ventricular dysfunction resulting in decreased cardiac output elicits a series of adaptational processes that attempt to compensate for some of the decrement in myocardial function. One of the key manifestations of these compensatory processes is cardiac hypertrophy, which is characterized by a marked increase in myocyte size and an increase in contractile proteins. The benefits resulting from these compensatory adaptational mechanisms, however, are only transient, and within a period of months to years, the changes induced in the myocardium fail to sustain cardiac output at a level that is sufficient to meet the demands of the body; subsequently, physical performance is impaired. Typically, progressive dilation and thinning of the left ventricle occur along with progression of CHF. The mechanisms responsible for the thinning of ventricular tissue and loss of left ventricular mass are poorly understood; traditionally, such loss has been attributed to tissue necrosis based on the morphologic observation of dead cardiac myocytes. Very recently, there have been data suggesting that apoptosis, a form of programmed cell death (PCD), occurs in the heart and may be responsible, at least in part, for the progression of CHF and the chronic loss of left ventricular function and mass. Evidence for a role of apoptosis/PCD in the progression of heart failure has been obtained from a variety of observations, including in vitro studies of cardiac myocytes in culture, experimental animal models of cardiac injury, and cardiac tissue obtained from patients with CHF. Thus, apoptosis/PCD may be a critical mechanism involved in the progressive loss of cardiac myocytes, which ultimately results in end-stage heart failure. In this brief review, the evidence for apoptosis/PCD in cardiac myocytes is presented and its potential role in the progression of CHF is analyzed. In particular, the genomic basis for apoptosis in cardiac myocytes is explored, and its relevance to the identification of novel targets for future pharmacological interventions is discussed. (Trends Cardiovasc Med 1997;7:249-255). © 1997, Elsevier Science Inc.

11.
Cardiovasc Res ; 45(3): 560-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10728377

ABSTRACT

Apoptosis is a form of cell death that involves discrete genetic and molecular programs, de novo protein expression and a unique cellular phenotype. Evidence for the existence of apoptosis in the human heart has been reported in various cardiac diseases, including ischemic and non-ischemic heart failure, myocardial infarction and arrhythmias. Among the most potent stimuli that elicit cardiomyocyte apoptosis are: oxygen radicals (including NO), cytokines (FAS/TNF alpha-receptor signaling), stress conditions (chemical or physical, e.g., radiation), sphingolipid metabolites (ceramide) and autocoids, e.g., angiotensin II. Apoptosis of cardiac myocytes may contribute to progressive pump-failure, arrhythmias and cardiac remodeling. The recognition of numerous molecular targets associated with cardiomyocyte apoptosis may provide novel therapeutic strategies for diverse cardiac ailments, as recently suggested by pharmacologic studies in experimental animals. This review paper is aimed to highlight the role of protein kinase signaling pathways in apoptosis with special attention to the stress-activated protein kinases (SAPK) and mitogen-activated protein kinases (MAPK) systems.


Subject(s)
Apoptosis , Cardiovascular Diseases/physiopathology , MAP Kinase Signaling System , Myocardium/metabolism , Oxidative Stress , Second Messenger Systems , Adrenergic alpha-Antagonists/therapeutic use , Adrenergic beta-Antagonists/therapeutic use , Animals , Apoptosis/drug effects , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Humans , Phospholipids/metabolism , Protein Kinases/metabolism
12.
Cardiovasc Res ; 35(3): 414-21, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9415285

ABSTRACT

Recent advances in molecular biology techniques have provided powerful tools for discovery of novel genes relevant to both biological and pathological processes. mRNA differential display is an emerging technique for novel gene discovery and it has been successfully applied to many physiological and pathological conditions including normal development, cell differentiation, cancer, cardiovascular disease, inflammation and CNS disorders. In the present work, we briefly illustrate the critical procedure and highlight most recent technical improvements and modifications of this technology. Based upon the successful applications of this technique in cardiovascular research, it may provide a valuable and powerful investigational tool for the identification of novel therapeutic targets in cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/therapy , Genetic Techniques , Genetic Therapy/methods , RNA, Messenger/analysis , Gene Expression , Humans , Polymerase Chain Reaction , Sequence Analysis, DNA , Transcription, Genetic
13.
Cardiovasc Res ; 28(12): 1808-14, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7867034

ABSTRACT

OBJECTIVE: The aim was to investigate whether interleukin-1 beta (IL-1 beta) plays a role in modulating the adhesion of monocytes and neutrophils to vascular smooth muscle cells, and to identify what molecules on these cells may be involved in the adhesion. METHODS: Rat aortic smooth muscle cells were challenged with IL-1 beta and tested for adhesion of prelabelled monocytes and neutrophils. Northern analysis, reverse transcription/polymerase chain reaction (RT/PCR), and immunocytochemical staining were used to measure the changes of intercellular adhesion molecule-1 (ICAM-1) and other adhesion molecules in response to IL-1 beta stimulation. Neutralising antibody against ICAM-1 was used to demonstrate a role of ICAM-1 in this IL-1 beta induced adhesion. RESULTS: IL-1 beta induced the adhesion of monocytes and neutrophils to aortic smooth muscle cells in a concentration and time dependent manner. IL-1 beta-induced adhesion was inhibited by preincubation of the cells with an IL-1 receptor antagonist (IL-1ra). Northern analysis and RT/PCR showed that ICAM-1 mRNA represents a predominant adhesion molecule induced by IL-1 beta, and that the expression of ICAM-1 mRNA precedes and parallels the induced adhesion profiles of aortic smooth muscle cells for leucocytes. Immunocytochemical staining confirmed the IL-1 beta induced ICAM-1 expression on the smooth muscle cells. Moreover, a monoclonal anti-rat ICAM-1 antibody produced a concentration dependent inhibition of the IL-1 beta induced adhesion of monocytes and neutrophils to the smooth muscle cells. CONCLUSIONS: IL-1 beta actively regulates functional ICAM-1 expression in vascular smooth muscle cells. The IL-1 beta-induced expression of ICAM-1 on the smooth muscle cells may be an important contributor to the increased adhesion by monocytes and neutrophils to these cells and suggests that IL-1 beta might play a role in the proinflammatory and immune functions of the modified smooth muscle cells during atherosclerosis and restenosis.


Subject(s)
Intercellular Adhesion Molecule-1/immunology , Interleukin-1/immunology , Leukocytes/immunology , Muscle, Smooth, Vascular/immunology , Animals , Arteriosclerosis/immunology , Base Sequence , Blotting, Northern , Cell Adhesion/immunology , Cells, Cultured , DNA Primers , Humans , Immunohistochemistry , Molecular Sequence Data , Monocytes/immunology , Muscle, Smooth, Vascular/cytology , Neutrophils/immunology , Polymerase Chain Reaction , Rats
14.
Cardiovasc Res ; 28(3): 400-6, 1994 Mar.
Article in English | MEDLINE | ID: mdl-7909721

ABSTRACT

OBJECTIVE: Oxygen radical mediated endothelial injury plays an important role in cardiovascular disease. Carvedilol, a new beta blocker and antihypertensive agent, has been shown to have antioxidant activity. The aim of this study was to determine whether carvedilol protects oxygen radical induced endothelial injury. METHODS: Cultured bovine pulmonary artery (BPAEC) and human umbilical vein endothelial cells (HUVEC) were used and oxygen radicals were generated by xanthine-xanthine oxidase or phorbol myristate acetate (PMA) activated human neutrophils. Cell injury was assessed by lactate dehydrogenase (LDH) release and cell death, or 51 Cr release from prelabelled BPAEC. The electron paramagnetic resonance (EPR) spin trapping technique was used to detect the amount of radical spin adducts formed in cell lipids. RESULTS: Carvedilol dose dependently inhibited xanthine-xanthine oxidase induced LDH release from BPAEC and HUVEC, with IC50 values of 3.8 microM and 2.6 microM, respectively, and significantly reduced cell death by xanthine-xanthine oxidase. Other beta blockers tested (propranolol, labetalol, pindolol, and celiprolol) showed a mild effect or no effect at all. Increasing the time of pretreatment with carvedilol enhanced its cell protective effect against oxidative stress. Carvedilol also protected BPAEC dose dependently from PMA activated, neutrophil induced cell injury. Carvedilol had no effect on xanthine oxidase activity. EPR study confirmed that xanthine-xanthine oxidase induced the formation of lipid derived radicals in cell lipids and carvedilol scavenged free radicals, as indicated by the decreased EPR signal. CONCLUSIONS: Carvedilol protects endothelial cells against oxygen radical mediated cell injury and death by scavenging free radicals. The prevention of oxidative injury to endothelial cells might potentially contribute to the clinical beneficial effects of carvedilol as an antihypertensive agent.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Antihypertensive Agents/pharmacology , Carbazoles/pharmacology , Endothelium, Vascular/drug effects , Propanolamines/pharmacology , Vasodilator Agents/pharmacology , Animals , Carbazoles/chemistry , Carvedilol , Cattle , Cells, Cultured , Electron Spin Resonance Spectroscopy , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , L-Lactate Dehydrogenase/metabolism , Neutrophils/physiology , Propanolamines/chemistry , Pulmonary Artery/cytology , Reactive Oxygen Species/metabolism , Tetradecanoylphorbol Acetate/metabolism , Xanthine , Xanthine Oxidase/metabolism , Xanthines/metabolism
15.
Cardiovasc Res ; 26(5): 518-25, 1992 May.
Article in English | MEDLINE | ID: mdl-1359930

ABSTRACT

OBJECTIVE: The aim was to evaluate in a minipig model of acute myocardial infarction the cardioprotection provided by the beta adrenoceptor blocking and vasodilating activities present in carvedilol; comparison was made to the pure beta adrenoceptor antagonist, propranolol. METHODS: Experiments were performed in 25 Yucatan minipigs (9-12 kg), randomly assigned to receive vehicle (n = 7), carvedilol 0.3 mg.kg-1 (n = 6), carvedilol 1 mg.kg-1 (n = 6), or propranolol 1 mg.kg-1 (n = 6). Myocardial infarction was produced by occlusion of the left anterior descending coronary artery for 45 min followed by 4 h of reperfusion. Vehicle, carvedilol (0.3 and 1 mg.kg-1) or propranolol (1 mg.kg-1) were given intravenously 15 min before the coronary artery occlusion. At the end of the reperfusion period, infarct size was determined using Evans blue dye and triphenyltetrazolium chloride staining. Infarct volumes were visualised using computer assisted three dimensional image analysis of the stained myocardial tissue sections. Myeloperoxidase activity was measured in tissue samples removed from normal, infarcted, and at risk areas. RESULTS: Carvedilol (1 mg.kg-1) reduced infarct size by over 90% without producing pronounced changes in systemic haemodynamic variables. The ability of carvedilol to reduce infarct size was clearly dose dependent. Thus infarct size, which represented 27.5(SEM 2.3)% of the area at risk in the vehicle treated group, was only 13.1(4.0)% (p < 0.05) and 2.4(1.5)% (p < 0.01) in pigs treated with carvedilol at 0.3 and 1 mg.kg-1, respectively. In animals treated with propranolol (1 mg.kg-1), infarct size represented 10.9(2.4)% of the area at risk (p < 0.05). The 60% and 91% reductions in infarct size produced by propranolol (1 mg.kg-1) and carvedilol (1 mg.kg-1), respectively, were clearly evident upon three dimensional image analysis. The reduction in infarct size was significantly greater for carvedilol (1 mg.kg-1) compared to propranolol (1 mg.kg-1) at equivalent beta adrenoceptor blocking doses. Pretreatment with propranolol did not reduce the increases in myeloperoxidase activity observed in the area at risk or in the infarcted area. In contrast, carvedilol produced a dose dependent reduction in myeloperoxidase activity in these areas. CONCLUSIONS: Carvedilol limits myocardial necrosis resulting from coronary artery occlusion and reperfusion in a more pronounced manner than the pure beta adrenoceptor antagonist, propranolol. The cardioprotective effect of carvedilol, which reduced infarct size by 91%, may result from the combined effects of beta adrenoceptor blockade and vasodilatation, and possibly also from inhibition of intracellular calcium overload in cardiac cells resulting from antagonism of myocardial alpha 1 adrenoceptors and/or calcium channel blockade. The cardioprotection provided by carvedilol may ultimately be of benefit in hypertensive patients who are at risk for acute myocardial infarction.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Antihypertensive Agents/therapeutic use , Carbazoles/therapeutic use , Myocardial Infarction/prevention & control , Propanolamines/therapeutic use , Vasodilator Agents/therapeutic use , Animals , Blood Pressure/drug effects , Carvedilol , Disease Models, Animal , Heart Rate/drug effects , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Peroxidase/metabolism , Propranolol/therapeutic use , Swine , Swine, Miniature
16.
Neurosci Biobehav Rev ; 20(3): 445-52, 1996.
Article in English | MEDLINE | ID: mdl-8880734

ABSTRACT

The original notion that the brain represented an "immune-privileged" organ lacking the capability to produce an inflammatory response to an injury, would appear no longer tenable. Indeed, accumulating evidence during the last decade has shown that the CNS can mount a well-defined inflammatory response to a variety of insults including trauma, ischemia, transplantation, viral infections, toxins as well as neurodegenerative processes. Many aspects of this centrally-derived inflammatory response parallel, to some extent, the nature of such a reaction in the periphery. Through the recent application of molecular biological techniques, new concepts are rapidly emerging as to the molecular mechanisms associated with the development of brain injury. In particular, the importance of cytokines, especially TNF alpha and IL-1 beta, as well as adhesion molecules, has been emphasized in the propagation and maintenance of a CNS inflammatory response. This review will summarize recent observations as to the involvement of these inflammatory mediators in CNS injury and lay claim to the possibility that inhibitors of peripheral inflammation may also be of benefit in treating CNS injuries such as stroke, head trauma, Alzheimer's disease and multiple sclerosis.


Subject(s)
Brain Injuries/physiopathology , Cytokines/physiology , Animals , Inflammation/physiopathology , Tumor Necrosis Factor-alpha/metabolism
17.
Neurosci Biobehav Rev ; 21(1): 31-44, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8994207

ABSTRACT

A review of the effects of reducing brain temperature on ischemic brain injury is presented together with original data describing the systematic evaluation of the effects of brain cooling on brain injury produced by transient focal ischemia. Male spontaneously hypertensive rate were subjected to transient middle cerebral artery occlusion (TMCAO; 80, 120 or 160 min) followed by 24 h of reperfusion. During TMCAO, the exposed skull was bathed with isotonic saline at various temperatures to control skull and deeper brain temperatures. Rectal temperature was always constant at 37 degrees C. Initial studies indicated that skull temperature was decreased significantly (i.e. to 32-33 degrees C) just as a consequence of surgical exposure of the artery. Subsequent studies indicated that maintaining skull temperature at 37 degrees C compared to 32 degrees C significantly (p < 0.05) increased the infarct size following 120 or 160 min TMCAO. In other studies, 80 min TMCAO was held constant, but deeper brain temperature could be varied by regulating skull temperature at different levels. At 36-38 degrees C brain temperature, infarct volumes of 102 +/- 10 to 91 +/- 9 mm3 occurred following TMCAO. However, at a brain temperature of 34 degrees C, a significantly (p < 0.05) reduced infarct volume of 37 +/- 10 mm3 was observed. Absolutely no brain infarction was observed if the brain was cooled to 29 degrees C during TMCAO. Middle cerebral artery exposure and maintaining brain temperature at 37 degrees C without artery occlusion did not produce any cerebral injury. These data indicated the importance of controlling brain temperature in cerebral ischemia and that reducing brain temperature during ischemia produces a brain temperature-related decrease in focal ischemic damage. Brain cooling of 3 degrees C and 8 degrees C can provide dramatic and complete, respectively, neuroprotection from transient focal ischemia. Multiple mechanisms for reduced brain temperature-induced neuroprotection have been identified and include reduced metabolic rate and energy depletion, decreased excitatory transmitter release, reduced alterations in ion flux, and reduced vascular permeability, edema, and blood-brain barrier disruption. Cerebral hypothermia is clearly the most potent therapeutic approach to reducing experimental ischemic brain injury identified to date, and this is emphasized by the present data which demonstrate complete neuroprotection in transient focal stroke. Certainly all available information warrants the evaluation of brain cooling for potential implementation in the treatment of human stroke.


Subject(s)
Body Temperature/physiology , Brain Ischemia/therapy , Hypothermia/therapy , Animals , Humans , Male , Rats , Rats, Inbred SHR
18.
Neurosci Biobehav Rev ; 16(2): 219-33, 1992.
Article in English | MEDLINE | ID: mdl-1630732

ABSTRACT

A review of the sensitivity of genetically hypertensive rats to cerebral ischemia was presented together with original data describing the systematic comparison of the effects of focal ischemia (permanent and temporary with reperfusion) performed in hypertensive and normotensive rats (i.e., blood pressures verified in conscious instrumented rats). Microsurgical techniques were used to isolate and occlude the middle cerebral artery (MCAO) of spontaneously hypertensive (SHR), Sprague-Dawley (SD) and Wistar Kyoto (WKY) rats at the level of the inferior cerebral vein. Following permanent (24 h) MCAO, persistent and similar decreases in local microvascular perfusion (i.e., to 15.6 +/- 1.7% of pre-MCAO levels) were verified in the primary ischemic zone of the cortex for all strains using Laser-Doppler flowmetry. A contralateral hemiplegia that occurred following MCAO, evidenced by forelimb flexion and muscle weakness, was greater in SHR (neurological grade = 2.0 +/- 0.1) than SD (1.0 +/- 0.4) or WKY (0.7 +/- 0.4) rats (N = 7-9, p less than 0.05). SHR also exhibited sensory motor deficits following MCAO compared to sham-operation, with decreased normal placement response of the hindlimb (% normal = 20 vs. 83, N = 23-30, p decreased rota-rod (41 +/- 7 vs. 126 +/- 19 on rod, N = 10-15, p less than 0.05) and balance beam (25 +/- 5 vs. 116 +/- 29 s on beam, N = 5-7, p less than 0.05) performance. However, an index of general motor activity was not affected by permanent MCAO. Triphenyltetrazolium-stained forebrain tissue analyzed by planimetry revealed a significantly larger and more consistent cortical infarction in SHR (hemispheric infarction = 27.9 +/- 1.5%) compared to SD (15.4 +/- 4.1%) and WKY (4.0 +/- 2.4%) rats (N = 7-9, p less than 0.05), occupying predominantly the frontal and parietal areas. Also, a significant degree of ipsilateral hemispheric swelling (4.6 +/- 0.9%, N = 7-9, p less than 0.05) and increased brain water content (78.4 +/- 0.3% to 80.4 +/- 0.2%, N = 8-9, p less than 0.05) was identified in SHR that was not observed in SD or WKY rats. A novel model of temporary MCAO also was evaluated in the hypertensive and normotensive rat strains. Initially, the effect of increasing MCAO-time followed by 24 h reperfusion in SHR was studied. During temporary MCAO (20 to 300 min), persistent and stable decreases in local microvascular perfusion (i.e., to 15-20% of pre-MCAO levels) were verified in the primary ischemic zones of the cortex.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Brain Ischemia/pathology , Hypertension/pathology , Animals , Brain Ischemia/genetics , Humans , Hypertension/genetics , Rats , Rats, Inbred SHR , Rats, Inbred Strains , Rats, Inbred WKY
19.
Endocrinology ; 129(2): 965-9, 1991 Aug.
Article in English | MEDLINE | ID: mdl-1649752

ABSTRACT

Membranes prepared from the medullary region of the porcine kidney displayed high affinity, high density (Kd, 0.12 nM; binding capacity, 127 fmol/mg protein) receptors for calcitonin gene-related peptide (CGRP). Human CGRP (hCGRP), rat CGRP (rCGRP), and the hCGRP analog [hCGRP-(8-37)] competed for the binding of [125I]hCGRP, whereas salmon calcitonin (sCT) and CGRP-(22-37) were very weak in displacing [125I]hCGRP binding. In accordance with these binding data, CGRP stimulated adenylate cyclase activity in these membrane preparations in a concentration-dependent manner, with an EC50 similar to that of the Kd for binding. In the same preparations, sCT was ineffective in stimulating adenylate cyclase activity, suggesting that porcine kidney medullary membranes possess receptors specific for CGRP. Further hCGRP-(8-37), a CGRP antagonist, inhibited CGRP-stimulated adenylate cyclase activity in a competitive manner. Covalent cross-linking of [125I]hCGRP to these membranes resulted in the specific labeling of one major band at approximately 30,000 mol wt and two minor bands at about 58,000 and 78,000 mol wt. The presence of CGRP receptors and their coupling to adenylate cyclase suggest a role for CGRP in kidney function, such as local regulation of the microcirculation, electrolyte transport, or water homeostasis in the porcine kidney.


Subject(s)
Adenylyl Cyclases/metabolism , Kidney Medulla/metabolism , Receptors, Cell Surface/metabolism , Animals , Binding, Competitive , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/pharmacology , Cell Membrane/metabolism , Cross-Linking Reagents , Humans , Molecular Weight , Rats , Receptors, Calcitonin , Receptors, Cell Surface/analysis , Receptors, Cell Surface/chemistry , Second Messenger Systems , Swine
20.
Endocrinology ; 111(2): 608-13, 1982 Aug.
Article in English | MEDLINE | ID: mdl-7047152

ABSTRACT

To determine if the subnormal blood pressure recovery after hemorrhage in Brattleboro rats is due to secondary abnormalities in the renin-angiotensin or sympathetic nervous systems, we measured the hemodynamic, catecholamine, and renin activity responses to moderate acute hemorrhage in anesthetized Brattleboro rats. Results were compared to responses in groups of animals matched for either age or weight. Blood pressure recovery was significantly blunted (P less than 0.01) in Brattleboro rats compared to that in either control group, but heart rate responses were similar. Basal plasma norepinephrine was significantly higher in Brattleboro rats than in controls (P less than 0.001), but the response to hemorrhage was not significantly different. Both plasma epinephrine levels and renin activity were significantly higher before hemorrhage and increased more after hemorrhage in vasopressin-deficient animals. Plasma vasopressin in controls increased approximately 10-fold, reaching levels of 790 +/- 140 pg/ml in age-matched controls and 425 +/- 60 pg/ml in weight-matched controls. Vasopressin levels in Brattleboro rats were undetectable both before and after hemorrhage. We conclude from these data that the subnormal blood pressure recovery observed in vasopressin-deficient rats is not due to secondary abnormalities of the renin-angiotensin or sympathetic nervous systems, but, instead, is related more directly to the vasopressin deficiency.


Subject(s)
Hemodynamics , Hemorrhage/physiopathology , Renin-Angiotensin System , Vasopressins/deficiency , Animals , Blood Pressure , Heart Rate , Norepinephrine/blood , Rats , Renin/blood , Time Factors , Vasopressins/blood
SELECTION OF CITATIONS
SEARCH DETAIL