Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cardiovasc Drugs Ther ; 27(2): 109-15, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23318690

ABSTRACT

BACKGROUND: Recent studies in rabbits have demonstrated that platelet P2Y12 receptor antagonists are cardioprotective, and that the mechanism is surprisingly not related to blockade of platelet aggregation but rather to triggering of the same signal transduction pathway seen in pre- and postconditioning. We wanted to determine whether this same cardioprotection could be documented in a primate model and whether the protection was limited to P2Y12 receptor antagonists or was a class effect. METHODS: Thirty-one macaque monkeys underwent 90-min LAD occlusion/4-h reperfusion. RESULTS: The platelet P2Y12 receptor blocker cangrelor started just prior to reperfusion significantly decreased infarction by an amount equivalent to that seen with ischemic postconditioning (p < 0.001). For any size of risk zone, infarct size in treated hearts was significantly smaller than that in control hearts. OM2, an investigational murine antibody against the primate collagen receptor glycoprotein (GP) VI, produced similar protection (p < 0.01) suggesting a class effect. Both cangrelor and OM2 were quite effective at blocking platelet aggregation (94 % and 97 %, respectively). CONCLUSIONS: Thus in a primate model in which infarct size could be determined directly platelet anti-aggregatory agents are cardioprotective. The important implication of these investigations is that patients with acute myocardial infarction who are treated with platelet anti-aggregatory agents prior to revascularization may already be in a postconditioned state. This hypothesis may explain why in recent clinical trials postconditioning-mimetic interventions which were so protective in animal models had at best only a modest effect.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Antibodies/administration & dosage , Myocardial Infarction/drug therapy , Platelet Aggregation Inhibitors/administration & dosage , Platelet Membrane Glycoproteins/immunology , Purinergic P2Y Receptor Antagonists/administration & dosage , Adenosine Monophosphate/administration & dosage , Animals , Blood Pressure/drug effects , Heart Rate/drug effects , Macaca fascicularis , Male , Myocardial Infarction/physiopathology , Platelet Aggregation/drug effects
2.
J Cardiovasc Pharmacol Ther ; 18(3): 251-62, 2013 May.
Article in English | MEDLINE | ID: mdl-23233653

ABSTRACT

BACKGROUND: Blockade of platelet activation during primary percutaneous intervention for acute myocardial infarction is standard care to minimize stent thrombosis. To determine whether antiplatelet agents offer any direct cardioprotective effect, we tested whether they could modify infarction in a rabbit model of ischemia/reperfusion caused by reversible ligation of a coronary artery. METHODS AND RESULTS: The P2Y12 (adenosine diphosphate) receptor blocker cangrelor administered shortly before reperfusion in rabbits undergoing 30-minute regional ischemia/3-hour reperfusion reduced infarction from 38% of ischemic zone in control hearts to only 19%. Protection was dose dependent and correlated with the degree of inhibition of platelet aggregation. Protection was comparable to that seen with ischemic postconditioning (IPOC). Cangrelor protection, but not its inhibition of platelet aggregation, was abolished by the same signaling inhibitors that block protection from IPOC suggesting protection resulted from protective signaling rather than anticoagulation. As with IPOC, protection was lost when cangrelor administration was delayed until 10 minutes after reperfusion and no added protection was seen when cangrelor and IPOC were combined. These findings suggest both IPOC and cangrelor may protect by the same mechanism. No protection was seen when cangrelor was used in crystalloid-perfused isolated hearts indicating some component in whole blood is required for protection. Clopidogrel had a very slow onset of action requiring 2 days of treatment before platelets were inhibited, and only then the hearts were protected. Signaling inhibitors given just prior to reperfusion blocked clopidogrel's protection. Neither aspirin nor heparin was protective. CONCLUSIONS: Clopidogrel and cangrelor protected rabbit hearts against infarction. The mechanism appears to involve signal transduction during reperfusion rather than inhibition of intravascular coagulation. We hypothesize that both drugs protect by activating IPOC's protective signaling to prevent reperfusion injury. If true, patients receiving P2Y12 inhibitors before percutaneous intervention may already be postconditioned thus explaining failure of recent clinical trials of postconditioning drugs.


Subject(s)
Cardiotonic Agents/pharmacology , Coronary Vessels/drug effects , Heart/drug effects , Myocardial Reperfusion Injury/prevention & control , Platelet Aggregation Inhibitors/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/metabolism , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/antagonists & inhibitors , Adenosine Monophosphate/pharmacology , Animals , Cardiotonic Agents/antagonists & inhibitors , Clopidogrel , Coronary Vessels/metabolism , Female , In Vitro Techniques , Ischemic Postconditioning , Male , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/therapy , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Perfusion , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Purinergic P2Y Receptor Antagonists/chemistry , Rabbits , Receptors, Purinergic P2Y12/chemistry , Signal Transduction/drug effects , Ticlopidine/analogs & derivatives , Ticlopidine/antagonists & inhibitors , Ticlopidine/pharmacology
3.
Handb Exp Pharmacol ; (204): 211-36, 2011.
Article in English | MEDLINE | ID: mdl-21695642

ABSTRACT

Intermittent claudication (IC) is one of the most frequent forms of lower extremity peripheral arterial disease (PAD) and is most commonly caused by arterial atherosclerosis. Its clinical manifestation includes fatigue, discomfort, or pain occurring in limb muscles due to exercise-induced ischemia, thus limiting the ability of IC patients to walk and exercise. In addition to lifestyle changes (diet, exercise, and smoking cessation), pharmacological treatments are needed. Pathologically, atherosclerotic lesions cause a mismatch in oxygen supply and metabolic demand in the leg muscles during walking/exercise. This subjects the muscles to repeated ischemia and reperfusion injury that can alter structure and oxidative metabolism, resulting in insufficient utilization of oxygen supply. Despite extensive research efforts, cilostazol and pentoxifylline are the only drugs indicated for relieving the symptoms of IC, with cilostazol demonstrating significant improvement in walking distance and quality of life in these patients. Originally developed as a PDE3 inhibitor, cilostazol was later found to have several other pharmacological actions, and its success has been attributed to its multifactorial actions on platelets, endothelium, smooth muscle, and lipid profiles. Using cilostazol as an example, we discuss the rationales and pitfalls of targeting PDEs in IC, and potential strategies for the development of new and more effective pharmacological treatments.


Subject(s)
Intermittent Claudication/drug therapy , Phosphodiesterase Inhibitors/therapeutic use , Atherosclerosis/etiology , Blood Platelets/enzymology , Drug Discovery , Endothelium, Vascular/enzymology , Humans , Intermittent Claudication/etiology , Muscle, Smooth, Vascular/enzymology , Nitric Oxide/physiology , Phosphoric Diester Hydrolases/analysis
4.
Handb Exp Pharmacol ; (204): 487-510, 2011.
Article in English | MEDLINE | ID: mdl-21695653

ABSTRACT

Protozoan parasites of the order kinetoplastida are the causative agents of three of the world's most important neglected human diseases: African trypanosomiasis, American trypanosomiasis, and leishmaniasis. Current therapies are limited, with some treatments having serious and sometimes lethal side effects. The growing number of cases that are refractory to treatment is also of concern. With few new drugs in development, there is an unmet medical need for new, more effective, and safer medications. Recent studies employing genetic and pharmacological techniques have begun to shed light on the role of the cyclic nucleotide phosphodiesterases in the life cycle of these pathogens and suggest that these important regulators of cyclic nucleotide signaling may be promising new targets for the treatment of parasitic diseases.


Subject(s)
Leishmaniasis/drug therapy , Phosphodiesterase Inhibitors/therapeutic use , Trypanosomiasis/drug therapy , Animals , Crystallization , Humans , Kinetoplastida/enzymology , Leishmaniasis/enzymology , Nucleotides, Cyclic/physiology , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/physiology , Signal Transduction/physiology , Trypanosomiasis/enzymology
5.
Article in English | MEDLINE | ID: mdl-21558761

ABSTRACT

Serial transmission electron microscopy of human megakaryocytes (MKs) revealed their polyploidization and gradual maturation through consecutive transition in characteristics of various organelles and others. At the beginning of differentiation, MK with ploidy 32N, e.g., has 16 centrosomes in the cell center surrounded by 32N nucleus. Each bundle of microtubules (MTs) emanated from the respective centrosome supports and organizes 16 equally volumed cytoplasmic compartments which together compose one single 32N MK. During the differentiation, single centriole separated from the centriole pair, i.e., centrosome, migrates to the most periphery of the cell through MT bundle, corresponding to a half of the interphase array originated from one centrosome, supporting one "putative cytoplasmic compartment" (PCC). Platelet demarcation membrane (DM) is constructed on the boundary surface between neighbouring PCCs. Matured PCC, composing of a tandem array of platelet territories covered by a sheet of DM is designated as protoplatelet. Eventually, the rupture of MK results in release of platelets from protoplatelets. (Communicated by Tadamitsu Kishimoto, M.J.A.).


Subject(s)
Blood Platelets/cytology , Cell Differentiation , Megakaryocytes/cytology , Models, Biological , Blood Platelets/ultrastructure , Centrioles/metabolism , Centrioles/ultrastructure , Humans , Megakaryocytes/ultrastructure , Polyploidy
6.
Eur J Pharmacol ; 651(1-3): 73-6, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21093427

ABSTRACT

Neurodegeneration is thought to be a component of schizophrenia pathology, and some antipsychotics appear to slow degenerative changes in patients. Aripiprazole, the first partial dopamine D(2) receptor agonist approved for the treatment of schizophrenia, is suggested to be neuroprotective based on non-clinical studies using transformed cell lines and in vivo stress and lesion paradigms. However, aripiprazole-induced neuroprotection has not been studied in a neuronal glutamate toxicity assay, which may model aspects of neurodegeneration occurring in schizophrenia. This study examined whether therapeutically relevant concentrations of aripiprazole protect rat embryonic cortical neurons from glutamate toxicity in biochemical and high-content imaging assays. Aripiprazole inhibited glutamate-induced neurotoxicity by 40% in a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay, in contrast to risperidone and olanzapine, which had little neuroprotective activity. This neuroprotective effect of aripiprazole was not mediated by the activation of serotonin 5-HT(1A) or dopamine D(2) receptors, Akt or glycogen-synthase kinase-3ß signaling (GSK-3ß), or through the inhibition of poly-ADP ribose polymerase (PARP). Further experiments are required to determine the biochemical nature of aripiprazole-induced neuroprotection and whether any such activity might have clinical relevance.


Subject(s)
Cerebral Cortex/cytology , Cytoprotection/drug effects , Glutamic Acid/toxicity , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Piperazines/pharmacology , Quinolones/pharmacology , Animals , Aripiprazole , Benzodiazepines/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Dopamine D2 Receptor Antagonists , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Molecular Imaging , Neurons/cytology , Olanzapine , Poly(ADP-ribose) Polymerase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, Serotonin, 5-HT1A/metabolism , Risperidone/pharmacology , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Signal Transduction/drug effects , Tetrazolium Salts/metabolism , Thiazoles/metabolism
7.
Circ J ; 74(1): 181-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19966507

ABSTRACT

BACKGROUND: The mechanisms underlying the ability of cilostazol to improve walking distance in patients with intermittent claudication (IC) are not fully understood, but may be related to its phosphodiesterase type 3 (PDE3) and adenosine uptake inhibition. In the present study the effect of cilostazol on blood flow and interstitial adenosine concentration was compared with that of the PDE3 inhibitor, milrinone, and the adenosine uptake inhibitor, draflazine. METHODS AND RESULTS: Rabbit gastrocnemius muscle blood flow was measured under resting, contracting and ischemic conditions. Interstitial adenosine was sampled by microdialysis. None of the drugs affected tissue blood flow at rest. Blood flow in electrically stimulated muscle was 2- to 3-fold higher in vehicle-, milrinone- and draflazine-treated animals. However, cilostazol caused an 8-fold increase. Ligation of the femoral artery decreased blood flow in the stimulated muscle in all groups to a similar degree. Cilostazol and draflazine increased the dialysate adenosine concentration during the first 10 min of muscle contraction, but had no effect during ischemia, most likely because of the high AMP deaminase activity in skeletal muscle. CONCLUSIONS: Cilostazol increases blood flow in the gastrocnemius muscle during contraction and it is this effect that may be partially responsible for the improved walking distance in IC patients. (Circ J 2010; 74: 181 - 187).


Subject(s)
Muscle Contraction/physiology , Muscle, Skeletal/blood supply , Regional Blood Flow/drug effects , Tetrazoles/pharmacology , Vasodilator Agents/pharmacology , AMP Deaminase/metabolism , Adenosine/antagonists & inhibitors , Adenosine/metabolism , Animals , Cilostazol , Dose-Response Relationship, Drug , Electric Stimulation , Male , Milrinone/pharmacology , Models, Animal , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Phosphodiesterase 3 Inhibitors , Piperazines/pharmacology , Rabbits
8.
Basic Res Cardiol ; 105(1): 119-28, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19669077

ABSTRACT

Ischemic pre- (IPC) and post- (IPOC) conditioning are very protective in laboratory animals, but it has not been possible to measure their anti-infarct potency in human hearts. Non-human primates are genetically closer to humans than other laboratory animals, but until now there have been no studies of IPC or IPOC in any primate species. Accordingly the left anterior descending coronary artery of cynomolgus monkeys was occluded for 90 min and reperfused for 4 h. In control animals, only 44% of the risk zone infarcted indicating cynomolgus myocardium is much more resistant to infarction than that of rabbits or rats. The regression line for the infarct-risk zone plot was very linear (r = 0.99), and intersected the risk zone axis at 0.82 cm3. Even small changes in infarct size could be detected as a shift in this line. Collateral flow in 12 monkeys was 6.6% of flow to normal myocardium and not a covariate of infarct size. IPC with two cycles of 10-min coronary occlusion/10-min reperfusion reduced infarction to near zero indicating that the innate resistance to infarction was not caused by constitutive preconditioning. Wortmannin, an antagonist of phosphatidylinositol 3-kinase (PI3-K), administered just before release of the 90-min coronary occlusion attenuated IPC's infarct-sparing effect by approximately 50% suggesting that PI3-K was involved in preconditioning's protection. IPOC with six cycles of 30-s reperfusion/30-s coronary reocclusion, a very protective protocol in most species, was much less protective than IPC. We conclude that ischemic preconditioning is extremely protective in cynomolgus hearts despite their sparse collateralization but, surprisingly, the protocol of IPOC used in this study offers less protection.


Subject(s)
Disease Models, Animal , Ischemic Preconditioning, Myocardial , Myocardial Infarction/prevention & control , Animals , Arrhythmias, Cardiac/etiology , Blood Gas Analysis , Blood Pressure , Coronary Circulation , Heart Rate , Macaca fascicularis , Male , Myocardial Infarction/complications , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology
9.
J Biomed Sci ; 16: 92, 2009 Oct 13.
Article in English | MEDLINE | ID: mdl-19825180

ABSTRACT

BACKGROUND: Adipogenesis is a complex process that involves many genes/proteins at different stages of differentiation. In order to identify genes critical for adipogenesis, we took a novel approach based on phenotype change of individual cell, to search for genes with regulatory roles in adipogenesis genome-wide in 3T3-L1 cells. METHODS: Lentivirus-based inducible random homologous knockdown was used for the screening of functional gene that altered lipid formation in the adipocyte during differentiation. RESULTS: In the present study, we reported the identification of an alternatively spliced mitochondrial oxodicarboxylate carrier (ODC), so named ODC-AS. ODC-AS is different from ODC by replacing 22 amino acids with 29 amino acids at the N-terminal. ODC was widely expressed in most tissues in mouse as determined by multi-tissue cDNA panel polymerase chain reaction. However, ODC-AS was only detected in adipose tissue and in iris and sclera-choroid complex of the eye. The expression of ODC-AS in 3T3-L1 was detected after the induction of differentiation, and reached a peak at day 4 and then reduced thereafter, whereas no ODC transcript detected in the cells neither before nor after differentiation. Knocking down of ODC-AS expression by RNA interference led to significant reduction in lipid accumulation as determined by triglyceride measurement and Nile Red staining, as well as adipogenic marker CEBPalpha, PPARgamma, aP2 and CD36. Although both ODC and ODC-AS are expressed in white and brown adipose tissues, only the expression of ODC-AS was down-regulated in brown adipose tissue by cold exposure. CONCLUSION: These results implicate that ODC-AS may promote lipid accumulation during adipocyte differentiation and play an important role in the regulation of lipid metabolism in adipose tissues.


Subject(s)
Adipogenesis/genetics , Alternative Splicing , Carboxylic Acids/metabolism , Mitochondrial Membrane Transport Proteins/physiology , Organic Anion Transporters/physiology , 3T3-L1 Cells , Adipocytes/cytology , Animals , Cell Differentiation , Gene Expression Profiling/methods , Gene Expression Regulation , Genome , Lentivirus/genetics , Lipids/chemistry , Mice , Mitochondrial Membrane Transport Proteins/genetics , Organ Specificity , Organic Anion Transporters/genetics , Phenotype
10.
Blood ; 111(4): 2007-14, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18056842

ABSTRACT

During blood feeding, mosquitoes inject saliva containing a mixture of molecules that inactivate or inhibit various components of the hemostatic response to the bite injury as well as the inflammatory reactions produced by the bite, to facilitate the ingestion of blood. However, the molecular functions of the individual saliva components remain largely unknown. Here, we describe anopheline antiplatelet protein (AAPP) isolated from the saliva of Anopheles stephensi, a human malaria vector mosquito. AAPP exhibited a strong and specific inhibitory activity toward collagen-induced platelet aggregation. The inhibitory mechanism involves direct binding of AAPP to collagen, which blocks platelet adhesion to collagen and inhibits the subsequent increase in intracellular Ca(2+) concentration ([Ca(2+)]i). The binding of AAPP to collagen effectively blocked platelet adhesion via glycoprotein VI (GPVI) and integrin alpha(2)beta(1). Cell adhesion assay showed that AAPP inhibited the binding of GPVI to collagen type I and III without direct effect on GPVI. Moreover, intravenously administered recombinant AAPP strongly inhibited collagen-induced platelet aggregation ex vivo in rats. In summary, AAPP is a malaria vector mosquito-derived specific antagonist of receptors that mediate the adhesion of platelets to collagen. Our study may provide important insights for elucidating the effects of mosquito blood feeding against host hemostasis.


Subject(s)
Collagen/pharmacology , Insect Proteins/pharmacology , Platelet Adhesiveness/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/physiology , Platelet Membrane Glycoproteins/physiology , Salivary Proteins and Peptides/pharmacology , Animals , Anopheles , Blood Platelets/drug effects , Blood Platelets/physiology , Insect Proteins/genetics , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/drug effects , Protein Binding , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Salivary Glands , Salivary Proteins and Peptides/genetics
11.
Cell Signal ; 19(8): 1765-71, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17482796

ABSTRACT

Phosphodiesterase type 3 (PDE3) is an important regulator of cAMP-mediated responses within the cardiovascular system. PDE3 exists as two subtypes: PDE3A and PDE3B, with distinct cellular and subcellular locations. Due to the lack of subtype-specific pharmacological tools, the definitive role of each subtype in regulating cardiovascular function has not been determined. In this study, we investigated platelet and cardiac function, using PDE3A and PDE3B gene knockout (KO) mice. Platelet-rich-plasma was prepared from the blood of KO and age-matched wild-type (WT) mice. PGE1 (1 microg/mL) almost completely inhibited aggregation of platelets from WT, PDE3A KO and PDE3B KO mice. In platelets from WT mice, cilostamide (100 microM), a selective PDE3 inhibitor, blocked collagen- and ADP-induced aggregation. In contrast, cilostamide had no effect on aggregation of platelets from PDE3A KO mice. In PDE3B KO mice, inhibition of collagen- and ADP-induced platelet aggregation was similar to that in WT mice. The resting intra-platelet cAMP concentration in platelets from PDE3A KO mice was twice that in the WT platelets. After PGE1 (0.1 microg/mL) stimulation, intra-cellular cAMP concentration was increased significantly more in platelets from PDE3A KO mice compared to WT mice. In vivo, PDE3A KO mice were protected against collagen/epinephrine-induced pulmonary thrombosis and death, while no such protection was observed in PDE3B KO mice. The heart rate of PDE3A KO mice was significantly higher, compared with age-matched WT mice, while that of PDE3B KO mice was similar to WT. There was no difference in cardiac contractility between PDE3A or PDE3B KO mice. Heart rate and contractility were increased in a similar dose-dependent fashion by isoproterenol in both types of KO mice. Cilostamide increased heart rate and contractility in WT and PDE3B KO but not in PDE3A KO mice. Compared to WT and PDE3B KO mice, cyclic AMP-PDE activity in membrane fractions prepared from the hearts of PDE3A KO mice was lower and not inhibited by cilostamide. The data suggest that PDE3A is the main subtype of PDE3 expressed in platelets and cardiac ventricular myocytes, and is responsible for the functional changes caused by PDE3 inhibition.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Blood Platelets/metabolism , Gene Expression Regulation, Enzymologic , Myocardium/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/analysis , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Animals , Cyclic AMP/analysis , Cyclic Nucleotide Phosphodiesterases, Type 3 , Mice , Mice, Knockout , Platelet Aggregation/physiology , Platelet-Rich Plasma/metabolism
12.
Arterioscler Thromb Vasc Biol ; 27(5): 1199-205, 2007 May.
Article in English | MEDLINE | ID: mdl-17322098

ABSTRACT

BACKGROUND: Inhibition of GPVI has been proposed as a useful antithrombotic strategy; however, in vivo proof-of-concept animal studies targeting GPVI are lacking. We evaluated a novel anti-human GPVI monoclonal antibody OM4 Fab in rats. METHODS AND RESULTS: OM4 Fab specifically inhibited collagen-induced aggregation of rat platelets in vitro with an IC50 of 20 to 30 microg/mL but not ADP and AA-induced platelet aggregation. After intravenous administration of OM4 Fab, a rapid inhibition of ex vivo platelet aggregation was observed with a gradual recovery within 60 to 90 minutes which corresponded to the decline in OM4 Fab plasma concentration and time-dependent decrease in platelet-bound OM4 Fab. In contrast to previous reports in mice, intravenous OM4 Fab did not deplete platelet GPVI. Injection of OM4 IgG caused acute thrombocytopenia. In a modified Folts model of cyclic flow reduction in rat carotid artery, the number of complete occlusions was significantly reduced by intravenous administration of OM4 Fab (20 mg/kg) before or after mechanical injury to the vessel, without prolongation of bleeding time. CONCLUSION: Fab fragment of the monoclonal antibody OM4 effectively inhibits collagen induced platelet aggregation in vitro and ex vivo, and in vivo thrombosis in rats without prolonging bleeding time. Antibodies against GPVI may have therapeutic potential, inhibiting thrombosis without prolonging bleeding time.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hemorrhage/epidemiology , Immunoglobulin Fab Fragments/immunology , Immunoglobulin G/immunology , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/antagonists & inhibitors , Thrombosis/prevention & control , Animals , Antibodies, Monoclonal/administration & dosage , Bleeding Time , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Hemorrhage/etiology , Incidence , Injections, Intravenous , Platelet Count , Platelet Membrane Glycoproteins/metabolism , Rats , Risk Factors , Thrombosis/blood , Thrombosis/immunology
13.
Thromb Res ; 119(3): 319-29, 2007.
Article in English | MEDLINE | ID: mdl-16566959

ABSTRACT

Recent progress in the understanding of thrombus formation has suggested an important role for glycoprotein (GP) VI in this process. To clarify the exact role in detail, it is necessary to use specific, high affinity inhibitory antibodies. However, possibly due to the conserved structure of GPVI among species, it has been difficult to obtain potent antibodies. In this study, we developed highly potent anti-human GPVI monoclonal antibodies using GPVI knockout mice for immunization. Fab fragments of these antibodies, named OM1 and OM2, potently inhibit collagen-induced platelet aggregation. The IC(50) values for OM1 and OM2 are 0.6+/-0.05 and 1.7+/-0.5 microg/mL, respectively, showing potency greater than, or equal to that of abciximab (1.7+/-0.3 microg/mL), an anti-GPIIb/IIIa antibody. Fab fragments of OM1 and OM2 also potently inhibit collagen-induced ATP release, thromboxane A(2) formation, and platelet adhesion to immobilized collagen under static and flow conditions. Interestingly, platelet aggregation induced with collagen-related peptide was potently inhibited by OM2 but not OM1, indicating that OM1 recognizes an epitope that is different from collagen-related peptide-binding site on GPVI. These results suggest that OM1 and OM2 may be useful tools to understand the role of GPVI in thrombus formation. Furthermore, these antibodies have the potential to be developed as a new class of therapeutic tool.


Subject(s)
Antibodies, Monoclonal/pharmacology , Immunoglobulin Fab Fragments/pharmacology , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Blood Platelets/cytology , Blood Platelets/metabolism , COS Cells , Chlorocebus aethiops , Humans , Immunization , Immunoglobulin Fab Fragments/immunology , Mice , Mice, Knockout , Platelet Adhesiveness/immunology , Platelet Aggregation/immunology , Platelet Membrane Glycoproteins/deficiency
14.
J Psychopharmacol ; 21(6): 620-7, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17092971

ABSTRACT

Aripiprazole, (+)terguride, OPC-4392 and (-)3-PPP have been classified as dopamine D(2) receptor partial agonists based largely on their activity in second messenger-based assays of dopamine D(2) receptor signalling. Nevertheless, signal transduction amplification might result in these compounds behaving as dopamine D(2) receptor full agonists at a more downstream level of signalling. We compared the intrinsic activity (E(max), expressed as a percentage of the maximal effect of dopamine) of aripiprazole, (+)terguride, OPC-4392 and (-)3-PPP using second (calcium (Ca(2+)) mobilization) and third (extracellular signal-regulated kinase 2 (ERK2) phosphoprotein expression) messenger readouts of cloned human dopamine D(2long) (hD(2L)) receptor signalling in CHO cells. These compounds were all less potent and displayed lower intrinsic activity in the Ca(2+) assay (aripiprazole = 24.3%, (+)terguride = 56.9%, OPC-4392 = 58.6% and (-)3-PPP = 75.1%), and aripiprazole (E(max) = 54.5%) displayed a substantially lower intrinsic activity than (+)terguride (E(max) = 92.3%), OPC-4392 (E(max) = 93.1%) and (-)3-PPP (E(max) = 101.1%) in the more downstream-based ERK2 phosphoprotein expression assay. These drug effects on Ca(2+) mobilization and ERK2 phosphoprotein expression were mediated through dopamine hD(2L) receptors, as they all were blocked by (-)raclopride, whereas (-)raclopride and other dopamine D(2) receptor antagonists (haloperidol, risperidone, ziprasidone, olanzapine, clozapine and quetiapine) were inactive on their own in both assays. These data are consistent with clinical evidence that only dopamine D(2) receptor partial agonists with a sufficiently low enough intrinsic activity will prove effective against the positive symptoms of schizophrenia, and also highlight the importance of using downstream-based assays in the discovery of novel D(2) receptor partial agonist therapeutics.


Subject(s)
Antipsychotic Agents/pharmacology , Calcium Signaling/drug effects , Dopamine Agonists/pharmacology , MAP Kinase Signaling System/drug effects , Receptors, Dopamine D2/agonists , Animals , Antipsychotic Agents/therapeutic use , Aripiprazole , CHO Cells , Cloning, Molecular , Cricetinae , Cricetulus , Dopamine/metabolism , Dopamine Agonists/therapeutic use , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans , Immunoblotting , Lisuride/analogs & derivatives , Lisuride/pharmacology , Microscopy, Confocal , Mitogen-Activated Protein Kinase 1/metabolism , Phosphorylation , Piperazines/pharmacology , Piperidines/pharmacology , Quinolones/pharmacology , Raclopride/pharmacology , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Schizophrenia/drug therapy , Schizophrenia/metabolism , Time Factors , Transfection
15.
Prog Neuropsychopharmacol Biol Psychiatry ; 31(2): 348-56, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17070976

ABSTRACT

Clinical evidence suggests that dopamine D(2) receptor partial agonists must have a sufficiently low intrinsic activity to be effective as antipsychotics. Here, we used dopamine D(2) receptor signaling assays to compare the in vitro functional characteristics of the antipsychotic aripiprazole with other dopamine D(2) receptor partial agonists (7-{3-[4-(2,3-dimethylphenyl)-piperazinyl]propoxy}-2(1H)-quinolinone [OPC-4392], (-)-3-(3-hydroxy-phenyl)-N-n-propylpiperidine [(-)3-PPP] and (+)terguride) and dopamine D(2) receptor antagonists. Aripiprazole and OPC-4392 were inactive in a guanosine-5'-O-(3-[(35)S]thio)-triphosphate ([(35)S]GTPgammaS) binding assay using Chinese Hamster Ovary (CHO) cell membranes expressing cloned human dopamine D(2Long) (hD(2L)) receptors, whereas (-)3-PPP and (+)terguride displayed low intrinsic activity. Aripiprazole also had no effect on [(35)S]GTPgammaS binding to CHO-hD(2L) cells, while OPC-4392, (-)3-PPP and (+)terguride were partial agonists. In contrast, aripiprazole, OPC-4392, (-)3-PPP, and (+)terguride were inactive in a [(35)S]GTPgammaS binding assay using rat striatal membranes. However, at a more downstream level of CHO-hD(2L) cell signalling, these drugs all behaved as dopamine hD(2L) receptor partial agonists, with aripiprazole displaying an intrinsic activity 2 to 3-fold lower (inhibition of forskolin-induced adenosine 3',5'-cyclic monophosphate accumulation) and almost half as high (enhancement of adenosine triphosphate-stimulated [(3)H]arachidonic acid release) as OPC-4392, (-)3-PPP and (+)terguride. Dopamine activity was blocked in each case by (-)raclopride, which was inactive on its own in every assay, as were the antipsychotics haloperidol, olanzapine, ziprasidone and clozapine. Together, these data, whilst preclinical in nature, are consistent with clinical evidence suggesting the favorable antipsychotic profile of aripiprazole, compared with the other clinically ineffective partial agonists, is dependent on its low intrinsic activity at dopamine D(2) receptors. This study also highlights the limitations of using [(35)S]GTPgammaS binding assays to identify dopamine D(2) receptor partial agonists.


Subject(s)
Cell Membrane/metabolism , Dopamine Agonists/pharmacology , Receptors, Dopamine D2/physiology , Signal Transduction/physiology , Animals , Arachidonic Acid/metabolism , Binding, Competitive , Cell Membrane/drug effects , Cells, Cultured , Corpus Striatum/cytology , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Humans , Male , Neurons/cytology , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sulfur Isotopes/pharmacokinetics , Transfection/methods
16.
Thromb Haemost ; 96(2): 167-75, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16894460

ABSTRACT

Recent progress in the understanding of thrombus formation has suggested an important role of glycoprotein (GP)VI. In contrast to its pivotal role in collagen-induced platelet activation, it has been suggested that its blockade does not induce massive bleeding tendency. To demonstrate the dissociation between inhibitory effect on platelet aggregation and bleeding by GPVI blockade, we examined the effects of Fab fragment of OM2, an anti-human GPVI monoclonal antibody on ex vivo collagen-induced platelet aggregation and skin bleeding time after intravenous injection in cynomolgus monkeys. In a dose-escalation study, OM2 potently (> 80%) inhibited collagen-induced platelet aggregation at the cumulative dose of 0.2 mg/kg with a slight prolongation of bleeding time (1.3 times baseline value). Furthermore, at 18.8 mg/kg, the highest dose tested, prolongation of bleeding time was still mild (1.9 times). In contrast, abciximab, Fab fragment of anti-GPIIb/IIIa antibody prolonged bleeding time by 5.0 times at 0.35 mg/kg, the lowest effective dose on platelet aggregation. In a pharmacodynamic study, a bolus injection of OM2 at 0.4 mg/kg produced potent inhibition of collagen-induced aggregation up to six hours after injection, showing longer half-life than that of abciximab. The injection of OM2 Fab did not induce thrombocytopenia and GPVI depletion in monkeys. These results suggest that blockade of GPVI by antibody can exert a potent inhibitory effect on collagen-induced platelet aggregation with a milder prolongation of bleeding time than blockade of GPIIb/IIIa. This study indicates that OM2 has the potential to be developed as a new class of therapeutic tool.


Subject(s)
Antibodies/chemistry , Bleeding Time/methods , Platelet Function Tests/methods , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/metabolism , Animals , Blotting, Western , Collagen/chemistry , Immunoglobulin G/chemistry , Macaca fascicularis , Platelet Adhesiveness , Platelet Aggregation , Platelet Membrane Glycoproteins/immunology , Time Factors
17.
Eur J Pharmacol ; 540(1-3): 53-6, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16730695

ABSTRACT

Dopamine potently increased calcium mobilization in Chinese hamster ovary cells expressing human dopamine D2Long receptors (CHO-D2L cells), and increased guanosine-5'-O-(3-[35S]thio)-triphosphate binding to CHO-D2L cell and rat striatal membranes. These effects of dopamine were blocked by the dopamine D2 receptor antagonist (-)raclopride. In contrast to the findings of a recent controversial study, phencyclidine, ketamine and dizocilpine (MK-801) lacked dopamine D2 receptor full agonist, partial agonist and antagonist activity in these assays, suggesting their psychotomimetic effects, and activity in rodent models of schizophrenia, are associated with N-methyl-d-aspartate receptor blockade rather than a direct interaction with dopamine D2 receptors.


Subject(s)
Dizocilpine Maleate/pharmacology , Ketamine/pharmacology , Phencyclidine/pharmacology , Receptors, Dopamine D2/metabolism , Animals , Binding, Competitive/drug effects , CHO Cells , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Cricetulus , Dopamine/pharmacology , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Gene Expression/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Raclopride/pharmacology , Rats , Receptors, Dopamine D2/genetics , Sulfur Radioisotopes
18.
Thromb Res ; 118(3): 371-80, 2006.
Article in English | MEDLINE | ID: mdl-16139873

ABSTRACT

Platelet glycoprotein VI (GPVI) is now considered to be a major player in platelet-collagen adhesive interactions leading to thrombus formation. GPVI blockade, or its depletion, has been shown in mice to result in complete protection against arterial thrombosis, without significant prolongation of bleeding time. GPVI may therefore represent a useful antithrombotic target. In order to reaffirm the role of GPVI in platelet-collagen interactions, we developed GPVI(null) mice by targeted disruption methodology. GPVI(null) mice platelets failed to respond to a high dose of fibrillar collagen, or convulxin, a GPVI agonist, but showed a normal response to other agonists such as ADP, PMA and arachidonic acid. We report, for the first time, that a proportion of GPVI(null) mice is protected against lethal thromboembolism, induced by the infusion of a mixture of collagen and epinephrine. Greater than 55% of GPVI(null) mice survived the challenge, whereas the maximal survival from the other genotypes was 17% (n=18 per genotype). Washed platelets obtained from GPVI(null) mice showed >90% reduction in adhesion to fibrillar collagen under static conditions. Platelet adhesion to collagen under dynamic conditions using a high shear rate (2600 s(-1)) was dramatically reduced using blood from GPVI(null) mice, while platelets from wild-type and heterozygous animals showed a similar amount of adhesion. Animals from each genotype had essentially similar tail bleeding time, suggesting that a complete deficiency of GPVI, at least in mice, does not result in an enhanced bleeding tendency. These observations clearly establish that blockade of GPVI may attenuate platelet-collagen interactions without adversely affecting the bleeding time.


Subject(s)
Bleeding Time , Blood Coagulation/drug effects , Fibrillar Collagens , Platelet Membrane Glycoproteins/metabolism , Pulmonary Embolism/chemically induced , Pulmonary Embolism/metabolism , Animals , Mice , Mice, Knockout , Platelet Membrane Glycoproteins/genetics , Pulmonary Embolism/prevention & control
19.
Cardiovasc Drugs Ther ; 19(1): 41-8, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15883755

ABSTRACT

PURPOSE: To determine the antiplatelet effect of cilostazol (Pletal) and its interaction with dipyridamole in in vitro and in vivo rabbit models, and to see if it can be dissociated from bleeding time prolongation. METHODS: In vitro collagen-induced platelet aggregation was measured by an impedance-based aggregometer. The in vivo antithrombotic effect was evaluated in a rabbit carotid artery cyclic flow reduction (CFR) model, in which repetitive thrombosis was induced by mechanical injuries of the artery and stenosis. Template bleeding time was determined in rabbit ear arterioles and hindlimb nail cuticles. RESULTS: In vitro platelet aggregation was slightly inhibited by 4 microM cilostazol (22 +/- 6%), and modestly by 13 microM (57 +/- 3% of aggregation). While dipyridamole itself up to 13 microM had no significant inhibition, it potentiated the effect from cilostazol: in the presence of 4 microM dipyridamole, 4 microM cilostazol inhibited aggregation by 47 +/- 6%. Dipyridamole also potentiated the CFR reducing effect of cilostazol: combination of dipyridamole (no effect by itself) and cilostazol at 1 microM decreased CFRs to levels achieved by 3-4 microM cilostazol alone. Bleeding times were similar in controls and animals treated with cilostazol, or with cilostazol and dipyridamole. In contrast, aspirin (4 mg/kg), while reducing CFRs, significantly increased bleeding time. CONCLUSION: These results suggest that dipyridamole potentiates the antiplatelet effect of cilostazol without prolongation of the bleeding time, implying a potential novel combination antithrombotic therapy.


Subject(s)
Carotid Artery Thrombosis/drug therapy , Dipyridamole/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation/drug effects , Tetrazoles/therapeutic use , Animals , Bleeding Time , Carotid Artery Thrombosis/blood , Cilostazol , Dipyridamole/administration & dosage , Dipyridamole/pharmacology , Disease Models, Animal , Drug Synergism , In Vitro Techniques , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/pharmacology , Rabbits , Tetrazoles/administration & dosage , Tetrazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...