Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biochem Soc Trans ; 44(2): 606-12, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27068977

ABSTRACT

Since the identification of the proteinase-activated receptor (PAR) family as mediators of serine protease activity in the 1990s, there has been tremendous progress in the elucidation of their pathophysiological roles. The development of drugs that target PARs has been the focus of many laboratories for the potential treatment of thrombosis, cancer and other inflammatory diseases. Understanding the mechanisms of PAR activation and G protein signalling pathways evoked in response to the growing list of endogenous proteases has yielded great insight into receptor regulation at the molecular level. This has led to the development of new selective modulators of PAR activity, particularly PAR1. The mixed success of targeting PARs has been best exemplified in the context of inhibiting PAR1 as a new antiplatelet therapy. The development of the competitive PAR1 antagonist, vorapaxar (Zontivity), has clearly shown the value in targeting PAR1 in acute coronary syndrome (ACS); however the severity of associated bleeding with this drug has limited its use in the clinic. Due to the efficacy of thrombin acting via PAR1, strategies to selectively inhibit specific PAR1-mediated G protein signalling pathways or to target the second thrombin platelet receptor, PAR4, are being devised. The rationale behind these alternative approaches is to bias downstream thrombin activity via PARs to allow for inhibition of pro-thrombotic pathways but maintain other pathways that may preserve haemostatic balance and improve bleeding profiles for widespread clinical use. This review summarizes the structural determinants that regulate PARs and the modulators of PAR activity developed to date.


Subject(s)
Platelet Aggregation Inhibitors/pharmacology , Receptors, Proteinase-Activated/drug effects , Humans , Hydrolysis , Lactones/pharmacology , Lactones/therapeutic use , Ligands , Platelet Aggregation Inhibitors/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Receptors, Proteinase-Activated/metabolism , Signal Transduction , Thrombosis/drug therapy
2.
Trends Pharmacol Sci ; 30(1): 8-16, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19058861

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by activation of the coagulation cascade and it has long been suspected that coagulation is an essential component of this still largely idiopathic group of diseases. The realization that coagulation factors are not only passive mediators in the propagation of coagulation, but also actively engage different cell types by activating proteinase-activated receptors (PARs) has provided mechanistic insight into how coagulation cascade propagation might participate in the progression of IBD. The emergence of PAR(2) as a key player in the progression of IBD has focused attention on its agonist, coagulation factor Xa (FXa). Recent findings on FXa and PAR(2) link the coagulation cascade to the progression of IBD. Here, we propose that FXa-induced PAR(2) activation has an important role in orchestrating intestinal inflammatory and fibroproliferative responses, and that PAR(2) might provide a novel therapeutic target for the management of IBD.


Subject(s)
Blood Coagulation , Factor Xa/metabolism , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/physiopathology , Signal Transduction , Animals , Humans , Models, Biological , Receptors, Proteinase-Activated/drug effects , Receptors, Proteinase-Activated/metabolism
3.
Am Heart J ; 157(2): 253-62, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19185631

ABSTRACT

The platelet, once thought to be solely involved in clot formation, is now known to be a key mediator in various other processes such as inflammation, thrombosis, and atherosclerosis. Therefore, antiplatelet agents have become paramount in the prevention and management of various cardiovascular diseases. However, the currently most widely used antiplatelet drugs, aspirin and clopidogrel, have been shown to reduce the risk of serious vascular events only by approximately one quarter. Similarly, oral glycoprotein IIb/IIIa antagonists have been associated with excess mortality, thus restricting the use of parental glycoprotein IIb/IIIa antagonists to the treatment of acute clinical conditions. Thus, for the prevention of cardiovascular diseases, there is still a clinical need for antiplatelet drugs with higher antithrombotic efficacy but with safety profiles that allow for a preventive long-term administration. Thrombin signaling through the protease-activated receptors (PARs) has been shown to influence a wide range of physiologic and pathologic responses in cardiovascular systems. Thus, interference with PARs appears to be a promising strategy to develop new antiplatelet agents with higher efficacy. This review focuses on the cardiovascular actions of PARs that play a role in normal cardiovascular physiology and that are likely to contribute to cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/drug therapy , Platelet Aggregation Inhibitors/pharmacology , Receptors, Proteinase-Activated/drug effects , Cardiovascular System/drug effects , Humans , Platelet Aggregation Inhibitors/therapeutic use
4.
Br J Pharmacol ; 153 Suppl 1: S263-82, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18059329

ABSTRACT

Proteinases like thrombin, trypsin and tissue kallikreins are now known to regulate cell signaling by cleaving and activating a novel family of G-protein-coupled proteinase-activated receptors (PARs 1-4) via exposure of a tethered receptor-triggering ligand. On their own, short synthetic PAR-selective PAR-activating peptides (PAR-APs) mimicking the tethered ligand sequences can activate PARs 1, 2 and 4 and cause physiological responses both in vitro and in vivo. Using the PAR-APs as sentinel probes in vivo, it has been found that PAR activation can affect the vascular, renal, respiratory, gastrointestinal, musculoskeletal and nervous systems (both central and peripheral nervous system) and can promote cancer metastasis and invasion. In general, responses triggered by PARs 1, 2 and 4 are in keeping with an innate immune inflammatory response, ranging from vasodilatation to intestinal inflammation, increased cytokine production and increased or decreased nociception. Further, PARs have been implicated in a number of disease states, including cancer and inflammation of the cardiovascular, respiratory, musculoskeletal, gastrointestinal and nervous systems. In addition to activating PARs, proteinases can cause hormone-like effects by other signalling mechanisms, like growth factor receptor activation, that may be as important as the activation of PARs. We, therefore, propose that the PARs themselves, their activating serine proteinases and their associated signalling pathways can be considered as attractive targets for therapeutic drug development. Thus, proteinases in general must now be considered as 'hormone-like' messengers that can signal either via PARs or other mechanisms.


Subject(s)
Peptide Hydrolases/physiology , Receptors, Proteinase-Activated/physiology , Signal Transduction/physiology , Animals , Humans , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Receptors, Proteinase-Activated/antagonists & inhibitors , Receptors, Proteinase-Activated/drug effects , Signal Transduction/drug effects
5.
Brain Res ; 1210: 230-9, 2008 May 19.
Article in English | MEDLINE | ID: mdl-18410909

ABSTRACT

Periventricular germinal matrix hemorrhage is a devastating complication of preterm birth. Inflammation appears to play a role in brain damage after premature birth and hypoxia. The effects of rat blood plasma and serum on cytokine expression by cultured rat microglial cells were investigated. We analyzed mRNA expression levels of tumor necrosis factor (TNF)-alpha, interleukin-6 and protease activated receptor-1 and -4 by quantitative RT-PCR. Protein expression for TNFalpha was done using immunocytochemistry and ELISPOT assays. Plasma and serum had dose dependent toxic effects on microglia as measured by lactate dehydrogenase release assay and activated caspase-3 immunocytochemistry. High concentrations of plasma enhanced TNFalpha mRNA expression and protein production, while high concentrations of serum enhanced IL-6 mRNA expression. This study suggests that soluble components of blood might be differentially responsible for up regulating production of the cytokines TNFalpha and IL-6 by microglia from immature rodent brain.


Subject(s)
Blood Proteins/immunology , Cytokines/metabolism , Encephalitis/immunology , Microglia/immunology , Microglia/metabolism , Animals , Animals, Newborn , Birth Injuries/immunology , Birth Injuries/metabolism , Birth Injuries/physiopathology , Blood Proteins/pharmacology , Caspase 3/drug effects , Caspase 3/metabolism , Cells, Cultured , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/physiopathology , Cytokines/genetics , Dose-Response Relationship, Drug , Encephalitis/etiology , Encephalitis/metabolism , Immunohistochemistry , Interleukin-6/genetics , Interleukin-6/metabolism , L-Lactate Dehydrogenase/drug effects , L-Lactate Dehydrogenase/metabolism , Premature Birth/immunology , Premature Birth/metabolism , Premature Birth/physiopathology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Receptor, PAR-1/drug effects , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Receptors, Proteinase-Activated/drug effects , Receptors, Proteinase-Activated/genetics , Receptors, Proteinase-Activated/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics , Up-Regulation/immunology
6.
Curr Opin Pharmacol ; 7(6): 593-7, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17964216

ABSTRACT

Increased sensitivity of visceral nociceptive pathways contributes to symptoms in an array of clinical gastrointestinal conditions, however, the search for a consistently effective pharmacological agent to treat these conditions remain elusive. Modulation of visceral nociceptive pathways can occur at peripheral, spinal and supra-spinal sites and a dizzying array of potential drug targets exists. Till date, only tricyclic anti-depressants (TCAs) such as amitriptyline and, more recently, selective serotonin reuptake inhibitors (SSRIs) such as citalopram have demonstrated convincing visceral anti-nociceptive properties and clinical benefit in a limited population of patients with visceral hypersensitivity. Unfortunately, there is an incomplete understanding of the receptors and/or primary site of action at which these compounds exert their effects and significant side effects are often encountered. There is a continuing and concerted effort underway to develop target-specific visceral analgesic/anti-hyperalgesic compounds and the aim of this article is to provide a concise update on the most recent advances in this area.


Subject(s)
Neural Pathways/drug effects , Pain/physiopathology , Acid Sensing Ion Channels , Animals , Human Growth Hormone/analogs & derivatives , Human Growth Hormone/pharmacology , Humans , Membrane Proteins/drug effects , Nerve Tissue Proteins/drug effects , Probiotics , Receptors, Adrenergic, alpha-2/drug effects , Receptors, Proteinase-Activated/drug effects , Sodium Channels/drug effects , TRPV Cation Channels/drug effects
7.
Platelets ; 19(5): 352-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18791941

ABSTRACT

Recent studies indicate connections between periodontitis and atherothrombosis, and the periodontal pathogen Porphyromonas gingivalis has been found within atherosclerotic lesions. P. gingivalis-derived proteases, designated gingipains activate human platelets, probably through a "thrombin-like" activity on protease-activated receptors (PARs). However, the potential interplay between P. gingivalis and other physiological platelet activators has not been investigated. The aim of this study was to elucidate consequences and mechanisms in the interaction between P. gingivalis and the stress hormone epinephrine. By measuring changes in light transmission through platelet suspensions, we found that P. gingivalis provoked aggregation, whereas epinephrine alone never had any effect. Intriguingly, pre-treatment of platelets with a low, sub-threshold number of P. gingivalis (i.e. a density that did not directly provoke platelet aggregation) resulted in a marked aggregation response when epinephrine was added. This synergistic action was not inhibited by the cyclooxygenas inhibitor aspirin. Furthermore, fura-2-measurements revealed that epinephrine caused an intracellular Ca(2+) mobilization in P. gingivalis pre-treated platelets, whereas epinephrine alone had no effect. Inhibition of the arg-specific gingipains, but not the lys-specific gingipains, abolished the aggregation and the Ca(2+) response provoked by epinephrine. Similar results were achieved by separate blockage of platelet alpha(2)-adrenergic receptors and PARs. In conclusion, the present study shows that a sub-threshold number of P. gingivalis sensitizes platelets to epinephrine. We suggest that P. gingivalis-derived arg-specific gingipains activates a small number of PARs on the surface of the platelets. This leads to an unexpected Ca(2+) mobilization and a marked aggregation response when epinephrine subsequently binds to the alpha(2)-adrenergic receptor. The present results are consistent with a direct connection between periodontitis and stress, and describe a novel mechanism that may contribute to pathological platelet activation.


Subject(s)
Adhesins, Bacterial/physiology , Blood Platelets/microbiology , Cysteine Endopeptidases/physiology , Epinephrine/pharmacology , Platelet Aggregation/drug effects , Porphyromonas gingivalis/physiology , Apyrase/pharmacology , Aspirin/pharmacology , Blood Platelets/drug effects , Calcium Signaling/drug effects , Cyclooxygenase Inhibitors/pharmacology , Gingipain Cysteine Endopeptidases , Humans , In Vitro Techniques , Leupeptins/pharmacology , Nephelometry and Turbidimetry , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Porphyromonas gingivalis/enzymology , Porphyromonas gingivalis/pathogenicity , Protease Inhibitors/pharmacology , Pyrroles/pharmacology , Quinazolines/pharmacology , Receptors, Proteinase-Activated/drug effects , Receptors, Proteinase-Activated/physiology , Virulence , Yohimbine/pharmacology
8.
Hematol Oncol Clin North Am ; 21(1): 103-13, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17258121

ABSTRACT

The protease-activated receptors (PARs) are a unique family of vascular receptors that confer on cells an ability to sense, and respond to, local changes in the proteolytic environment. They are activated by serine proteases of the blood coagulation cascade, notably thrombin, and are linked to thrombotic and inflammatory effector pathways. In surgery with cardiopulmonary bypass (CPB), thrombin is generated in large quantities in the extracorporeal circuit and can exert systemic effects by way of platelet and endothelial PAR1. Aprotinin (Trasylol), a serine protease inhibitor used in cardiac surgery, preserves platelet function, and attenuates the inflammatory response by protecting the PAR 1 receptor on platelets and endothelium.


Subject(s)
Hemostasis , Inflammation , Receptors, Proteinase-Activated , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Aprotinin/pharmacology , Cardiopulmonary Bypass , Hemostasis/drug effects , Hemostasis/physiology , Humans , Inflammation/drug therapy , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Receptors, Proteinase-Activated/drug effects , Receptors, Proteinase-Activated/physiology , Thrombosis/drug therapy
9.
Curr Opin Pharmacol ; 6(4): 428-34, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16777485

ABSTRACT

Coagulation cascade and innate immunity are intimately linked in their endeavor to organize the body's response to injury. Protease-activated receptors (PARs) are important mediators of inflammatory response that can be activated by proteases of the coagulation cascade. Their recent discovery has shed new light on the crosstalk between coagulation and innate immunity. Recent studies have investigated the physiological relevance of PARs in the context of immunity and vascular injury, suggesting that these receptors could be used as therapeutic targets for the treatment of pathologies related to innate immunity, endothelial functions and coagulation processes.


Subject(s)
Blood Coagulation , Immunity, Innate , Receptors, Proteinase-Activated/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Arthritis/immunology , Arthritis/metabolism , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/metabolism , Hemostasis , Humans , Peptide Hydrolases/metabolism , Receptors, Proteinase-Activated/drug effects , Receptors, Proteinase-Activated/immunology , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/metabolism , Sepsis/immunology , Sepsis/metabolism , Thrombosis/blood , Thrombosis/immunology , Thrombosis/metabolism
10.
J Leukoc Biol ; 78(1): 202-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15788441

ABSTRACT

Human mononuclear phagocytes have recently been shown to express constitutively and even more so, upon stimulation with bacteria, fungi, lipopolysaccharide (LPS), zymosan, or thrombin platelet basic protein (PBP). This CXC chemokine as well as platelet factor 4 (PF4), which is located genomically at a short distance from the PBP, were previously considered to be specific markers for the megakaryocyte cell lineage. Both chemokines have signaling and antimicrobial activity. In the present studies, transcriptional and expressional regulation of PF4 and related chemokines was studied in human monocytes. As shown by quantitative mRNA analysis, Western blots, radioimmunoprecipitation of cell extracts, and immunofluorescence and quantitatively with enzyme-linked immunosorbent assay, human monocytes express PF4 in the same order of magnitude as the known, regulated CXC chemokine interleukin (IL)-8. Expression of PF4 is up-regulated at the mRNA and protein level by thrombin and mediated by proteinase-activated receptors (PARs), resulting in a 32- to 128-fold higher mRNA level and leading to an up-to-sixfold increase of the peptide concentration in monocyte culture supernatants. Thrombin and the synthetic ligand of PAR-1 and PAR-2, SFLLRN, also induced comparable increases in the levels of mRNA for PBP, IL-8, regulated on activation, normal T expressed and secreted (RANTES), monocyte chemoattractant protein-1, and macrophage-inflammatory protein-1alpha and increased synthesis of these chemokines as shown by immunofluorescence or a quantitative immunobead-based method. The induction of increased mRNA levels for all chemokines by SFLLRN was unsurpassed by LPS, zymosan, interferon-gamma (IFN-gamma), tumor necrosis factor alpha (TNF-alpha), and IL-1. Activation of monocytes through PARs represents an alternate activation mechanism, independent from IFN-gamma, TNF-alpha, or other signaling pathways.


Subject(s)
Chemotaxis, Leukocyte/immunology , Monocytes/immunology , Platelet Factor 4/immunology , Receptors, Proteinase-Activated/immunology , Signal Transduction/immunology , Cells, Cultured , Chemokine CCL2/drug effects , Chemokine CCL2/immunology , Chemokine CCL4 , Chemokine CCL5/immunology , Chemokines, CXC/immunology , Chemotaxis, Leukocyte/drug effects , Humans , Inflammation Mediators/pharmacology , Interleukin-8/immunology , Interleukin-8/metabolism , Macrophage Inflammatory Proteins/immunology , Monocytes/drug effects , Monocytes/metabolism , Peptide Fragments/pharmacology , Platelet Factor 4/genetics , RNA, Messenger/metabolism , Receptor, PAR-1/drug effects , Receptor, PAR-1/immunology , Receptor, PAR-2/drug effects , Receptor, PAR-2/immunology , Receptors, Proteinase-Activated/drug effects , Signal Transduction/drug effects , Thrombin/metabolism , Transcriptional Activation/genetics , Up-Regulation/drug effects , Up-Regulation/immunology
11.
Thromb Haemost ; 115(5): 905-10, 2016 05 02.
Article in English | MEDLINE | ID: mdl-26887783

ABSTRACT

Vorapaxar, a novel antiplatelet thrombin PAR-1 inhibitor, has been evaluated in the successful TRA2P trial and the failed TRACER trial. The drug is currently approved for post myocardial infarction and peripheral artery disease indications with concomitant use of clopidogrel and/or aspirin. The FDA ruled that the vorapaxar safety profile is acceptable. However, both trials revealed excess diplopia (double vision) usually reversible after vorapaxar. The diplopia risk appears to be small (about 1 extra case per 1,000 treated subjects), but real. Overall, there were 10 placebo and 34 vorapaxar diplopia cases (p=0.018) consistent for TRACER (2 vs 13 cases; p=0.010) and for TRA2P (8 vs 21 cases; p=0.018). Hence, we review the FDA-confirmed evidence and discuss potential causes and implications of such a surprising adverse association, which may be related to off-target PAR receptor mismodulation in the eye.


Subject(s)
Diplopia/chemically induced , Lactones/adverse effects , Platelet Aggregation Inhibitors/adverse effects , Pyridines/adverse effects , Receptor, PAR-1/antagonists & inhibitors , Receptors, Proteinase-Activated/drug effects , Animals , Aspirin/administration & dosage , Clopidogrel , Drug Approval , Eye/drug effects , Eye/metabolism , Humans , Lactones/administration & dosage , Peripheral Arterial Disease/drug therapy , Pyridines/administration & dosage , Randomized Controlled Trials as Topic , Receptors, Proteinase-Activated/metabolism , Ticlopidine/administration & dosage , Ticlopidine/analogs & derivatives , United States , United States Food and Drug Administration
12.
Biomed Pharmacother ; 59(3): 70-5, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15795099

ABSTRACT

The activation of the coagulation system in cancer patients is a well-known phenomenon responsible for recurrent clinical problems. A number of fascinating molecular mechanisms have been recognized showing that the tumor not only activates the coagulation system, but vice versa, activated coagulation proteins are able to induce molecular effects in tumor cells. The molecular basis is the expression of defined membrane receptors by tumor cells that are activated, for example, by thrombin. As the liberation of thrombin from prothrombin is one of the key events in coagulation, it's impact upon biological processes, such as cancerogenesis and metastasation, seems to be a regular pathophysiological consequence. These perceptions are not only interesting for the comprehension of cancerogenesis, metastasation, and clinical phenomena, but they also have a high impact upon modern strategies of tumor therapy. Especially, the development of clinically useful coagulation inhibitors, such as modern low molecular weight heparins or melagatran, created the possibility of therapies that combine cell biological approaches with apoptosis-inducing principals such as chemotherapy. Several clinical studies that demonstrate the implication of these strategies have already been published recently. In this article the cell biological basics for these approaches are reviewed.


Subject(s)
Anticoagulants/therapeutic use , Blood Coagulation , Hemostatics/pharmacology , Neoplasms/complications , Receptors, Proteinase-Activated/drug effects , Thrombin/physiology , Thrombosis , Blood Coagulation/drug effects , Blood Coagulation/physiology , Female , Humans , Male , Neoplasm Metastasis , Thrombosis/etiology , Thrombosis/physiopathology , Thrombosis/prevention & control
13.
Curr Opin Pharmacol ; 4(6): 551-6, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15525542

ABSTRACT

Serine proteases from the circulation, inflammatory cells, digestive glands and microorganisms can signal to cells by cleaving protease-activated receptors (PARs), a family of four G-protein-coupled receptors. Proteases cleave PARs at specific sites to expose tethered ligand domains that bind to and activate the cleaved receptors. Despite this irreversible mechanism of activation, PAR signaling is tightly regulated to prevent the uncontrolled stimulation of cells. Although PARs are found in all organ systems, protease signaling is of particular interest in the gastrointestinal tract, where proteases regulate neurotransmission, secretion, motility, epithelial permeability and intestinal inflammation, and can thus contribute to disease.


Subject(s)
Gastrointestinal Tract/physiology , Receptors, Proteinase-Activated/physiology , Serine Endopeptidases/physiology , Signal Transduction/drug effects , Animals , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/physiopathology , Models, Biological , Receptors, Proteinase-Activated/drug effects , Serine Endopeptidases/pharmacology
14.
J Leukoc Biol ; 76(5): 1010-8, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15316029

ABSTRACT

The antimicrobial activity of a number of chemokines has recently come into focus of research about innate immunity. We have previously shown that platelet basic protein (PBP), which gives rise to several antimicrobial peptides of platelets, is also expressed in human monocytes. In the present studies, we show that exposure of human monocytes to bacteria or microbial components (lipopolysaccharide and zymosan) induces a several-fold greater expression of derivates of PBP. Also, activation of proteinase-activated receptors (PARs) by thrombin or the synthetic peptide ligand SFLLRN of PAR-1 significantly increased PBP expression, presumably on the transcriptional level, as evidenced by higher mRNA levels. Derivates of PBP appeared to reach phago-lysosomes, as higher concentration was found in latex phagosomes isolated by a flotation method. By the gel-overlay technique, two bactericidal derivatives of PBP could be visualized, which were immunoreactive with anti-PBP antibody in Western blots. By matrix-assisted laser desorption/ionization time of flight and surface-enhanced laser desorption and ionization techniques, it was confirmed that the bands corresponded to PBP derivates. After immunofixation with a monoclonal antibody to PBP, the major peptide in zymosan-stimulated monocytes was identified to correspond by molecular weight to connective tissue-activating peptide III, which has been reported to be a major antimicrobial PBP derivate also in platelets. Our observations indicate that PBP and its derivates are constituents of the antimicrobial arsenal of human monocytes. Their increased expression after exposure to microorganisms allows a rapid host response to pathogens.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Bacterial Infections/immunology , Chemokines/metabolism , Monocytes/immunology , Cell Line , Chemokines/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Monocytes/metabolism , Peptide Fragments/pharmacology , Peptides/genetics , Peptides/metabolism , Phagosomes/drug effects , Phagosomes/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Proteinase-Activated/drug effects , Receptors, Proteinase-Activated/metabolism , Thrombin/pharmacology , Up-Regulation/drug effects , Up-Regulation/physiology , Zymosan/pharmacology , beta-Thromboglobulin
15.
Eur J Pharmacol ; 471(2): 141-7, 2003 Jun 20.
Article in English | MEDLINE | ID: mdl-12818702

ABSTRACT

Functions of thrombin as a modulator of inflammation and tissue repair are mediated by the proteinase-activated receptor (PAR) family. Some of these effects may be induced by activation of mast cells. To characterize the degranulation of rat peritoneal mast cells in response to PAR agonists, the effects of thrombin, trypsin and peptide agonists of PARs (PAR-AP, proteinase-activated receptor-activating peptides) on secretion were investigated. The release of beta-hexosaminidase by thrombin (0.01-1 microM) was concentration-dependent and mediated via PAR(1), as evidenced by cathepsin G (100 microM)-induced inactivation of PAR(1) and thrombin-stimulated PAR(1) desensitization. Trypsin (1 microM) accelerated histamine secretion. The PAR(1)-AP, TRAP (SFFLRN, 1-100 microM) and the PAR(2)-AP SLIGRL (5-100 microM) caused the release of histamine, and beta-hexosaminidase from inflammatory mast cells were obtained from a model of acute peritonitis in rats. Relative to the response to compound 48/80, the thrombin- and TRAP-induced release of beta-hexosaminidase was higher in inflammatory mast cells than in the control. This suggests that additional exposure of PAR(1) on mast cells to PAR agonists or an increase in PARs sensitivity to PAR agonists probably occurred during acute inflammation.


Subject(s)
Inflammation/physiopathology , Mast Cells/metabolism , Mast Cells/physiology , Receptors, Proteinase-Activated/agonists , Acute Disease , Animals , Cell Degranulation/drug effects , Dose-Response Relationship, Drug , Histamine/chemistry , Injections, Intraperitoneal , Ketotifen/administration & dosage , Ketotifen/pharmacokinetics , Male , Mast Cells/drug effects , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacokinetics , Peritoneum/cytology , Peritoneum/drug effects , Peritoneum/metabolism , Peritonitis/chemically induced , Rats , Rats, Wistar , Receptor, PAR-1/drug effects , Receptors, Proteinase-Activated/drug effects , Receptors, Thrombin/drug effects , Thioglycolates/administration & dosage , Thioglycolates/adverse effects , Thrombin/administration & dosage , Thrombin/pharmacokinetics , Trypsin/administration & dosage , Trypsin/pharmacokinetics , beta-N-Acetylhexosaminidases/chemistry , p-Methoxy-N-methylphenethylamine/administration & dosage , p-Methoxy-N-methylphenethylamine/pharmacokinetics
16.
World J Gastroenterol ; 20(34): 11977-84, 2014 Sep 14.
Article in English | MEDLINE | ID: mdl-25232234

ABSTRACT

Gastric cancer (GC) is the fourth most common cancer in the world and the second cause of cancer-related death. Gastric carcinogenesis is a multifactorial process, in which environmental and genetic factors interact to activate multiple intracellular signals thus leading to uncontrolled growth and survival of GC cells. One such a pathway is regulated by proteinase activated-receptors (PARs), seven transmembrane-spanning domain G protein-coupled receptors, which comprise four receptors (i.e., PAR-1, PAR-2, PAR-3, and PAR-4) activated by various proteases. Both PAR-1 and PAR-2 are over-expressed on GC cells and their activation triggers and/or amplifies intracellular pathways, which sustain gastric carcinogenesis. There is also evidence that expression of either PAR-1 or PAR-2 correlates with depth of wall invasion and metastatic dissemination and inversely with the overall survival of patients. Consistently, data emerging from experimental models of GC suggest that both these receptors can be important targets for therapeutic interventions in GC patients. In contrast, PAR-4 levels are down-regulated in GC and correlate inversely with the aggressiveness of GC, thus suggesting a negative role of this receptor in the control of GC. In this article we review the available data on the expression and role of PARs in GC and discuss whether manipulation of PAR-driven signals may be useful for interfering with GC cell behavior.


Subject(s)
Biomarkers, Tumor/metabolism , Receptors, Proteinase-Activated/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Drug Design , Humans , Molecular Targeted Therapy , Receptors, Proteinase-Activated/drug effects , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology
17.
J Physiol Sci ; 64(2): 119-28, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24421180

ABSTRACT

Proteases play a diverse role in health and disease. An excessive concentration of proteases has been found in the feces of patients with inflammatory bowel disease or irritable bowel syndrome and been implicated in the pathogenesis of such disorders. This study examined the effect of the serine protease, trypsin, on intestinal epithelial anion secretion when added to the luminal side. A mucosal-submucosal sheet of the mouse cecum was mounted in Ussing chambers, and the short-circuit current (I sc) was measured. Trypsin added to the mucosal (luminal) side increased I sc with an ED50 value of approximately 10 µM. This I sc increase was suppressed by removing Cl(-) from the bathing solution. The I sc increase induced by 10-100 µM trypsin was substantially suppressed by tetrodotoxin, and partially inhibited by a neurokinin-1 receptor antagonist, but not by a muscarinic or nicotinic ACh-receptor antagonist. The trypsin-induced I sc increase was also significantly inhibited by a 5-hydroxytryptamine-3 receptor (5-HT3) antagonist and substantially suppressed by the simultaneous addition of both 5-HT3 and 5-HT4 receptor antagonists. We conclude that luminal trypsin activates the enteric reflex to induce anion secretion, 5-HT and substance P playing important mediating roles in this secreto-motor reflex. Luminal proteases may contribute to the cause of diarrhea occurring with some intestinal disorders.


Subject(s)
Cecum/innervation , Chlorides/metabolism , Enteric Nervous System/drug effects , Intestinal Mucosa/innervation , Intestinal Secretions/drug effects , Trypsin/pharmacology , Animals , Cecum/metabolism , Electric Conductivity , Enteric Nervous System/physiology , Intestinal Mucosa/metabolism , Intestinal Secretions/metabolism , Male , Mice , Neurotransmitter Agents/pharmacology , Receptors, Proteinase-Activated/drug effects , Receptors, Proteinase-Activated/metabolism , Reflex/drug effects , Serotonin/metabolism , Substance P/metabolism , Time Factors
18.
J Cardiovasc Transl Res ; 7(2): 203-11, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24203054

ABSTRACT

The coagulation protease cascade plays an essential role in hemostasis. In addition, a clot contributes to host defense by limiting the spread of pathogens. Coagulation proteases induce intracellular signaling by cleavage of cell surface receptors called protease-activated receptors (PARs). These receptors allow cells to sense changes in the extracellular environment, such as infection. Viruses activate the coagulation cascade by inducing tissue factor expression and by disrupting the endothelium. Virus infection of the heart can cause myocarditis, cardiac remodeling, and heart failure. A recent study using a mouse model have shown that tissue factor, thrombin, and PAR-1 signaling all positively regulate the innate immune during viral myocarditis. In contrast, PAR-2 signaling was found to inhibit interferon-ß expression and the innate immune response. These observations suggest that anticoagulants may impair the innate immune response to viral infection and that inhibition of PAR-2 may be a new strategy to reduce viral myocarditis.


Subject(s)
Blood Coagulation , Myocarditis/metabolism , Myocytes, Cardiac/metabolism , Receptors, Proteinase-Activated/metabolism , Signal Transduction , Animals , Anticoagulants/therapeutic use , Blood Coagulation/drug effects , Humans , Immunity, Innate , Myocarditis/blood , Myocarditis/drug therapy , Myocarditis/immunology , Myocarditis/virology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/immunology , Myocytes, Cardiac/virology , Receptors, Proteinase-Activated/drug effects , Signal Transduction/drug effects
19.
J Physiol Pharmacol ; 63(6): 571-6, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23388472

ABSTRACT

To clarify the relationship between Helicobacter pylori (H. pylori), a risk factor for gastritis, peptic ulcer and gastric cancer, and proteinase-activated receptors (PARs) that contribute to inflammatory responses, we determined and characterized the biological activity of H. pylori components in the mammalian cells that express PARs. The activity of H. pylori extracts was assessed in distinct cell lines with high expression of PAR1 (RGM1 cells), PAR2 (A549 cells), or PAR2 and PAR4 (HCT-15 cells). A PAR1-activating peptide (AP), but not H. pylori extracts, caused prostaglandin E2 (PGE2) release in RGM1 cells. On the other hand, H. pylori extracts produced release of PGE2 and interleukin-8 (IL-8) in A549 and HCT-15 cells, respectively, as a PAR2-AP did. The activity of H. pylori extracts in A549 cells was not affected by a proteinase inhibitor or exposure to boiling, but abolished by inhibitors of lipopolysaccharide (LPS), IRAK-1/4 or NF-κB. The activity of H. pylori extracts in HCT-15 cells was partially suppressed by boiling or the proteinase inhibitor. In rat platelets that express PAR4 and PAR3, like a PAR4-AP, H. pylori extracts induced aggregation when assessed in platelet rich plasma, an effect unaffected by the proteinase inhibitor, but did not cause aggregation of washed rat platelets that responded to the PAR4-AP or thrombin. The present study thus shows the biological activities of H. pylori extracts in A549 and HCT-15 cells or rat platelets, and suggests that they are not mediated by any PAR-activating proteinases, but may involve the other pathogenic factors including LPS.


Subject(s)
Helicobacter pylori/metabolism , Receptors, Proteinase-Activated/metabolism , Animals , Bacterial Proteins/metabolism , Blood Platelets/metabolism , Cell Line, Tumor , Dinoprostone/metabolism , Hot Temperature , Humans , Interleukin-8/metabolism , Lipopolysaccharides/metabolism , Male , Oligopeptides/pharmacology , Platelet Aggregation , Protease Inhibitors/pharmacology , Protein Denaturation , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Receptors, Proteinase-Activated/drug effects , Receptors, Thrombin/metabolism , Thrombin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL