Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
J Allergy Clin Immunol ; 150(6): 1534-1544, 2022 12.
Article in English | MEDLINE | ID: mdl-35779668

ABSTRACT

BACKGROUND: Mast cells are implicated in the pathogenesis of asthma, but the underlying mechanisms are not fully elucidated. Under asthmatic conditions, mast cells can relocalize to the epithelial layer and may thereby affect the functional properties of the airway epithelial cells. OBJECTIVES: Activated mast cells release large quantities of proteases from their secretory granules, including chymase and tryptase. Here we investigated whether these proteases may affect airway epithelial cells. METHODS: Primary small airway epithelial cells were treated with tryptase or chymase, and the effects on epithelial cell viability, proliferation, migration, cytokine output, and transcriptome were evaluated. RESULTS: Airway epithelial cells were relatively refractory to tryptase. In contrast, chymase had extensive effects on multiple features of the epithelial cells, with a particular emphasis on processes related to extracellular matrix (ECM) remodeling. These included suppressed expression of ECM-related genes such as matrix metalloproteinases, which was confirmed at the protein level. Further, chymase suppressed the expression of the fibronectin gene and also caused degradation of fibronectin released by the epithelial cells. Chymase was also shown to suppress the migratory capacity of the airway epithelial cells and to degrade the cell-cell contact protein E-cadherin on the epithelial cell surface. CONCLUSION: Our findings suggest that chymase may affect the regulation of ECM remodeling events mediated by airway epithelial cells, with implications for the impact of mast cells in inflammatory lung diseases such as asthma.


Subject(s)
Extracellular Matrix , Mast Cells , Humans , Epithelial Cells
2.
J Allergy Clin Immunol ; 149(2): 718-727, 2022 02.
Article in English | MEDLINE | ID: mdl-34331992

ABSTRACT

BACKGROUND: Mast cells (MCs) have a profound impact on allergic asthma. Under such conditions, MCs undergo degranulation, resulting in the release of exceptionally large amounts of MC-restricted proteases. However, the role of these proteases in asthma is only partially understood. OBJECTIVES: We sought to test our hypothesis that MC proteases can influence the functionality of human lung fibroblasts (HLFs). METHODS: Primary HLFs were treated with MC chymase or tryptase, followed by assessment of parameters related to fibroblast function. RESULTS: HLFs underwent major morphologic changes in response to chymase, showing signs of cellular contraction, but were refractory to tryptase. However, no effects of chymase on HLF viability or proliferation were seen. Chymase, but not tryptase, had a major impact on the output of extracellular matrix-associated compounds from the HLFs, including degradation of fibronectin and collagen-1, and activation of pro-matrix metalloprotease 2. Further, chymase induced the release of various chemotactic factors from HLFs. In line with this, conditioned medium from chymase-treated HLFs showed chemotactic activity on neutrophils. Transcriptome analysis revealed that chymase induced a proinflammatory gene transcription profile in HLFs, whereas tryptase had minimal effects. CONCLUSIONS: Chymase, but not tryptase, has a major impact on the phenotype of primary airway fibroblasts by modifying their output of extracellular matrix components and by inducing a proinflammatory phenotype.


Subject(s)
Asthma/etiology , Chymases/toxicity , Fibroblasts/drug effects , Lung/drug effects , Mast Cells/enzymology , Apoptosis/drug effects , Cells, Cultured , Collagen Type I/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lung/metabolism , Lung/pathology , Mast Cells/physiology , Transcriptome , Tryptases/toxicity
3.
J Allergy Clin Immunol ; 144(4S): S31-S45, 2019 10.
Article in English | MEDLINE | ID: mdl-30772496

ABSTRACT

Mast cells (MCs), which are best known for their detrimental role in patients with allergic diseases, act in a diverse array of physiologic and pathologic functions made possible by the plurality of MC types. Their various developmental avenues and distinct sensitivity to (micro-) environmental conditions convey extensive heterogeneity, resulting in diverse functions. We briefly summarize this heterogeneity, elaborate on molecular determinants that allow MCs to communicate with their environment to fulfill their tasks, discuss the protease repertoire stored in secretory lysosomes, and consider different aspects of MC signaling. Furthermore, we describe key MC governance mechanisms (ie, the high-affinity receptor for IgE [FcεRI]), the stem cell factor receptor KIT, the IL-4 system, and both Ca2+- and phosphatase-dependent mechanisms. Finally, we focus on distinct physiologic functions, such as chemotaxis, phagocytosis, host defense, and the regulation of MC functions at the mucosal barriers of the lung, gastrointestinal tract, and skin. A deeper knowledge of the pleiotropic functions of MC mediators, as well as the molecular processes of MC regulation and communication, should enable us to promote beneficial MC traits in physiology and suppress detrimental MC functions in patients with disease.


Subject(s)
Chemotaxis/immunology , Intestinal Mucosa/immunology , Mast Cells/immunology , Phagocytosis , Respiratory Mucosa/immunology , Signal Transduction/immunology , Animals , Calcium/immunology , Humans , Interleukin-4/immunology , Intestinal Mucosa/pathology , Lysosomes/immunology , Lysosomes/pathology , Mast Cells/pathology , Proto-Oncogene Proteins c-kit/immunology , Receptors, IgE/immunology , Respiratory Mucosa/pathology
4.
Int J Mol Sci ; 21(14)2020 Jul 17.
Article in English | MEDLINE | ID: mdl-32709152

ABSTRACT

Tryptase is a tetrameric serine protease located within the secretory granules of mast cells. In the secretory granules, tryptase is stored in complex with negatively charged heparin proteoglycans and it is known that heparin is essential for stabilizing the enzymatic activity of tryptase. However, recent findings suggest that enzymatically active tryptase also can be found in the nucleus of murine mast cells, but it is not known how the enzmatic activity of tryptase is maintained in the nuclear milieu. Here we hypothesized that tryptase, as well as being stabilized by heparin, can be stabilized by DNA, the rationale being that the anionic charge of DNA could potentially substitute for that of heparin to execute this function. Indeed, we showed that double-stranded DNA preserved the enzymatic activity of human ß-tryptase with a similar efficiency as heparin. In contrast, single-stranded DNA did not have this capacity. We also demonstrated that DNA fragments down to 400 base pairs have tryptase-stabilizing effects equal to that of intact DNA. Further, we showed that DNA-stabilized tryptase was more efficient in degrading nuclear core histones than heparin-stabilized enzyme. Finally, we demonstrated that tryptase, similar to its nuclear localization in murine mast cells, is found within the nucleus of primary human skin mast cells. Altogether, these finding reveal a hitherto unknown mechanism for the stabilization of mast cell tryptase, and these findings can have an important impact on our understanding of how tryptase regulates nuclear events.


Subject(s)
DNA/chemistry , Mast Cells/enzymology , Tryptases/chemistry , Cells, Cultured , Enzyme Stability , Humans , Mast Cells/chemistry , Mast Cells/cytology , Skin/chemistry , Skin/cytology , Skin/enzymology
6.
J Biol Chem ; 288(19): 13885-96, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23548907

ABSTRACT

BACKGROUND: Sunflower trypsin inhibitor-1 (SFTI-1) and Momordica cochinchinensis trypsin inhibitor-II (MCoTI-II) are potent protease inhibitors comprising a cyclic backbone. RESULTS: Elucidation of structure-activity relationships for SFTI-1 and MCoTI-II was used to design inhibitors with enhanced inhibitory activity. CONCLUSION: An analog of MCoTI-II is one of the most potent inhibitors of matriptase. SIGNIFICANCE: These results provide a solid basis for the design of selective peptide inhibitors of matriptase with therapeutic potential. The type II transmembrane serine protease matriptase is a key activator of multiple signaling pathways associated with cell proliferation and modification of the extracellular matrix. Deregulated matriptase activity correlates with a number of diseases, including cancer and hence highly selective matriptase inhibitors may have therapeutic potential. The plant-derived cyclic peptide, sunflower trypsin inhibitor-1 (SFTI-1), is a promising drug scaffold with potent matriptase inhibitory activity. In the current study we have analyzed the structure-activity relationships of SFTI-1 and Momordica cochinchinensis trypsin inhibitor-II (MCoTI-II), a structurally divergent trypsin inhibitor from Momordica cochinchinensis that also contains a cyclic backbone. We show that MCoTI-II is a significantly more potent matriptase inhibitor than SFTI-1 and that all alanine mutants of both peptides, generated using positional scanning mutagenesis, have decreased trypsin affinity, whereas several mutations either maintain or result in enhanced matriptase inhibitory activity. These intriguing results were used to design one of the most potent matriptase inhibitors known to date with a 290 pm equilibrium dissociation constant, and provide the first indication on how to modulate affinity for matriptase over trypsin in cyclic peptides. This information might be useful for the design of more selective and therapeutically relevant inhibitors of matriptase.


Subject(s)
Peptides, Cyclic/chemistry , Plant Proteins/chemistry , Serine Endopeptidases/chemistry , Serine Proteinase Inhibitors/chemistry , Amino Acid Sequence , Amino Acid Substitution , Catalytic Domain , Helianthus/chemistry , Humans , Hydrogen Bonding , Kinetics , Molecular Dynamics Simulation , Molecular Sequence Data , Momordica/chemistry , Nuclear Magnetic Resonance, Biomolecular , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/genetics , Plant Proteins/chemical synthesis , Plant Proteins/genetics , Protein Binding , Structure-Activity Relationship , Surface Properties
7.
ChemMedChem ; 18(18): e202300218, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37424408

ABSTRACT

The zymogens of the neutrophil serine proteases elastase, proteinase 3, and cathepsin G are converted proteolytically into their pro-inflammatory active forms by the action of cathepsin C. The inhibition of this cysteine protease therefore is an interesting therapeutic approach for the treatment of inflammatory disorders with a high neutrophil burden such as COPD. Based on E-64c-hydrazide as lead structure, we have recently developed a covalently acting cathepsin C inhibitor using a n-butyl residue attached at the amine nitrogen of the hydrazide moiety to efficiently address the deep hydrophobic S2 pocket. To further optimize the affinity and selectivity profile of this inhibitor, the S1'-S2' area was now investigated by a combinatorial approach, showing that Nle-tryptamide is a ligand superior to the initially used Leu-isoamylamide. Using the neutrophil precursor line U937 as a cell culture model, this optimized inhibitor blocks the intracellular cathepsin C activity and thereby suppresses the activation of neutrophil elastase.


Subject(s)
Cathepsin C , Hydrazines , Cathepsin C/metabolism , Hydrazines/pharmacology , Leukocyte Elastase/metabolism , Serine Proteases , Leucine
8.
Nat Commun ; 14(1): 1910, 2023 04 06.
Article in English | MEDLINE | ID: mdl-37024468

ABSTRACT

PRG4 is an extracellular matrix protein that maintains homeostasis through its boundary lubricating and anti-inflammatory properties. Altered expression and function of PRG4 have been associated with joint inflammatory diseases, including osteoarthritis. Here we show that mast cell tryptase ß cleaves PRG4 in a dose- and time-dependent manner, which was confirmed by silver stain gel electrophoresis and mass spectrometry. Tryptase-treated PRG4 results in a reduction of lubrication. Compared to full-length, cleaved PRG4 further activates NF-κB expression in cells overexpressing TLR2, -4, and -5. In the destabilization of the medial meniscus model of osteoarthritis in rat, tryptase ß and PRG4 colocalize at the site of injury in knee cartilage and is associated with disease severity. When human primary synovial fibroblasts from male osteoarthritis patients or male healthy subjects treated with tryptase ß and/or PRG4 are subjected to a quantitative shotgun proteomics and proteome changes are characterized, it further supports the role of NF-κB activation. Here we show that tryptase ß as a modulator of joint lubrication in osteoarthritis via the cleavage of PRG4.


Subject(s)
Cartilage, Articular , Osteoarthritis , Humans , Male , Animals , Rats , Tryptases/metabolism , Proteoglycans/metabolism , Lubrication , NF-kappa B/metabolism , Osteoarthritis/metabolism , Inflammation/metabolism , Cartilage, Articular/metabolism
9.
Carcinogenesis ; 33(12): 2507-19, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23042094

ABSTRACT

In America and Western Europe, prostate cancer is the second leading cause of death in men. Emerging evidence suggests that chronic inflammation is a major risk factor for the development and metastatic progression of prostate cancer. We previously reported that the chemopreventive polyphenol curcumin inhibits the expression of the proinflammatory cytokines CXCL1 and -2 leading to diminished formation of breast cancer metastases. In this study, we analyze the effects of curcumin on prostate carcinoma growth, apoptosis and metastasis. We show that curcumin inhibits translocation of NFκB to the nucleus through the inhibition of the IκB-kinase (IKKß, leading to stabilization of the inhibitor of NFκB, IκBα, in PC-3 prostate carcinoma cells. Inhibition of NFκB activity reduces expression of CXCL1 and -2 and abolishes the autocrine/paracrine loop that links the two chemokines to NFκB. The combination of curcumin with the synthetic IKKß inhibitor, SC-541, shows no additive or synergistic effects indicating that the two compounds share the target. Treatment of the cells with curcumin and siRNA-based knockdown of CXCL1 and -2 induce apoptosis, inhibit proliferation and downregulate several important metastasis-promoting factors like COX2, SPARC and EFEMP. In an orthotopic mouse model of hematogenous metastasis, treatment with curcumin inhibits statistically significantly formation of lung metastases. In conclusion, chronic inflammation can induce a metastasis prone phenotype in prostate cancer cells by maintaining a positive proinflammatory and prometastatic feedback loop between NFκB and CXCL1/-2. Curcumin disrupts this feedback loop by the inhibition of NFκB signaling leading to reduced metastasis formation in vivo.


Subject(s)
Antineoplastic Agents/therapeutic use , Chemokine CXCL1/antagonists & inhibitors , Chemokine CXCL2/antagonists & inhibitors , Curcumin/therapeutic use , Prostatic Neoplasms/drug therapy , Animals , Apoptosis , Cell Line, Tumor , Chemokine CXCL1/genetics , Chemokine CXCL2/genetics , Humans , Male , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Neoplasm Metastasis , Prostatic Neoplasms/pathology , Signal Transduction , Xenograft Model Antitumor Assays
10.
J Innate Immun ; 14(5): 433-446, 2022.
Article in English | MEDLINE | ID: mdl-34937018

ABSTRACT

Previous research has indicated an intimate functional communication between mast cells (MCs) and neutrophils during inflammatory conditions, but the nature of such communication is not fully understood. Activated neutrophils are known to release DNA-containing extracellular traps (neutrophil extracellular traps [NETs]) and, based on the known ability of tryptase to interact with negatively charged polymers, we here hypothesized that tryptase might interact with NET-contained DNA and thereby regulate NET formation. In support of this, we showed that tryptase markedly enhances NET formation in phorbol myristate acetate-activated human neutrophils. Moreover, tryptase was found to bind vividly to the NETs, to cause proteolysis of core histones and to cause a reduction in the levels of citrullinated histone-3. Secretome analysis revealed that tryptase caused increased release of numerous neutrophil granule compounds, including gelatinase, lactoferrin, and myeloperoxidase. We also show that DNA can induce the tetrameric, active organization of tryptase, suggesting that NET-contained DNA can maintain tryptase activity in the extracellular milieu. In line with such a scenario, DNA-stabilized tryptase was shown to efficiently degrade numerous pro-inflammatory compounds. Finally, we showed that tryptase is associated with NET formation in vivo in a melanoma setting and that NET formation in vivo is attenuated in mice lacking tryptase expression. Altogether, these findings reveal that NET formation can be regulated by MC tryptase, thus introducing a novel mechanism of communication between MCs and neutrophils.


Subject(s)
Extracellular Traps , Animals , DNA/metabolism , Extracellular Traps/metabolism , Histones/metabolism , Humans , Mice , Neutrophils/metabolism , Tryptases/metabolism
11.
J Biol Chem ; 285(40): 30686-97, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20667834

ABSTRACT

Protein misfolding with loss-of-function of the enzyme phenylalanine hydroxylase (PAH) is the molecular basis of phenylketonuria in many individuals carrying missense mutations in the PAH gene. PAH is complexly regulated by its substrate L-Phenylalanine and its natural cofactor 6R-L-erythro-5,6,7,8-tetrahydrobiopterin (BH(4)). Sapropterin dihydrochloride, the synthetic form of BH(4), was recently approved as the first pharmacological chaperone to correct the loss-of-function phenotype. However, current knowledge about enzyme function and regulation in the therapeutic setting is scarce. This illustrates the need for comprehensive analyses of steady state kinetics and allostery beyond single residual enzyme activity determinations to retrace the structural impact of missense mutations on the phenylalanine hydroxylating system. Current standard PAH activity assays are either indirect (NADH) or discontinuous due to substrate and product separation before detection. We developed an automated fluorescence-based continuous real-time PAH activity assay that proved to be faster and more efficient but as precise and accurate as standard methods. Wild-type PAH kinetic analyses using the new assay revealed cooperativity of activated PAH toward BH(4), a previously unknown finding. Analyses of structurally preactivated variants substantiated BH(4)-dependent cooperativity of the activated enzyme that does not rely on the presence of l-Phenylalanine but is determined by activating conformational rearrangements. These findings may have implications for an individualized therapy, as they support the hypothesis that the patient's metabolic state has a more significant effect on the interplay of the drug and the conformation and function of the target protein than currently appreciated.


Subject(s)
Biopterins/analogs & derivatives , Coenzymes/chemistry , Phenylalanine Hydroxylase/chemistry , Phenylalanine/chemistry , Allosteric Regulation/genetics , Biopterins/chemistry , Biopterins/metabolism , Biopterins/therapeutic use , Coenzymes/metabolism , Coenzymes/therapeutic use , Enzyme Activation/genetics , Fluorescence , Humans , Kinetics , Mutation, Missense , Phenylalanine/genetics , Phenylalanine/metabolism , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Phenylketonurias/drug therapy , Phenylketonurias/enzymology , Phenylketonurias/genetics
12.
Hum Mol Genet ; 18(9): 1612-23, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19224950

ABSTRACT

Newborn screening (NBS) for medium-chain acyl-CoA dehydrogenase deficiency (MCADD) revealed a higher birth prevalence and genotypic variability than previously estimated, including numerous novel missense mutations in the ACADM gene. On average, these mutations are associated with milder biochemical phenotypes raising the question about their pathogenic relevance. In this study, we analyzed the impact of 10 ACADM mutations identified in NBS (A27V, Y42H, Y133H, R181C, R223G, D241G, K304E, R309K, I331T and R388S) on conformation, stability and enzyme kinetics of the corresponding proteins. Partial to total rescue of aggregation by co-overexpression of GroESL indicated protein misfolding. This was confirmed by accelerated thermal unfolding in all variants, as well as decreased proteolytic stability and accelerated thermal inactivation in most variants. Catalytic function varied from high residual activity to markedly decreased activity or substrate affinity. Mutations mapping to the beta-domain of the protein predisposed to severe destabilization. In silico structural analyses of the affected amino acid residues revealed involvement in functionally relevant networks. Taken together, our results substantiate the hypothesis of protein misfolding with loss-of-function being the common molecular basis in MCADD. Moreover, considerable structural alterations in all analyzed variants do not support the view that novel mutations found in NBS bear a lower risk of metabolic decompensation than that associated with mutations detected in clinically ascertained patients. Finally, the detailed insight into how ACADM missense mutations induce loss of MCAD function may provide guidance for risk assessment and counseling of patients, and in future may assist delineation of novel pharmacological strategies.


Subject(s)
Acyl-CoA Dehydrogenase/chemistry , Acyl-CoA Dehydrogenase/deficiency , Lipid Metabolism, Inborn Errors/enzymology , Neonatal Screening , Protein Folding , Acyl-CoA Dehydrogenase/genetics , Amino Acid Substitution , Enzyme Stability , Female , Humans , Infant, Newborn , Kinetics , Lipid Metabolism, Inborn Errors/genetics , Male , Molecular Conformation , Molecular Sequence Data , Mutation, Missense
13.
Am J Hum Genet ; 83(1): 5-17, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18538294

ABSTRACT

A significant share of patients with phenylalanine hydroxylase (PAH) deficiency benefits from pharmacological doses of tetrahydrobiopterin (BH(4)), the natural PAH cofactor. Phenylketonuria (PKU) is hypothesized to be a conformational disease, with loss of function due to protein destabilization, and the restoration of enzyme function that is observed in BH(4) treatment might be transmitted by correction of protein misfolding. To elucidate the molecular basis of functional impairment in PAH deficiency, we investigated the impact of ten PAH gene mutations identified in patients with BH(4)-responsiveness on enzyme kinetics, stability, and conformation of the protein (F55L, I65S, H170Q, P275L, A300S, S310Y, P314S, R408W, Y414C, Y417H). Residual enzyme activity was generally high, but allostery was disturbed in almost all cases and pointed to altered protein conformation. This was confirmed by reduced proteolytic stability, impaired tetramer assembly or aggregation, increased hydrophobicity, and accelerated thermal unfolding--with particular impact on the regulatory domain--observed in most variants. Three-dimensional modeling revealed the involvement of functionally relevant amino acid networks that may communicate misfolding throughout the protein. Our results substantiate the view that PAH deficiency is a protein-misfolding disease in which global conformational changes hinder molecular motions essential for physiological enzyme function. Thus, PKU has evolved from a model of a genetic disease that leads to severe neurological impairment to a model of a treatable protein-folding disease with loss of function.


Subject(s)
Motion , Phenylalanine Hydroxylase/deficiency , Phenylalanine Hydroxylase/metabolism , Phenylketonurias/enzymology , Phenylketonurias/genetics , Administration, Oral , Allosteric Regulation , Amino Acid Metabolism, Inborn Errors , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Biopterins/administration & dosage , Biopterins/analogs & derivatives , Biopterins/therapeutic use , Catalytic Domain , Computer Simulation , Dimerization , Endopeptidase K/pharmacology , Enzyme Stability , Female , Hot Temperature , Humans , Hydrogen Bonding , Hydrolysis , Hydrophobic and Hydrophilic Interactions , Infant, Newborn , Kinetics , Luminescence , Male , Models, Molecular , Mutation, Missense , Phenylalanine/blood , Phenylalanine/metabolism , Phenylalanine Hydroxylase/analysis , Phenylalanine Hydroxylase/chemistry , Phenylalanine Hydroxylase/genetics , Protein Conformation , Protein Denaturation , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary/genetics , Protein Subunits/chemistry , Protein Subunits/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Static Electricity
14.
Bioorg Med Chem ; 19(23): 7236-43, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22047802

ABSTRACT

ß-Tryptase, a mast-cell specific serine protease with trypsin-like activity, has emerged in the last years as a promising novel therapeutic target in the field of allergic inflammation. Recently, we have developed a potent and selective ß-tryptase inhibitor based on the natural product cyclotheonamide E4 by implementing a basic P3 residue that addresses the determinants of the extended substrate specificity of ß-tryptase. To further improve the affinity/selectivity profile of this lead structure, we have now investigated ß-homo-3-aminomethylphenylalanine as S1 ligand. In contrast to the corresponding ß-homo amino acids derived from lysine or arginine, we demonstrate that this particular basic ß-homo amino acid is a privileged S1 ligand for the development of ß-tryptase inhibitors. Besides affinity, selectivity and reduced basicity, these novel cyclotheonamide E4 analogs show excellent stability in human plasma and serum.


Subject(s)
Arginine/chemistry , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Phenylalanine/analogs & derivatives , Serine Proteinase Inhibitors/chemistry , Tryptases/antagonists & inhibitors , Biomimetic Materials/chemistry , Drug Stability , Humans , Ligands , Peptides, Cyclic/blood , Peptides, Cyclic/chemical synthesis , Phenylalanine/chemistry , Serine Proteinase Inhibitors/blood , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/pharmacology , Structure-Activity Relationship , Tryptases/metabolism
15.
J Immunol ; 183(4): 2223-31, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19625657

ABSTRACT

The cathelicidin LL-37 represents a potent antimicrobial and cell-stimulating agent, most abundantly expressed in peripheral organs such as lung and skin during inflammation. Because mast cells (MC) overtake prominent immunomodulatory roles in these organs, we wondered whether interactions exist between MC and LL-37. In this study, we show for the first time to our knowledge that physiological concentrations of LL-37 induce degranulation in purified human lung MC. Intriguingly, as a consequence LL-37 rapidly undergoes limited cleavage by a released protease. The enzyme was identified as beta-tryptase by inhibitor studies and by comparison to the recombinant protease. Examining the resulting LL-37 fragments for their functional activity, we found that none of the typical capacities of intact LL-37, i.e., MC degranulation, bactericidal activity, and neutralization of LPS, were retained. Conversely, we found that another inflammatory protein, the platelet-derived chemokine CXCL4, protects LL-37 from cleavage by beta-tryptase. Interestingly, CXCL4 did not act as a direct enzyme inhibitor, but destabilized active tetrameric beta-tryptase by antagonizing the heparin component required for the integrity of the tetramer. Altogether our results suggest that interaction of LL-37 and MC initiates an effective feedback loop to limit cathelicidin activity during inflammation, whereas CXCL4 may represent a physiological counter-regulator of beta-tryptase activity.


Subject(s)
Cathelicidins/metabolism , Mast Cells/enzymology , Mast Cells/immunology , Platelet Factor 4/physiology , Tryptases/physiology , Antimicrobial Cationic Peptides , Cathelicidins/antagonists & inhibitors , Cathelicidins/physiology , Cell Degranulation/immunology , Cells, Cultured , Feedback, Physiological/immunology , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Lung/enzymology , Lung/immunology , Lung/metabolism , Mast Cells/metabolism , Protein Processing, Post-Translational/immunology , Tryptases/metabolism
16.
Biochem J ; 428(3): 473-82, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20337595

ABSTRACT

Pathogenic bacteria, including Pseudomonas aeruginosa, interact with and engage the host plasminogen (Plg) activation system, which encompasses the urokinase (uPA)-type Plg activator, and is involved in extracellular proteolysis, including matrilysis and fibrinolysis. We hypothesized that secreted bacterial proteases might contribute to the activation of this major extracellular proteolytic system, thereby participating in bacterial dissemination. We report that LasB, a thermolysin-like metalloprotease secreted by Ps. aeruginosa, converts the human uPA zymogen into its active form (kcat=4.9 s-1, Km=8.9 microM). Accordingly, whereas the extracellular secretome from the LasB-expressing pseudomonal strain PAO1 efficiently activates pro-uPA, the secretome from the isogenic LasB-deficient strain PDO240 is markedly less potent in pro-uPA activation. Still, both secretomes induce some metalloprotease-independent activation of the human zymogen. The latter involves a serine protease, which we identified via both recombinant protein expression in Escherichia coli and purification from pseudomonal cultures as protease IV (PIV; kcat=0.73 s-1, Km=6.2 microM). In contrast, neither secretomes nor the pure proteases activate Plg. Along with this, LasB converts Plg into mini-Plg and angiostatin, whereas, as reported previously, it processes the uPA receptor, inactivates the plasminogen activator inhibitor 1, and activates pro-matrix metalloproteinase 2. PIV does not target these factors at all. To conclude, LasB and PIV, although belonging to different protease families and displaying quite different substrate specificities, both activate the urokinase-type precursor of the Plg activation cascade. Direct pro-uPA activation, as also reported for other bacterial proteases, might be a frequent phenomenon that contributes to bacterial virulence.


Subject(s)
Bacterial Proteins/metabolism , Peptide Hydrolases/metabolism , Pseudomonas aeruginosa/enzymology , Urokinase-Type Plasminogen Activator/metabolism , Humans , Kinetics , Plasminogen/metabolism , Pseudomonas aeruginosa/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Urokinase-Type Plasminogen Activator/genetics
17.
J Biol Chem ; 284(51): 35612-20, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19820233

ABSTRACT

Protein folding mechanisms have remained elusive mainly because of the transient nature of intermediates. Leech-derived tryptase inhibitor (LDTI) is a Kazal-type serine proteinase inhibitor that is emerging as an attractive model for folding studies. It comprises 46 amino acid residues with three disulfide bonds, with one located inside a small triple-stranded antiparallel beta-sheet and with two involved in a cystine-stabilized alpha-helix, a motif that is widely distributed in bioactive peptides. Here, we analyzed the oxidative folding and reductive unfolding of LDTI by chromatographic and disulfide analyses of acid-trapped intermediates. It folds and unfolds, respectively, via sequential oxidation and reduction of the cysteine residues that give rise to a few 1- and 2-disulfide intermediates. Species containing two native disulfide bonds predominate during LDTI folding (IIa and IIc) and unfolding (IIa and IIb). Stop/go folding experiments demonstrate that only intermediate IIa is productive and oxidizes directly into the native form. The NMR structures of acid-trapped and further isolated IIa, IIb, and IIc reveal global folds similar to that of the native protein, including a native-like canonical inhibitory loop. Enzyme kinetics shows that both IIa and IIc are inhibitory-active, which may substantially reduce proteolysis of LDTI during its folding process. The results reported show that the kinetics of the folding reaction is modulated by the specific structural properties of the intermediates and together provide insights into the interdependence of conformational folding and the assembly of native disulfides during oxidative folding.


Subject(s)
Hirudo medicinalis/chemistry , Protein Folding , Proteins/chemistry , Amino Acid Motifs/physiology , Animals , Disulfides/chemistry , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction
19.
Biochem J ; 410(1): 157-65, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-17973626

ABSTRACT

The major opportunistic pathogen Staphylococcus aureus utilizes the human fibrinolytic system for invasion and spread via plasmin(ogen) binding and non-proteolytic activation. Because S. aureus secretes several proteases recently proposed as virulence factors, we explored whether these enzymes could add to the activation of the host's fibrinolytic system. Exposure of human pro-urokinase [pro-uPA (where uPA is urokinase-type plasminogen activator)] to conditioned growth media from staphylococcal reference strains results in an EDTA-sensitive conversion of the single-chain zymogen into its two-chain active form, an activity not observed in an aureolysin-deficient strain. Using purified aureolysin, we verified the capacity of this thermolysin-like metalloprotease to activate pro-uPA, with a 2.6 x 10(3) M(-1) x s(-1) catalytic efficiency. Moreover, activation also occurs in the presence of human plasma, as well as in conditioned growth media from clinical isolates. Finally, we establish that aureolysin (i) converts plasminogen into angiostatin and mini-plasminogen, the latter retaining its capacity to be activated by uPA and to hydrolyse fibrin, (ii) degrades the plasminogen activator inhibitor-1, and (iii) abrogates the inhibitory activity of alpha(2)-antiplasmin. Altogether, we propose that, in parallel with the staphylokinase-dependent activation of plasminogen, aureolysin may contribute significantly to the activation of the fibrinolytic system by S. aureus, and thus may promote bacterial spread and invasion.


Subject(s)
Bacterial Proteins/metabolism , Fibrinolysis , Metalloendopeptidases/metabolism , Metalloproteases/metabolism , Staphylococcus aureus/metabolism , Base Sequence , Culture Media, Conditioned , DNA Primers , Humans , Kinetics , Plasminogen Activator Inhibitor 1/metabolism , Polymerase Chain Reaction , Staphylococcus aureus/enzymology , Staphylococcus aureus/pathogenicity , Urokinase-Type Plasminogen Activator/metabolism , Virulence
20.
Cell Death Dis ; 10(9): 659, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31506436

ABSTRACT

It is well established that mast cell accumulation accompanies most malignancies. However, the knowledge of how mast cells functionally impact on tumors is still rudimentary. Here we addressed this issue and show that mast cells have anti-proliferative activity on melanoma cells and that this effect is dependent on tryptase, a tetrameric protease stored in mast cell granules. Mechanistically, tryptase was found to be endocytosed by melanoma cells as cargo of DNA-coated exosomes released from melanoma cells, followed by transport to the nucleus. In the nucleus, tryptase executed clipping of histone 3 and degradation of Lamin B1, accompanied by extensive nuclear remodeling. Moreover, tryptase degraded hnRNP A2/B1, a protein involved in mRNA stabilization and interaction with non-coding RNAs. This was followed by downregulated expression of the oncogene EGR1 and of multiple non-coding RNAs, including oncogenic species. Altogether, these findings establish a new principle for regulation of tumor cell proliferation.


Subject(s)
Cell Nucleus/enzymology , Cell Proliferation , Gene Expression Regulation, Neoplastic , Mast Cells/enzymology , Melanoma/enzymology , Neoplasm Proteins/metabolism , Tryptases/metabolism , Active Transport, Cell Nucleus/genetics , Animals , Cell Line, Tumor , Cell Nucleus/genetics , Exosomes , Mast Cells/pathology , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Tryptases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL