Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
2.
Naunyn Schmiedebergs Arch Pharmacol ; 394(1): 107-115, 2021 01.
Article in English | MEDLINE | ID: mdl-32840651

ABSTRACT

Tryptase is a serine protease that is released from mast cells during allergic responses. Tryptase inhibitors are being explored as treatments for allergic inflammation in the skin and respiratory system, most notably asthma. Here we report direct tryptase inhibition by natural product compounds. Candidate inhibitors were identified by computational screening of a large (98,000 compounds) virtual library of natural product compounds for tryptase enzymatic site binding. Biochemical assays were used to validate the predicted anti-tryptase activity in vitro, revealing a high (four out of six) success rate for predicting binding using the computational docking model. We further assess tryptase inhibition by a biflavonoid scaffold, whose structure-activity relationship is partially defined by assessing the potency of structurally similar analogs.


Subject(s)
Biflavonoids/pharmacology , Biological Products/pharmacology , Tryptases/antagonists & inhibitors , Biflavonoids/chemistry , Biological Products/chemistry , Molecular Docking Simulation , Structure-Activity Relationship , Tryptases/metabolism
3.
J Neuroinflammation ; 17(1): 144, 2020 May 04.
Article in English | MEDLINE | ID: mdl-32366312

ABSTRACT

BACKGROUND: Cardiac arrest survivors suffer from neurological dysfunction including cognitive impairment. Cerebral mast cells, the key regulators of neuroinflammation contribute to neuroinflammation-associated cognitive dysfunction. Mast cell tryptase was demonstrated to have a proinflammatory effect on microglia via the activation of microglial protease-activated receptor-2 (PAR-2). This study investigated the potential anti-neuroinflammatory effect of mast cell tryptase inhibition and the underlying mechanism of PAR-2/p-p38/NFκB signaling following asphyxia-induced cardiac arrest in rats. METHODS: Adult male Sprague-Dawley rats resuscitated from 10 min of asphyxia-induced cardiac arrest were randomized to four separate experiments including time-course, short-term outcomes, long-term outcomes and mechanism studies. The effect of mast cell tryptase inhibition on asphyxial cardiac arrest outcomes was examined after intranasal administration of selective mast cell tryptase inhibitor (APC366; 50 µg/rat or 150 µg/rat). AC55541 (selective PAR-2 activator; 30 µg/rat) and SB203580 (selective p38 inhibitor; 300 µg/rat) were used for intervention. Short-term neurocognitive functions were evaluated using the neurological deficit score, number of seizures, adhesive tape removal test, and T-maze test, while long-term cognitive functions were evaluated using the Morris water maze test. Hippocampal neuronal degeneration was evaluated by Fluoro-Jade C staining. RESULTS: Mast cell tryptase and PAR-2 were dramatically increased in the brain following asphyxia-induced cardiac arrest. The inhibition of mast cell tryptase by APC366 improved both short- and long-term neurological outcomes in resuscitated rats. Such behavioral benefits were associated with reduced expressions of PAR-2, p-p38, NFκB, TNF-α, and IL-6 in the brain as well as less hippocampal neuronal degeneration. The anti-neuroinflammatory effect of APC366 was abolished by AC55541, which when used alone, indeed further exacerbated neuroinflammation, hippocampal neuronal degeneration, and neurologic deficits following cardiac arrest. The deleterious effects aggregated by AC55541 were minimized by p38 inhibitor. CONCLUSIONS: The inhibition of mast cell tryptase attenuated neuroinflammation, led to less hippocampal neuronal death and improved neurological deficits following cardiac arrest. This effect was at least partly mediated via inhibiting the PAR-2/p-p38/NFκB signaling pathway. Thus, mast cell tryptase might be a novel therapeutic target in the management of neurological impairment following cardiac arrest.


Subject(s)
Brain/pathology , Heart Arrest/complications , Hypoxia-Ischemia, Brain/etiology , Inflammation/metabolism , Signal Transduction/physiology , Tryptases/antagonists & inhibitors , Animals , Asphyxia/complications , Brain/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Inflammation/etiology , MAP Kinase Signaling System/physiology , Male , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Receptor, PAR-2/metabolism
4.
J Med Chem ; 63(6): 3004-3027, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32057241

ABSTRACT

ß-Tryptase, a homotetrameric serine protease, has four identical active sites facing a central pore, presenting an optimized setting for the rational design of bivalent inhibitors that bridge two adjacent sites. Using diol, hydroxymethyl phenols or benzoyl methyl hydroxamates, and boronic acid chemistries to reversibly join two [3-(1-acylpiperidin-4-yl)phenyl]methanamine core ligands, we have successfully produced a series of self-assembling heterodimeric inhibitors. These heterodimeric tryptase inhibitors demonstrate superior activity compared to monomeric modes of inhibition. X-ray crystallography validated the dimeric mechanism of inhibition, and compounds demonstrated high selectivity against related proteases, good target engagement, and tryptase inhibition in HMC1 xenograft models. Screening 3872 possible combinations from 44 boronic acid and 88 diol derivatives revealed several combinations that produced nanomolar inhibition, and seven unique pairs produced greater than 100-fold improvement in potency over monomeric inhibition. These heterodimeric tryptase inhibitors demonstrate the power of target-driven combinatorial chemistry to deliver bivalent drugs in a small molecule form.


Subject(s)
Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Tryptases/antagonists & inhibitors , Animals , Boronic Acids/chemistry , Boronic Acids/pharmacology , Crystallography, X-Ray , Female , Humans , Mice , Molecular Docking Simulation , Protein Conformation/drug effects , Protein Multimerization/drug effects , Tryptases/chemistry , Tryptases/metabolism
5.
Cell ; 179(2): 417-431.e19, 2019 Oct 03.
Article in English | MEDLINE | ID: mdl-31585081

ABSTRACT

Severe asthma patients with low type 2 inflammation derive less clinical benefit from therapies targeting type 2 cytokines and represent an unmet need. We show that mast cell tryptase is elevated in severe asthma patients independent of type 2 biomarker status. Active ß-tryptase allele count correlates with blood tryptase levels, and asthma patients carrying more active alleles benefit less from anti-IgE treatment. We generated a noncompetitive inhibitory antibody against human ß-tryptase, which dissociates active tetramers into inactive monomers. A 2.15 Å crystal structure of a ß-tryptase/antibody complex coupled with biochemical studies reveal the molecular basis for allosteric destabilization of small and large interfaces required for tetramerization. This anti-tryptase antibody potently blocks tryptase enzymatic activity in a humanized mouse model, reducing IgE-mediated systemic anaphylaxis, and inhibits airway tryptase in Ascaris-sensitized cynomolgus monkeys with favorable pharmacokinetics. These data provide a foundation for developing anti-tryptase as a clinical therapy for severe asthma.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Asthma/therapy , Mast Cells/enzymology , Mast Cells/immunology , Tryptases/antagonists & inhibitors , Tryptases/immunology , Adolescent , Allosteric Regulation/immunology , Animals , Cell Line , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Rabbits
6.
J Clin Invest ; 129(10): 4180-4193, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31265436

ABSTRACT

Dengue virus (DENV) infection causes a characteristic pathology in humans involving dysregulation of the vascular system. In some patients with dengue hemorrhagic fever (DHF), vascular pathology can become severe, resulting in extensive microvascular permeability and plasma leakage into tissues and organs. Mast cells (MCs), which line blood vessels and regulate vascular function, are able to detect DENV in vivo and promote vascular leakage. Here, we identified that a MC-derived protease, tryptase, is consequential for promoting vascular permeability during DENV infection, through inducing breakdown of endothelial cell tight junctions. Injected tryptase alone was sufficient to induce plasma loss from the circulation and hypovolemic shock in animals. A potent tryptase inhibitor, nafamostat mesylate, blocked DENV-induced vascular leakage in vivo. Importantly, in two independent human dengue cohorts, tryptase levels correlated with the grade of DHF severity. This study defines an immune mechanism by which DENV can induce vascular pathology and shock.


Subject(s)
Capillary Permeability , Dengue Virus/metabolism , Dengue/enzymology , Endothelium, Vascular/enzymology , Mast Cells/enzymology , Shock/enzymology , Tight Junctions/metabolism , Tryptases/metabolism , Animals , Benzamidines , Cell Line , Dengue/drug therapy , Dengue/pathology , Dengue/virology , Endothelium, Vascular/pathology , Endothelium, Vascular/virology , Guanidines/pharmacology , Humans , Mast Cells/pathology , Mast Cells/virology , Mice , Shock/drug therapy , Shock/pathology , Shock/virology , Tight Junctions/pathology , Tryptases/antagonists & inhibitors , Tryptases/genetics
7.
Acc Chem Res ; 52(6): 1709-1720, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31150198

ABSTRACT

The guanidinium moiety, which is present in active sites of many enzymes, plays an important role in the binding of anionic substrates. In addition, it was also found to be an excellent binding motif for supramolecular chemistry. Inspired by Nature, scientists have developed artificial receptors containing guanidinium scaffolds that bind to a variety of oxoanions through hydrogen bonding and charge pairing interactions. However, the majority of binding studies is restricted to organic solvents. Polyguanidinium based molecules can form efficient complexes in aqueous solvents due to strong electrostatic interactions. However, they only have moderate association constants, which are significantly decreased in the presence of competing anions and salts. Hence, to improve the binding affinity of the guanidinium moiety, our group developed the cationic guanidiniocarbonyl pyrrole (GCP) moiety. This rigid planar analogue binds efficiently to oxoanions, like carboxylates even in aqueous solvents. The lower p Ka value (7-8) of GCP compared to guanidinium derivatives (p Ka 13) favors the formation of strong, hydrogen bonded ion pairs. In addition, carboxylate binding is further enhanced by additional amide hydrogen bond donors located at the five position of the pyrrole core. Moreover, the design has allowed for introducing secondary interactions between receptor side chains and guest molecules, which allows for optimizing binding specificity and selectivity. The spectroscopic data confirmed stabilization of guanidiniocarbonyl pyrrole/oxoanion complexes through a combination of ion pairing and multiple hydrogen bonding interactions. The key role of the ionic interaction in a polar solvent, is demonstrated by a zwitterion derivative of the guanidiniocarbonyl pyrrole, which self-assembles in both dimethyl sulfoxide and pure water with association constants of K > 1010 M-1 and K = 170 M-1, respectively. In this Account, we discuss strategies for making GCP functionalized compounds, in order to boost their ability to bind oxoanions. Then we explore how these building blocks have been incorporated into different synthetic molecules and peptide sequences, highlighting examples that demonstrated the versatility of this binding scaffold. For instance, the high oxoanion binding property of GCP-based compounds was exploited to generate a detectable signal for sensing applications, thus improving selectivity and sensitivity in aqueous solution. Moreover, peptides and molecules containing GCP have shown excellent gene transfections properties. Furthermore, the self-assembly and zwitterionic behavior of zwitterionic GCP analogues was used to develop variety of supramolecular architectures such as stable supramolecular ß-helix structure, linear supramolecular oligomers, one-dimensional rods or two-dimension sheets, fibers, vesicles, soft nanospheres, as well as stimuli responsive supramolecular gels.


Subject(s)
Guanidines/chemistry , Pyrroles/chemistry , Receptors, Artificial/chemistry , Arginine/chemistry , Carboxylic Acids/chemistry , DNA/chemistry , Enzyme Inhibitors/chemistry , Hydrogen Bonding , Peptides/chemistry , RNA/chemistry , Tryptases/antagonists & inhibitors
8.
Pharmacol Ther ; 199: 91-110, 2019 07.
Article in English | MEDLINE | ID: mdl-30877022

ABSTRACT

Mast cells are tissue-resident cells, which have been proposed to participate in various inflammatory diseases, among them the cardiovascular diseases (CVDs). For mast cells to be able to contribute to an inflammatory process, they need to be activated to exocytose their cytoplasmic secretory granules. The granules contain a vast array of highly bioactive effector molecules, the neutral protease tryptase being the most abundant protein among them. The released tryptase may act locally in the inflamed cardiac or vascular tissue, so contributing directly to the pathogenesis of CVDs. Moreover, a fraction of the released tryptase reaches the systemic circulation, thereby serving as a biomarker of mast cell activation. Actually, increased levels of circulating tryptase have been found to associate with CVDs. Here we review the biological relevance of the circulating tryptase as a biomarker of mast cell activity in CVDs, with special emphasis on the relationship between activation of mast cells in their tissue microenvironments and the pathophysiological pathways of CVDs. Based on the available in vitro and in vivo studies, we highlight the potential molecular mechanisms by which tryptase may contribute to the pathogenesis of CVDs. Finally, the synthetic and natural inhibitors of tryptase are reviewed for their potential utility as therapeutic agents in CVDs.


Subject(s)
Cardiovascular Diseases/metabolism , Mast Cells/enzymology , Tryptases/metabolism , Animals , Biomarkers/metabolism , Cardiovascular Diseases/drug therapy , Humans , Mast Cells/physiology , Tryptases/antagonists & inhibitors , Tryptases/chemistry
9.
Pharmacology ; 102(5-6): 233-243, 2018.
Article in English | MEDLINE | ID: mdl-30134249

ABSTRACT

ß-Tryptase is released from mast cells upon degranulation in response to allergic and inflammatory stimuli. Human tryptase is a homotetrameric serine protease with 4 identical active sites directed toward a central pore. These active sites present an optimized scenario for the rational design of bivalent inhibitors, which bridge 2 adjacent active sites. Using (3-[1-acylpiperidin-4-yl]phenyl)methanamine as the pharmacophoric core and a disiloxane linker to span 2 active sites we have successfully produced a novel bivalent tryptase inhibitor, compound 1a, with a comparable profile to previously described inhibitors. Pharmacological properties of compound 1a were studied in a range of in vitro enzymic and cellular screening assays, and in vivo xenograft models. This non-peptide inhibitor of tryptase demonstrated superior activity (IC50 at 100 pmol/L tryptase = 1.82 nmol/L) compared to monomeric modes of inhibition. X-ray crystallography validated the dimeric mechanism of inhibition, and 1a demonstrated good oral bioavailability and efficacy in HMC-1 xenograft models. Furthermore, compound 1a demonstrated extremely slow off rates and high selectivity against-related proteases. This highly potent, orally bioavailable and selective inhibitor of human tryptase will be an invaluable tool in future studies to explore the therapeutic potential of attenuating the activity of this elusive target.


Subject(s)
Mast Cells/drug effects , Silanes/chemistry , Silanes/pharmacology , Tryptases/antagonists & inhibitors , Animals , Cell Line , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Design , Humans , Immunohistochemistry , Male , Mast Cells/enzymology , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Pharmacokinetics , Silanes/analysis , Silanes/pharmacokinetics
10.
Int Arch Allergy Immunol ; 177(3): 199-206, 2018.
Article in English | MEDLINE | ID: mdl-30021208

ABSTRACT

BACKGROUND: Mast cells are key immune effector cells which release chemokines, proteases, and other inflammatory mediators upon activation by immunological stimuli. The aim of this study was to investigate the effects of co-releasing proteases on the kinetics of release of the chemokine monocyte chemoattractant protein-1 (MCP-1) in immunoglobulin E (IgE)-mediated activation of human mast cells. METHODS: Homogenous populations of mature and functional primary human mast cells were generated from CD34+ progenitors originated from buffy coats of healthy adult donors. The releases of MCP-1 from human mast cells in basal conditions and in response to FcεRI cross-linking were assessed at different time points. The effects of different types of protease inhibitors on MCP-1 release from these mast cells under stimulated or unstimulated conditions were also investigated. RESULTS: Cultured human mast cells released MCP-1 in basal conditions and its levels increased in a time-dependent manner. When stimulated by FcεRI cross-linking, the levels of MCP-1 detected in the medium gradually decreased over time after the initial peak induction. Such a decline in MCP-1 levels after IgE-dependent activation was completely prevented by pretreatment with a cocktail of protease inhibitors or the specific tryptase inhibitor APC366. CONCLUSIONS: Direct regulation of MCP-1 expression by co-release of tryptase in cultured human mast cells upon IgE-dependent activation demonstrates a role of the serglycin:serine protease axis in modulation of inflammatory reactions through proteolytic degradation of mediators such as chemokines.


Subject(s)
Chemokine CCL2/metabolism , Immunoglobulin E/immunology , Mast Cells/immunology , Proteoglycans/metabolism , Serine Proteases/metabolism , Tryptases/metabolism , Vesicular Transport Proteins/metabolism , Cell Degranulation/immunology , Cells, Cultured , Histamine Release/immunology , Humans , Mast Cells/physiology , Protease Inhibitors/pharmacology , Receptors, IgE/immunology , Receptors, IgE/metabolism , Tryptases/antagonists & inhibitors
11.
Clin Exp Med ; 18(3): 319-323, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29492715

ABSTRACT

Mast cells are recognized as critical components of the tumor stromal microenvironment in several solid and hematological malignancies, promoting angiogenesis and tumor growth. A correlation between mast cells infiltration, angiogenesis and tumor progression has been reported for pancreatic ductal adenocarcinoma as well. Mast cells contribute to the aggressiveness of the pancreatic ductal carcinoma enhancing the expression of several pro-angiogenic factors such as vascular endothelial growth factor, fibroblast growth factor-2, platelet-derived growth factor and angiopoietin-1 as well as stimulating the pancreatic cancer cells proliferation by IL-13 and tryptase. The disruption of this pro-angiogenic and proliferative stimulation by inhibiting the mast cells migration and degranulation is under investigation as a potential therapeutic approach in pancreatic ductal adenocarcinoma patients. This review will summarize the literature concerning the mast cells infiltration in the pancreatic ductal adenocarcinoma analyzing its role in angiogenesis and tumor progression.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Pancreatic Ductal/genetics , Gene Expression Regulation, Neoplastic , Mast Cells/drug effects , Neovascularization, Pathologic/prevention & control , Pancreatic Neoplasms/genetics , Angiopoietin-1/antagonists & inhibitors , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Carcinoma, Pancreatic Ductal/blood supply , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Cell Movement/drug effects , Disease Progression , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Interleukin-13/antagonists & inhibitors , Interleukin-13/genetics , Interleukin-13/metabolism , Mast Cells/metabolism , Mast Cells/pathology , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Tryptases/antagonists & inhibitors , Tryptases/genetics , Tryptases/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
12.
Arthritis Res Ther ; 19(1): 124, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28587618

ABSTRACT

BACKGROUND: Increasing evidences indicate that an unbalance between tryptases and their endogenous inhibitors, leading to an increased proteolytic activity, is implicated in the pathophysiology of rheumatoid arthritis. The aim of the present study was to evaluate the impact of tryptase inhibition on experimental arthritis. METHODS: Analysis of gene expression and regulation in the mouse knee joint was performed by RT-qPCR and in situ hybridization. Arthritis was induced in male C57BL/6 mice with mBSA/IL-1ß. Tryptase was inhibited by two approaches: a lentivirus-mediated heterologous expression of the human endogenous tryptase inhibitor, sperm-associated antigen 11B isoform C (hSPAG11B/C), or a chronic treatment with the synthetic tryptase inhibitor APC366. Several inflammatory parameters were evaluated, such as oedema formation, histopathology, production of IL-1ß, -6, -17A and CXCL1/KC, myeloperoxidase and tryptase-like activities. RESULTS: Spag11c was constitutively expressed in chondrocytes and cells from the synovial membrane in mice, but its expression did not change 7 days after the induction of arthritis, while tryptase expression and activity were upregulated. The intra-articular transduction of animals with the lentivirus phSPAG11B/C or the treatment with APC366 inhibited the increase of tryptase-like activity, the late phase of oedema formation, the production of IL-6 and CXCL1/KC. In contrast, neutrophil infiltration, degeneration of hyaline cartilage and erosion of subchondral bone were not affected. CONCLUSIONS: Tryptase inhibition was effective in inhibiting some inflammatory parameters associated to mBSA/IL-1ß-induced arthritis, notably late phase oedema formation and IL-6 production, but not neutrophil infiltration and joint degeneration. These results suggest that the therapeutic application of tryptase inhibitors to rheumatoid arthritis would be restrained to palliative care, but not as disease-modifying drugs. Finally, this study highlighted lentivirus-based gene delivery as an instrumental tool to study the relevance of target genes in synovial joint physiology and disease.


Subject(s)
Gene Transfer Techniques , Inflammation/metabolism , Knee Joint/metabolism , Tryptases/metabolism , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Arthritis, Experimental/therapy , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/therapy , Chondrocytes/metabolism , Cytokines/metabolism , Dipeptides/pharmacology , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/therapy , Knee Joint/drug effects , Knee Joint/pathology , Lentivirus/genetics , Male , Mice, Inbred C57BL , Synovial Membrane/metabolism , Tryptases/antagonists & inhibitors , Tryptases/genetics
13.
Expert Opin Ther Pat ; 27(8): 919-928, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28425830

ABSTRACT

INTRODUCTION: Tryptase is one of the main serine-proteinases located in the secretory granules of mast cells, and is released through degranulation, which is involved in the pathogenesis of allergic inflammatory disease, cardiovascular diseases, lung fibrosis and tumor. Therefore, inhibitors targeting tryptase may represent a new direction for the treatment of allergic inflammatory disease and other diseases. Areas covered: In this article, we discussed the history and development of tryptase inhibitors and described a variety of tryptase inhibitors via their structures and biological importance in clinical studies and drug development for tryptase-related diseases. Expert opinion: Initial tryptase inhibitors based on indole structure as the hydrophobic substituent on a benzylamine-piperidine template have low specificity and poor bioavailability. Therefore, designing new and specific inhibitors targeting tryptase should be involved in future clinical studies. Modifications toward indoles with varying N-substitution, introducing an amide bond, and growing the chain length contribute to an increase in the specific selectivity and potency of tryptase inhibitors. Tryptase has become the research hotspot to explore many related diseases. Therefore, there has been growing appreciation for the potential importance of the tryptase inhibitors as a target for treating these diseases.


Subject(s)
Drug Design , Serine Proteinase Inhibitors/pharmacology , Tryptases/antagonists & inhibitors , Animals , Humans , Hypersensitivity/drug therapy , Hypersensitivity/enzymology , Inflammation/drug therapy , Inflammation/enzymology , Mast Cells/enzymology , Mast Cells/metabolism , Patents as Topic , Serine Proteinase Inhibitors/administration & dosage , Tryptases/metabolism
14.
Biosci Rep ; 37(2)2017 04 30.
Article in English | MEDLINE | ID: mdl-28356487

ABSTRACT

Frog skin secretions contain complex peptidomes and peptidic protease inhibitors that are one of the biologically and structurally described groups of components. In the present study, by use of molecular 'shotgun' cloning and LC MS/MS fractionation sequencing, a novel Bowman-Birk-type heptadecapeptide (AALKGCWTKSIPPKPCF-amide), named Odorrana schmackeriTrypsin Inhibitor (OSTI), with a canonical Cys6-Cys16 disulfide bridge, was isolated and identified in piebald odorous frog (O. schmackeri) skin secretion. A synthetic replicate of OSTI-exhibited trypsin inhibitory activity with a Ki value of 0.3 ± 0.04 nM and also a tryptase inhibitory effect with a Ki of 2.5 ± 0.6 µM. This is the first time that this property has been reported for a peptide originating from amphibian sources. In addition, substituting lysine (K) with phenylalanine (F) at the presumed P1 position, completely abrogated the trypsin and tryptase inhibition, but produced a strong chymotrypsin inhibition with a Ki of 1.0 ± 0.1 µM. Thus, the specificity of this peptidic protease inhibitor could be optimized through modifying the amino acid residue at the presumed P1 position and this novel native OSTI, along with its analogue, [Phe9]-OSTI, have expanded the potential drug discovery and development pipeline directed towards alleviation of serine protease-mediated pathologies.


Subject(s)
Oligopeptides/chemistry , Oligopeptides/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Ranidae/metabolism , Skin/metabolism , Amino Acid Sequence , Animals , Base Sequence , Chromatography, High Pressure Liquid , Cloning, Molecular , DNA, Complementary/genetics , Oligopeptides/genetics , Oligopeptides/isolation & purification , Protease Inhibitors/isolation & purification , Ranidae/genetics , Skin/chemistry , Trypsin , Trypsin Inhibitors/chemistry , Trypsin Inhibitors/isolation & purification , Trypsin Inhibitors/pharmacology , Tryptases/antagonists & inhibitors
15.
Immunol Lett ; 177: 16-21, 2016 09.
Article in English | MEDLINE | ID: mdl-27393495

ABSTRACT

Mast cells (MCs) are localized in connective tissues and are more numerous near the boundaries between the external environment and the internal milieu including the skin, the respiratory tract, the gastrointestinal tract and the conjunctiva. In the gastrointestinal tract, MCs represent 1-5% of mononuclear cells in the lamina propria of the mucosa and in the submucosa, and they are also found inside the epithelium and deep in the muscle and serosal layers. The gastrointestinal MCs perform their biological functions, releasing mediators, as amines (histamine, serotonin), cytokines, proteases, lipid mediators (leukotrienes, prostaglandins), and heparin. MCs are involved in the pathogenesis of different inflammatory conditions and tumors of the gastrointestinal tract. The use of MCs' tryptase inhibitors or c-KitR tyrosine kinase inhibitors could represent a potential anti-MC therapeutic approach in all the inflammatory and tumor pathological conditions of the digestive tube in which MCs are involved.


Subject(s)
Gastrointestinal Neoplasms/immunology , Inflammation/immunology , Intestinal Mucosa/immunology , Mast Cells/physiology , Anti-Inflammatory Agents/therapeutic use , Cytokines/metabolism , Enzyme Inhibitors/therapeutic use , Gastrointestinal Neoplasms/drug therapy , Heparin/metabolism , Histamine/metabolism , Humans , Inflammation/drug therapy , Leukotrienes/metabolism , Mast Cells/drug effects , Prostaglandins/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Serotonin/metabolism , Tryptases/antagonists & inhibitors
16.
Mediators Inflamm ; 2016: 6431574, 2016.
Article in English | MEDLINE | ID: mdl-27378825

ABSTRACT

Mast cells are primary effector cells of allergy, and recruitment of mast cells in involved tissue is one of the key events in allergic inflammation. Tryptase is the most abundant secretory product of mast cells, but little is known of its influence on mast cell accumulation. Using mouse peritoneal model, cell migration assay, and flow cytometry analysis, we investigated role of tryptase in recruiting mast cells. The results showed that tryptase induced up to 6.7-fold increase in mast cell numbers in mouse peritoneum following injection. Inhibitors of tryptase, an antagonist of PAR-2 FSLLRY-NH2, and pretreatment of mice with anti-ICAM-1, anti-CD11a, and anti-CD18 antibodies dramatically diminished tryptase induced mast cell accumulation. On the other hand, PAR-2 agonist peptides SLIGRL-NH2 and tc-LIGRLO-NH2 provoked mast cell accumulation following injection. These implicate that tryptase induced mast cell accumulation is dependent on its enzymatic activity, activation of PAR-2, and interaction between ICAM-1 and LFA-1. Moreover, induction of trans-endothelium migration of mast cells in vitro indicates that tryptase acts as a chemoattractant. In conclusion, provocation of mast cell accumulation by mast cell tryptase suggests a novel self-amplification mechanism of mast cell accumulation. Mast cell stabilizers as well as PAR-2 antagonist agents may be useful for treatment of allergic reactions.


Subject(s)
Intercellular Adhesion Molecule-1/metabolism , Mast Cells/metabolism , Receptor, PAR-2/metabolism , Tryptases/metabolism , Animals , CD18 Antigens/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , L-Selectin/metabolism , Mast Cells/drug effects , Mice , Mice, Inbred BALB C , Receptor, PAR-2/antagonists & inhibitors , Tryptases/antagonists & inhibitors
17.
PLoS One ; 10(10): e0141169, 2015.
Article in English | MEDLINE | ID: mdl-26485396

ABSTRACT

Tryptic serine proteases of bronchial epithelium regulate ion flux, barrier integrity, and allergic inflammation. Inhibition of some of these proteases is a strategy to improve mucociliary function in cystic fibrosis and asthmatic inflammation. Several inhibitors have been tested in pre-clinical animal models and humans. We hypothesized that these inhibitors inactivate a variety of airway protease targets, potentially with bystander effects. To establish relative potencies and modes of action, we compared inactivation of human prostasin, matriptase, airway trypsin-like protease (HAT), and ß-tryptase by nafamostat, camostat, bis(5-amidino-2-benzimidazolyl)methane (BABIM), aprotinin, and benzamidine. Nafamostat achieved complete, nearly stoichiometric and very slowly reversible inhibition of matriptase and tryptase, but inhibited prostasin less potently and was weakest versus HAT. The IC50 of nafamostat's leaving group, 6-amidino-2-naphthol, was >104-fold higher than that of nafamostat itself, consistent with suicide rather than product inhibition as mechanisms of prolonged inactivation. Stoichiometric release of 6-amidino-2-naphthol allowed highly sensitive fluorometric estimation of active-site concentration in preparations of matriptase and tryptase. Camostat inactivated all enzymes but was less potent overall and weakest towards matriptase, which, however was strongly inhibited by BABIM. Aprotinin exhibited nearly stoichiometric inhibition of prostasin and matriptase, but was much weaker towards HAT and was completely ineffective versus tryptase. Benzamidine was universally weak. Thus, each inhibitor profile was distinct. Nafamostat, camostat and aprotinin markedly reduced tryptic activity on the apical surface of cystic fibrosis airway epithelial monolayers, suggesting prostasin as the major source of such activity and supporting strategies targeting prostasin for inactivation.


Subject(s)
Bronchi/drug effects , Epithelial Cells/drug effects , Serine Endopeptidases/chemistry , Serine Proteinase Inhibitors/pharmacology , Tryptases/antagonists & inhibitors , Aprotinin/pharmacology , Bronchi/cytology , Bronchi/enzymology , Catalytic Domain , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/enzymology , Esters , Gabexate/analogs & derivatives , Gabexate/pharmacology , Guanidines , Humans
18.
Bioorg Med Chem Lett ; 25(17): 3676-80, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26122211

ABSTRACT

A novel series of terminal and internal phosphonate esters based on our previously developed aryl carboxylate-type tryptase selective inhibitor 1 was synthesized. The potency of these synthesized compounds was assessed in vitro with an enzyme inhibition assay using three available serine proteases, that is, tryptase, trypsin, and thrombin. The internal phosphonate derivative 6 showed potent thrombin inhibitory activity with an IC50 value of 1.0 µM, whereas it exhibited no or only weak tryptase and trypsin inhibition at 10 µM. The Lineweaver-Burk plot analysis indicates that the inhibition pattern of thrombin with 6 is non-competitive in spite of the fact that the lead carboxylate compound 1 is competitive inhibitor. Therefore, the skeletal conversion of the carboxylate into a phosphonate alters the mode of molecular recognition of these inhibitors by thrombin.


Subject(s)
Antithrombins/chemistry , Antithrombins/pharmacology , Anticoagulants/chemistry , Anticoagulants/pharmacology , Carboxylic Acids/chemistry , Chemistry Techniques, Synthetic , Drug Design , Drug Discovery , Drug Evaluation, Preclinical/methods , Inhibitory Concentration 50 , Organophosphonates/chemistry , Structure-Activity Relationship , Tryptases/antagonists & inhibitors
19.
Exp Cell Res ; 332(2): 157-62, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25478999

ABSTRACT

Human mast cells (MCs) are a rich reservoir of neutral proteases, packed in large amounts in their granules and comprising a high fraction of all cellular proteins. Among these proteases, tryptase is involved in angiogenesis after its release from activated MC granules, as it has been demonstrated in different in vitro and in vivo assays. Moreover, tryptase-positive MCs increase in number and vascularization increases in a linear fashion in different solid and hematological tumors. This complex interplay between MCs and tumor angiogenesis have led to consider the therapeutic use of angiogenesis inhibitors, which specifically target the angiogenic activity of tryptase, such as gabexate mesilate and nafamostat mesilate, two inhibitors of trypsin-like serine proteases.


Subject(s)
Cytoplasmic Granules/enzymology , Mast Cells/enzymology , Tryptases/physiology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Angiogenic Proteins/physiology , Animals , Humans , Molecular Targeted Therapy , Neoplasms/blood supply , Neoplasms/drug therapy , Neoplasms/enzymology , Neovascularization, Pathologic , Tryptases/antagonists & inhibitors
20.
Biomed Res Int ; 2014: 154702, 2014.
Article in English | MEDLINE | ID: mdl-25295247

ABSTRACT

Angiogenesis is a complex process finely regulated by the balance between angiogenesis stimulators and inhibitors. As a result of proangiogenic factors overexpression, it plays a crucial role in cancer development. Although initially mast cells (MCs) role has been defined in hypersensitivity reactions and in immunity, it has been discovered that MCs have a crucial interplay on the regulatory function between inflammatory and tumor cells through the release of classical proangiogenic factors (e.g., vascular endothelial growth factor) and nonclassical proangiogenic mediators granule-associated (mainly tryptase). In fact, in several animal and human malignancies, MCs density is highly correlated with tumor angiogenesis. In particular, tryptase, an agonist of the proteinase-activated receptor-2 (PAR-2), represents one of the most powerful angiogenic mediators released by human MCs after c-Kit receptor activation. This protease, acting on PAR-2 by its proteolytic activity, has angiogenic activity stimulating both human vascular endothelial and tumor cell proliferation in paracrine manner, helping tumor cell invasion and metastasis. Based on literature data it is shown that tryptase may represent a promising target in cancer treatment due to its proangiogenic activity. Here we focused on molecular mechanisms of three tryptase inhibitors (gabexate mesylate, nafamostat mesylate, and tranilast) in order to consider their prospective role in cancer therapy.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Mast Cells/drug effects , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Tryptases/genetics , Cell Proliferation/drug effects , Gabexate/administration & dosage , Humans , Inflammation/drug therapy , Inflammation/pathology , Mast Cells/pathology , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Oligopeptides/metabolism , Tryptases/antagonists & inhibitors , Tumor Microenvironment/drug effects , Vascular Endothelial Growth Factor A
SELECTION OF CITATIONS
SEARCH DETAIL
...