Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Pharm Biomed Anal ; 86: 100-11, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23995753

ABSTRACT

Therapeutic drug monitoring (TDM) of antiretrovirals requires accurate and precise analysis of plasma drug concentrations. This work describes a simple, fast and sensitive UPLC-MS/MS method for determination of the commonly used protease inhibitors such as amprenavir, atazanavir, darunavir, indinavir, lopinavir, ritonavir, saquinavir and tipranavir, tenofovir a nucleoside reverse transcriptase inhibitor (NRTI), the non-NRTI such as efavirenz, nevirapine, etravirine, the CCR5 antagonist maraviroc as well as the more recent antiretrovirals, the integrase inhibitors such as raltegravir, elvitegravir and the new direct acting anti-HCV boceprevir. Adapted deuterated internal standard was added to plasma aliquots (100µl) prior to protein precipitation with methanol and acetonitrile. This method employed ultra-performance liquid chromatography coupled to tandem mass spectrometry with electrospray ionization mode. All compounds eluted within 4.2-min run time. Calibration curves were validated, with correlation coefficients (r(2)) higher than 0.997, for analysis of therapeutic concentrations reported in the literature. Inter- and intra-assay variations were <15%. Evaluation of accuracy shows a deviation <15% from target concentration at each quality control level. No significant matrix effect was observed for any of the antiretroviral studied. This new validated method fulfills all criteria for TDM of 15 antiretrovirals and boceprevir drugs and was successfully applied in routine TDM of antiretrovirals.


Subject(s)
Anti-Retroviral Agents/blood , Tandem Mass Spectrometry/methods , Tandem Mass Spectrometry/standards , Adenine/analogs & derivatives , Adenine/blood , Chromatography, High Pressure Liquid/methods , Chromatography, High Pressure Liquid/standards , Cyclohexanes/blood , Humans , Maraviroc , Mass Spectrometry/methods , Mass Spectrometry/standards , Nitriles , Organophosphonates/blood , Proline/analogs & derivatives , Proline/blood , Pyridazines/blood , Pyrimidines , Pyrrolidinones/blood , Quinolones/blood , Raltegravir Potassium , Tenofovir , Time Factors , Triazoles/blood
2.
PLoS One ; 7(9): e45273, 2012.
Article in English | MEDLINE | ID: mdl-22984630

ABSTRACT

Although prostanoids are known to be involved in regulation of the spontaneous beating rate of cultured neonatal rat cardiomyocytes, the various subtypes of prostanoid receptors have not been investigated in detail. In our experiments, prostaglandin (PG)F(2α) and prostanoid FP receptor agonists (fluprostenol, latanoprost and cloprostenol) produced a decrease in the beating rate. Two prostanoid IP receptor agonists (iloprost and beraprost) induced first a marked drop in the beating rate and then definitive abrogation of beating. In contrast, the prostanoid DP receptor agonists (PGD(2) and BW245C) and TP receptor agonists (U-46619) produced increases in the beating rate. Sulprostone (a prostanoid EP(1) and EP(3) receptor agonist) induced marked increases in the beating rate, which were suppressed by SC-19220 (a selective prostanoid EP(1) antagonist). Butaprost (a selective prostanoid EP(2) receptor agonist), misoprostol (a prostanoid EP(2) and EP(3) receptor agonist), 11-deoxy-PGE(1) (a prostanoid EP(2), EP(3) and EP(4) receptor agonist) did not alter the beating rate. Our results strongly suggest that prostanoid EP(1) receptors are involved in positive regulation of the beating rate. Prostanoid EP(1) receptor expression was confirmed by western blotting with a selective antibody. Hence, neonatal rat cardiomyocytes express both prostanoid IP and FP receptors (which negatively regulate the spontaneous beating rate) and prostanoid TP, DP(1) and EP(1) receptors (which positively regulate the spontaneous beating rate).


Subject(s)
Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Animals, Newborn , Blotting, Western , Cells, Cultured , Cloprostenol/pharmacology , Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/pharmacology , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Hydantoins/pharmacology , Iloprost/pharmacology , Latanoprost , Myocytes, Cardiac/metabolism , Prostaglandin D2/pharmacology , Prostaglandins F, Synthetic/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin E, EP1 Subtype/agonists , Receptors, Prostaglandin E, EP1 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP1 Subtype/physiology , Receptors, Thromboxane/agonists , Receptors, Thromboxane/physiology
3.
Fundam Clin Pharmacol ; 23(3): 279-92, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19453760

ABSTRACT

Using an isolated non-working rat heart model, this study investigated the mechanisms of pharmacological pre-conditioning (PC) induced by P2Y receptor stimulation with pyridoxal-5'-phosphate (PLP). After 6-hydroxydopamine pretreatment and a 15-min stabilization period, isolated rat hearts were perfused for 25 min then subjected to 40 min of global ischemia and 30 min of reperfusion (I/R); exposed for 15 min to 0.05 microM PLP bracketed for 25 min with broad-spectrum P2 antagonists (suramin or PPADS) or with more specific P2Y antagonists (AMPalphaS or MRS2578), 1 microM each, followed by a 5-min PLP-free perfusion before I/R; treated during 25 min with either glybenclamide (GLY, 1 microM), 5-hydroxydecanoic acid (5-HD, 100 microM), U73122 (0.5 microM), H89 (1 microM), or KN93 (1 microM), with an infusion starting 5 min before PLP. The main endpoints were the rate-pressure product (RPP), creatine kinase (CK) release and area necrosis. Recovery of RPP, measured 5 min after reperfusion, was rapidly improved by PLP, blocked by the P2 antagonists, and decreased with the different inhibitors. Fifteen minutes after the end of ischemia, CK release reached maximal values in all groups. PLP provided significant protection, whereas the P2 antagonists, 5-HD, a mitochondrial selective K(ATP) antagonist and GLY a non-selective K(ATP) channel blocker, suppressed the protective effect on myocardial injury. The suppression of the cardioprotective effects of PLP by AMPalphaS, the PKA inhibitor (H89), and phospholipase C blocker (U73122) is in agreement with the P2Y11 receptor as a receptor for PLP-induced PC. The suppression of the cardioprotective effects of PLP by MRS2578 and U73122 is in agreement with the P2Y6 receptor as a receptor for PLP-induced PC. Pre-ischemic exposure to nanomolar concentrations of PLP is protective against I/R. P2Y11 and P2Y6 represents the most likely candidate receptors for PLP-induced cardiac PC.


Subject(s)
Ischemic Preconditioning, Myocardial/methods , Myocardial Reperfusion Injury/prevention & control , Pyridoxal Phosphate/pharmacology , Receptors, Purinergic P2/drug effects , Animals , Creatine Kinase/drug effects , Creatine Kinase/metabolism , Disease Models, Animal , Male , Necrosis/pathology , Rats , Rats, Wistar , Receptors, Purinergic P2/metabolism , Signal Transduction/drug effects
4.
Life Sci ; 82(7-8): 376-82, 2008 Feb 13.
Article in English | MEDLINE | ID: mdl-18222489

ABSTRACT

Gelatinase A (matrix metalloproteinase-2, MMP-2) binds to several proteins through its collagen-binding domains (CBDs). Surface plasmon resonance analysis revealed a strong interaction between CBD123 and thrombospondin-1 (TSP-1), with a K(D) value of 2x10(-9) M. CBD123, as well as individual domains, behave as competitive inhibitors of the TSP-1-directed endocytic clearance of active MMP-2, but not of its latent form, by HT1080 fibrosarcoma cells. Enhanced level of active MMP-2 in conditioned medium was associated to increased matrigel invasion. Similarly, GGWSHWSPWSS and GGWSHW peptides, as tryptophan-rich peptides within properdin-repeat motifs (TSRs) of TSP-1, promoted MMP-2 accumulation and cell invasiveness. Our data document the importance of TSP-1 in promoting MMP-2-mediated cancer cell invasion through interaction between CBDs of the enzyme and TSRs motifs of TSP-1.


Subject(s)
Collagen/metabolism , Endocytosis/physiology , Fibrosarcoma/pathology , Matrix Metalloproteinase 2/metabolism , Neoplasm Invasiveness/pathology , Thrombospondin 1/metabolism , Binding Sites , Cell Line, Tumor , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/metabolism , Fibroblasts , Fibrosarcoma/metabolism , Humans , Neoplasm Invasiveness/physiopathology , Peptide Fragments/metabolism , Protein Binding
5.
FASEB J ; 21(9): 1968-78, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17341689

ABSTRACT

Elastin peptides (EPs) generated by hydrolysis of elastic fibers by elastinolytic enzymes display a wide spectrum of biological activities. Here, we investigated their influence on rat heart ischemia-mediated injury using the Langendorff ex vivo model. EPs, i.e., kappa elastin, at 1.32- and 660-nM concentrations, when administered before the ischemia period, elicited a beneficial influence against ischemia by accelerating the recovery rate of heart contractile parameters and by decreasing significantly creatine kinase release and heart necrosis area when measured at the onset of the reperfusion. All effects were S-Gal-dependent, as being reproduced by (VGVAPG)3 and as being inhibited by receptor antagonists, such as lactose and V14 peptide (VVGSPSAQDEASPL). EPs interaction with S-Gal triggered NO release and activation of PI3-kinase/Akt and ERK1/2 in human coronary endothelial cells (HCAECs) and rat neonatal cardiomyocytes (RCs). This signaling pathway, as designated as RISK, for reperfusion injury salvage kinase pathway, was shown to be responsible for the beneficial influence of EPs on ischemia/reperfusion injury on the basis of its inhibition by specific pharmacological inhibitors. EPs survival activity was attained at a concentration averaging that present into the blood circulation, supporting the contention that these matrikines might offer a natural protection against cardiac injury in young and adult individuals. Such protective effect might be lost with aging, since we found that hearts from 24-month-old rats did not respond to EPs.


Subject(s)
Cardiotonic Agents/pharmacology , Elastin/pharmacology , Ischemic Preconditioning, Myocardial/methods , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Myocardial Reperfusion Injury/prevention & control , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/drug effects , beta-Galactosidase/physiology , Amino Acid Sequence , Animals , Binding Sites , Cardiotonic Agents/therapeutic use , Coronary Circulation/drug effects , Culture Media, Serum-Free/pharmacology , Drug Evaluation, Preclinical , Elastin/chemistry , Elastin/therapeutic use , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Heart/growth & development , Humans , Male , Molecular Sequence Data , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Protein Binding , Rats , Rats, Wistar , Structure-Activity Relationship , Ventricular Pressure/drug effects
6.
Fundam Clin Pharmacol ; 21(1): 35-43, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17227443

ABSTRACT

Ischemic heart disease in diabetic patients might be linked to the accumulation of advanced-glycation end products (AGEs). In ischemic rat hearts, expression of receptor for AGEs and its ligands is significantly enhanced and involved in cardiac ischemia/reperfusion (I/R) injury even in the absence of diabetes. It has recently been reported that diabetic human myocardium cannot be protected by preconditioning. In this context, our hypothesis was that beta1-adrenergic preconditioning might be altered in the presence of AGEs. Using an isolated non-working rat heart model, this study investigated the effect of AGEs on cardioprotection induced by transient beta1-adrenoceptor (beta1-AR) stimulation with xamoterol (Xa). After 6-hydroxydopamine (6-OHDA) pre-treatment and a 20-min stabilization period, hearts were perfused at constant pressure for 20 min, then subjected to 40 min of global ischemia and 30 min of reperfusion (I/R, Ctrl); and exposed to 0.01 microm Xa for 5 min framed with or without 15.2 microm albumin (Alb) or glycated albumin (Gly Alb). The main endpoints were the mean coronary flow (MCF), the left ventricular end-diastolic pressure (LVEDP), rate-pressure product (RPP) and creatine kinase (CK) release and necrosis area. XA induced an increase in the MCF after I/R (t = 85 min), a protective effect on the LVEDP, an improvement in RPP, a decrease of CK release during reperfusion and a reduction of necrotic area. The beneficial effects induced by Xa during reperfusion were suppressed by the administration of Gly Alb during Xa infusion, whereas Alb did not hamper Xa-induced protection. These results suggest that AGEs suppress the cardioprotection resulting from the activation of beta1-ARs and thus might contribute to cardiovascular damages seen in diabetic patients.


Subject(s)
Adrenergic beta-1 Receptor Agonists , Adrenergic beta-Agonists , Ischemic Preconditioning, Myocardial , Serum Albumin/pharmacology , Xamoterol , Albumins/pharmacology , Animals , Blood Pressure/drug effects , Coronary Circulation/drug effects , Creatine Kinase/metabolism , Glycation End Products, Advanced/pharmacology , Heart/drug effects , Heart/physiopathology , In Vitro Techniques , Male , Myocardium/metabolism , Myocardium/pathology , Rats , Rats, Wistar , Glycated Serum Albumin
7.
J Pharm Biomed Anal ; 42(5): 593-600, 2006 Nov 16.
Article in English | MEDLINE | ID: mdl-16842961

ABSTRACT

A specific and sensitive liquid chromatography-tandem mass spectrometric (LC-MS-MS) method using an ion trap spectrometer was developed for quantitation of nefopam and desmethyl-nefopam in human plasma. Nefopam, desmethyl-nefopam and the internal standard (ethyl loflazepate) were extracted in a single step with diethyl ether from 1 mL of alkalinized plasma. The mobile phase consisted of acetonitrile with 0.1% formic acid (50:50, v:v). It was delivered at a flow-rate of 0.3 mL/min. The effluent was monitored by MS-MS in positive-ion mode. Ionisation was performed using an electrospray ion source operating at 200 degrees C. Nefopam and desmethyl-nefopam were identified and quantified in full scan MS-MS mode using a homemade MS-MS library. Calibration curves were linear over the concentration range of 0.78-100 ng/mL with determination coefficients >0.996. This method was fast (total run time<6 min), accurate (bias<12.5%), and reproducible (intra- and inter-assay precision<17.5%) with a quantitation limit of 0.78 ng/mL. The high specificity and sensitivity achieved by this method allowed the determination of nefopam and desmethyl-nefopam plasma levels in patients following either intermittent or continuous intravenous administration of nefopam.


Subject(s)
Chromatography, Liquid , Nefopam/analogs & derivatives , Tandem Mass Spectrometry , Calibration , Humans , Injections, Intravenous , Nefopam/blood , Nefopam/metabolism , Reference Standards , Reproducibility of Results , Sensitivity and Specificity
8.
Biochem Biophys Res Commun ; 345(2): 681-7, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16696942

ABSTRACT

UVA irradiation, dose-dependently (5-20 J/cm2), was shown to impair the morphogenic differentiation of human microvascular endothelial cells (HMECs) on Matrigel. Parallely, UVA down-regulated the expression of MMP-2 and MT1-MMP, both at the protein and the mRNA levels. On the contrary, the production of MMP-1 and TIMP-1 by HMECs increased following UVA treatment. The inhibitory effect of UVA on MMP expression and pseudotubes formation was mediated by UVA-generated singlet oxygen (1O2). The contribution of MT1-MMP, but not TIMP-1, to the regulation of HMECs' angiogenic phenotype following UVA irradiation was suggested using elastin-derived peptides and TIMP-1 blocking antibody, respectively.


Subject(s)
Angiogenic Proteins/metabolism , Endothelial Cells/radiation effects , Endothelium, Vascular/radiation effects , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinases/metabolism , Skin/cytology , Ultraviolet Rays , Angiogenic Proteins/genetics , Dose-Response Relationship, Radiation , Down-Regulation/radiation effects , Elastin/pharmacology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Humans , Matrix Metalloproteinases, Membrane-Associated , Phenotype , Singlet Oxygen/pharmacology , Tissue Inhibitor of Metalloproteinase-1/pharmacology
9.
Anticancer Res ; 25(4): 2617-25, 2005.
Article in English | MEDLINE | ID: mdl-16080502

ABSTRACT

Matrikines, i.e. matrix fragments with cytokine-like properties, have been ascribed a major role in regulating tumour progression. The invasive front of melanoma is characterised by intense fragmentation of dermal elastic fibres. Elastase-mediated elastolysis liberates elastin fragments, i.e. elastokines, that stimulate several aspects of melanoma progression such as to enhance melanoma cell invasion through type I collagen or increase angiogenesis. Induced-membrane-type 1 metalloprotease (MT1-MMP) expression following elastin receptor (S-Gal) occupancy by elastokines is responsible for those biological activities. Several matrix-derived peptides with a GXXPG consensus sequence adopting a type VIII beta-turn conformation were as potent as elastokines in promoting angiogenesis in a Matrigel assay, and galectin-3 also contains several similar repeats within its N-terminal domain. We propose that S-Gal might constitute a novel therapeutic target for controlling melanoma progression.


Subject(s)
Elastin/metabolism , Melanoma/metabolism , Melanoma/pathology , Amino Acid Sequence , Disease Progression , Humans , Matrix Metalloproteinases/metabolism , Molecular Sequence Data , Neoplasm Invasiveness , Receptors, Cell Surface/metabolism
10.
Clin Chem ; 51(9): 1666-72, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16020498

ABSTRACT

BACKGROUND: Identification of sulfonylureas in blood may be useful in the evaluation of hypoglycemic crises of unknown origin. The aim of the present study was to develop a highly selective liquid chromatography-electrospray tandem mass spectrometry (MS-MS) method using an ion-trap detector for rapid screening, identification, and quantification of sulfonylureas in human plasma. METHODS: After standard liquid-liquid extraction with glisoxepide as an internal standard, 8 sulfonylureas (glibenclamide, glipizide, gliclazide, glibornuride, glimepiride, carbutamide, chlorpropamide, and tolbutamide) were eluted from a C18 column within 10 min with an isocratic mobile phase. Drugs were identified and quantified in full-scan MS-MS mode by use of a homemade MS-MS library. We used the assay in 134 cases of hypoglycemic crises of unknown origin. RESULTS: No ion suppression effect was noted for the analytes at their specific retention-time windows. For all drugs, assay validation showed good linearity (r2>0.990) and acceptable imprecision and recovery based on commonly used criteria of acceptance. The mean extraction recoveries were 63%-87% for 5 sulfonylureas but <45% for 3 (carbutamide, chlorpropamide, and tolbutamide). Nevertheless, the high sensitivity of the MS instrument made possible detection and quantification of all 8 drugs at subtherapeutic to toxic concentrations with good precision. Sulfonylureas were found in 9 hypoglycemic patients. CONCLUSION: The described assay method allows accurate, rapid identification and quantification of 8 sulfonylureas in human plasma and can be used for specific diagnosis of factitious hypoglycemia caused by ingestion of these drugs.


Subject(s)
Hypoglycemic Agents/blood , Sulfonylurea Compounds/blood , Chromatography, Liquid , Diagnosis, Differential , Female , Humans , Hypoglycemia/chemically induced , Hypoglycemia/diagnosis , Hypoglycemic Agents/poisoning , Male , Middle Aged , Munchausen Syndrome/chemically induced , Munchausen Syndrome/diagnosis , Spectrometry, Mass, Electrospray Ionization , Sulfonylurea Compounds/poisoning , Toxicology/methods
11.
J Pharm Biomed Anal ; 39(3-4): 587-92, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-15949908

ABSTRACT

A sensitive high-performance liquid chromatography-tandem mass spectrometric (HPLC-MS-MS) method, using an ion trap spectrometer, was developed for quantitation of bupivacaine in human plasma. Bupivacaine and an internal standard (ropivacaine) were extracted in a single step from 100 microL of alkalinized plasma with diethyl-ether. The mobile phase consisted of acetonitrile with 0.1% formic acid (50:50, v/v), and was delivered at a flow rate of 0.3 mL/min. The effluent was detected by MS-MS in positive ion mode. Ionisation was performed, using an electrospray ion source, operating at 200 degrees C. The selected reaction monitoring transitions m/z 289-->m/z 140 and m/z 275-->m/z 126 were chosen for bupivacaine and ropivacaine, respectively. Calibration curves were linear over the concentration range of 3.90-500 microg/L with determination coefficients >0.996. The method is accurate (bias <10%) and reproducible (intra-assay and inter-assay precision <15%), with a quantitation limit of 3.90 microg/L, using only 100 microL of plasma. The high specificity and sensitivity, achieved by this fast method (total run-time <3 min), allowed the determination of bupivacaine plasma levels in pediatric patients, following epidural administration of bupivacaine.


Subject(s)
Biological Assay/methods , Bupivacaine/analysis , Bupivacaine/chemistry , Bupivacaine/pharmacokinetics , Chromatography, Liquid/methods , Mass Spectrometry/methods , Acetonitriles/chemistry , Amides/analysis , Amides/pharmacokinetics , Anesthetics, Local/analysis , Anesthetics, Local/chemistry , Calibration , Chemistry, Pharmaceutical/methods , Drug Industry/methods , Formates/chemistry , Humans , Ropivacaine , Sensitivity and Specificity , Spectrometry, Mass, Electrospray Ionization , Temperature , Time Factors
12.
Cardiovasc Res ; 66(3): 530-42, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15914118

ABSTRACT

OBJECTIVE: Using an isolated non-working rat heart model, this study investigated the mechanisms of pharmacological preconditioning (PC) induced by transient beta1-adrenoreceptor (beta1-AR) stimulation with xamoterol (XA). METHODS: After 6-hydroxydopamine (6-OHDA) pretreatment and a 20-min stabilization period, hearts were perfused at constant pressure for 20 min then subjected to 40 min of global ischemia and 30 min of reperfusion (I/R, Ctrl); exposed to 0.01 microM XA for 5 min with or without 10 microM atenolol (ATE), a specific antagonist of beta1-AR, followed by a 15-min XA-free perfusion before I/R (PC, ATE-PC, respectively); treated during 20 min with either phosphoinositide (PI) 3-kinase inhibitors, LY-294002 (LY, 15 microM), or wortmaninn (WO, 0.1 microM); protein kinase C (PKC) inhibitor, GF-109203X (GF, 4 nM); or protein kinase A (PKA) inhibitor, H89 (H89, 1 microM), with an infusion starting 3 min before XA (LY-PC, WO-PC, GF-PC, and H89-PC, respectively). The main endpoints were the mean coronary flow (MCF), the left ventricular end-diastolic pressure (LVEDP), rate-pressure product (RPP), and creatine kinase (CK) release. RESULTS: XA induced an increase in the MCF after I/R (t 105 min) and a protective effect on the LVEDP, which were blocked by ATE and abolished with the different inhibitors. The transient increase in RPP following XA infusion was blocked by ATE and was not modified by the inhibitors except for H89. Recovery of RPP, measured 25 min after reperfusion, was improved by XA, blocked by ATE, and decreased with the different inhibitors. Fifteen minutes after the end of ischemia, CK release reached maximal values in all groups. XA provided significant protection whereas ATE and the four inhibitors suppressed XA-induced protection. CONCLUSION: The transient preischemic exposure to nanomolar concentrations of a beta1-AR agonist is protective against I/R. PI 3-kinase, PKC, and PKA are implicated in the trigger phase of PC. These observations were confirmed by Western blots.


Subject(s)
Adrenergic beta-Agonists/therapeutic use , Cyclic AMP-Dependent Protein Kinases/metabolism , Ischemic Preconditioning, Myocardial , Myocardium/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Androstadienes/pharmacology , Animals , Blotting, Western/methods , Chromones/pharmacology , Coronary Circulation/drug effects , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Heart Rate/drug effects , Isoquinolines/pharmacology , Male , Morpholines/pharmacology , Myocardial Reperfusion Injury/prevention & control , Oxidopamine/pharmacology , Perfusion , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Wistar , Sulfonamides/pharmacology , Wortmannin , Xamoterol/therapeutic use
13.
J Cell Sci ; 118(Pt 2): 343-56, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15632106

ABSTRACT

Elastin-derived peptides display a wide range of biological activities in a number of normal and transformed cells but their involvement in angiogenesis has not been reported. In the present study, we show that kappa-elastin and VGVAPG hexapeptide elastin motif accelerated angiogenesis in the chick chorio-allantoic membrane in an in vivo model. They also stimulated pseudotube formation from human vascular and microvascular endothelial cells in the matrigel and collagen models as well as cell migration in an in vitro wound healing assay. Confocal and scanning electron microscopy analyses revealed the main reorganization of actin filaments mediated by elastin-derived peptides and changes in cell shape that correlated with a decrease of the cell form factor determined by computerized image analysis. Such elastin-derived peptide effects were attributed to upregulation of proMT1-MMP and proMMP-2 expression and activation at both the mRNA and protein levels. Batimastat, an inhibitor of furin convertase and TIMP-2, but not TIMP-1, totally abolished the influence of elastin-derived peptides (EDPs) on cell migration and tubulogenesis, thus favoring the involvement of MT1-MMP in such processes. To assess its contribution to EDP-mediated angiogenesis further, we used a small interfering RNA (siRNA) approach for specifically silencing MT1-MMP in human microvascular endothelial cells. Four sets of 21 bp siRNA duplexes targeting MT1-MMP mRNA were synthesized by in vitro transcription. Two of them proved to inhibit MT1-MMP expression efficiently but did not affect MT2-, MT3- and MT5-MMP expression. Seventy-two hours after transfection with 25 nM siRNAs EDP-induced MT1-MMP expression at the mRNA and protein levels was decreased fourfold. In parallel, proMMP-2 activation was inhibited. A scrambled siRNA, used as a negative control, had no effect. Finally, the effect of elastin peptides on pseudotube formation in MT1-MMP-siRNA transfected cells was totally abolished. These data emphasise the crucial role of MT1-MMP in the elastin-induced angiogenic phenotype of endothelial cells.


Subject(s)
Cell Movement/drug effects , Elastin/pharmacology , Endothelium, Vascular/physiology , Metalloendopeptidases/metabolism , Neovascularization, Physiologic/physiology , Peptide Fragments/pharmacology , Animals , Cell Movement/physiology , Chick Embryo , Dose-Response Relationship, Drug , Elastin/chemistry , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/drug effects , Neovascularization, Physiologic/drug effects , Phenotype , RNA, Small Interfering/pharmacology , Time Factors , Up-Regulation
14.
J Biol Chem ; 279(52): 54944-51, 2004 Dec 24.
Article in English | MEDLINE | ID: mdl-15489233

ABSTRACT

The low density lipoprotein receptor-related protein (LRP) mediates the endocytic clearance of various proteinases and proteinase.inhibitor complexes, including thrombospondin (TSP)-dependent endocytosis of matrix metalloproteinase (MMP)-2 (or gelatinase A), a key effector of extracellular matrix remodeling and cancer progression. However, the zymogen of MMP-2 (pro-MMP-2) mostly occurs in tissues as a complex with the tissue inhibitor of MMPs (TIMP-2). Here we show that clearance of the pro-MMP-2.TIMP-2 complex is also mediated by LRP, because addition of receptor-associated protein (RAP), a natural LRP ligand antagonist, inhibited endocytosis and lysosomal degradation of (125)I-pro-MMP-2.TIMP-2. Both TIMP-2 and the pro-MMP-2 collagen-binding domain independently competed for endocytosis of (125)I-pro-MMP-2.TIMP-2 complex. Surface plasmon resonance studies indicated that pro-MMP-2, TIMP-2, and pro-MMP-2.TIMP-2 directly interact with LRP in the absence of TSP. LRP-mediated endocytic clearance of (125)I-pro-MMP-2 was inhibited by anti-TSP antibodies and accelerated upon complexing with TSP-1, but these treatments had no effect on (125)I-pro-MMP-2.TIMP-2 uptake. This implies that mechanisms of clearance by LRP of pro-MMP-2 and pro-MMP-2.TIMP-2 complex are different. Interestingly, RAP did not inhibit binding of (125)I-pro-MMP-2.TIMP-2 to the cell surface. We conclude that clearance of pro-MMP-2.TIMP-2 complex is a TSP-independent two-step process, involving (i) initial binding to the cell membrane in a RAP-insensitive manner and (ii) subsequent LRP-dependent (RAP-sensitive) internalization and degradation.


Subject(s)
Endocytosis/physiology , Low Density Lipoprotein Receptor-Related Protein-1/physiology , Matrix Metalloproteinase 2/metabolism , Thrombospondins/physiology , Tissue Inhibitor of Metalloproteinase-2/metabolism , Binding Sites , Cell Line , Cell Membrane/metabolism , Collagen/metabolism , Culture Media, Conditioned , Fibrosarcoma , Gene Expression , Humans , Matrix Metalloproteinase 2/genetics , Protein Precursors/genetics , Protein Precursors/metabolism , Recombinant Proteins , Thrombospondin 1/metabolism , Thrombospondin 1/pharmacology , Tissue Inhibitor of Metalloproteinase-2/genetics , Transfection , Tumor Cells, Cultured
15.
Crit Rev Oncol Hematol ; 49(3): 203-20, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15036261

ABSTRACT

Neoangiogenesis, the formation of new blood capillaries from pre-existing vessels, plays an important role in a number of physiological and pathological processes, particularly in tumor growth and metastasis. Extracellular proteolysis by matrix metalloproteinases or other neutral proteinases is an absolute requirement for initiating tumor invasion and angiogenesis. Cryptic segments or pre-existing domains within larger proteins, most of them belonging to the extracellular matrix, can be exposed by conformational changes and/or generated by partial enzymatic hydrolysis. They can positively or negatively regulate important functions of endothelial cells including adhesion, migration, proliferation, cell survival and cell-cell interactions. Such regulations by cryptic segments and proteolytic fragments led to the concept of matricryptins and matrikines, respectively. Matrix metalloproteinases and matrikines in conjunction with other pro- or anti-angiogenic factors might act in concert at any step of the angiogenesis process. A number of matrikines have been identified as potent anti-angiogenic factors, which could provide a new alternative to anti-proteolytic strategies for the development of anti-angiogenic therapeutic molecules aimed at inhibiting tumor growth and metastasis. Some of them are currently being investigated in clinical trials.


Subject(s)
Matrix Metalloproteinases/physiology , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/physiology , Peptides/physiology , Extracellular Matrix/metabolism , Neoplasm Invasiveness
16.
Biochem Pharmacol ; 67(4): 643-54, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14757164

ABSTRACT

C18 unsaturated fatty acids were here found to inhibit proMMP (matrix metalloproteinase)-3 activation by plasmin. This effect was suppressed by lysine ligand competitors, indicating that it was mediated by binding to kringle domains. Surface plasmon resonance analysis demonstrated that oleic acid interacted to a similar extent with plasmin and kringle 5 (KD values of 3.4 x 10(-8) and 5.9 x 10(-8)M) while interaction with kringles 1-2-3 was 10-fold lower. Furthermore, oleic acid stimulated the amidolytic activity of plasmin and mini-plasmin, but not micro-plasmin. Oleic acid also enhanced u-PA (urokinase-type plasminogen activator)-mediated plasminogen activation over 50-fold. Taken together, these data indicate that inhibition of plasmin-induced proMMP-3 activation by unsaturated fatty acids was mediated through their preferential binding to kringle 5. The influence of elaidic acid on the plasmin/MMP-3/MMP-1 proteolytic cascade was assessed ex vivo. Exogenous addition of plasmin to dermal fibroblasts or supplementation of gingival fibroblast culture medium with plasminogen triggered this cascade. In both instances, elaidic acid totally abolished proMMP-3 and proMMP-1 activation. Additionally, a significant decrease in lattice retraction and collagen degradation in a range similar to that obtained with Batimastat was observed when human gingival fibroblasts were cultured in plasminogen-containing type I collagen gels, indicative of the dual influence of unsaturated fatty acids on MMP activation and activity. In conclusion, unsaturated fatty acids or molecules with similar structures could be attractive target for the development of natural pharmacological inhibitors directed against plasmin and/or MMPs in different pathological contexts such, skin UV irradiation, vascular diseases and tumour growth and invasion.


Subject(s)
Enzyme Precursors/metabolism , Fatty Acids, Unsaturated/pharmacology , Fibrinolysin/antagonists & inhibitors , Metalloendopeptidases/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Kringles , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase Inhibitors , Plasminogen/metabolism , Urokinase-Type Plasminogen Activator/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...