Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Pharmaceuticals (Basel) ; 11(2)2018 May 21.
Article in English | MEDLINE | ID: mdl-29883401

ABSTRACT

Matriptase-2 is a type II transmembrane serine protease and a key regulator of systemic iron homeostasis. Since the activation mechanism and several features of the physiological role of matriptase-2 are not fully understood, there is strong need for analytical tools to perform tasks such as distinguishing active and inactive matriptase-2. For this purpose we present a short biotinylated peptide derivative with a chloromethyl ketone group, biotin-RQRR-CMK, as an activity-based probe for matriptase-2. Biotin-RQRR-CMK was kinetically characterized and exhibited a second-order rate constant of inactivation (kinac/Ki) of 10,800 M−1 s−1 towards the matriptase-2 activity in the supernatant of transfected human embryonic kidney (HEK) cells. Biotin-RQRR-CMK was able to label active matriptase-2, as visualized in western blot experiments. Pretreatment with aprotinin, an active-site directed inhibitor of serine proteases, protected matriptase-2 from the reaction with biotin-RQRR-CMK.

2.
Cell Rep ; 21(8): 2090-2103, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29166602

ABSTRACT

The host metalloprotease meprin ß is required for mucin 2 (MUC2) cleavage, which drives intestinal mucus detachment and prevents bacterial overgrowth. To gain access to the cleavage site in MUC2, meprin ß must be proteolytically shed from epithelial cells. Hence, regulation of meprin ß shedding and activation is important for physiological and pathophysiological conditions. Here, we demonstrate that meprin ß activation and shedding are mutually exclusive events. Employing ex vivo small intestinal organoid and cell culture experiments, we found that ADAM-mediated shedding is restricted to the inactive pro-form of meprin ß and is completely inhibited upon its conversion to the active form at the cell surface. This strict regulation of meprin ß activity can be overridden by pathogens, as demonstrated for the bacterial protease Arg-gingipain (RgpB). This secreted cysteine protease potently converts membrane-bound meprin ß into its active form, impairing meprin ß shedding and its function as a mucus-detaching protease.


Subject(s)
Adhesins, Bacterial/metabolism , Cysteine Endopeptidases/metabolism , Metalloendopeptidases/metabolism , Metalloproteases/metabolism , Amino Acid Sequence/genetics , Animals , Cell Membrane/metabolism , Epithelial Cells/metabolism , Female , Gingipain Cysteine Endopeptidases , HEK293 Cells , Humans , Male , Metalloendopeptidases/genetics , Mice, Transgenic , Mucin-2/genetics , Mucin-2/metabolism
3.
Biopolymers ; 108(6)2017 Nov.
Article in English | MEDLINE | ID: mdl-28555756

ABSTRACT

Matriptase-2 plays a pivotal role in keeping iron concentrations within a narrow physiological range in humans. The opportunity to reduce matriptase-2 proteolytic activity may open a novel possibility to treat iron overload diseases, such as hereditary hemochromatosis and thalassemia. Here, we present 23 new analogues of trypsin inhibitor SFTI-1 designed to inhibit human matriptase-2. Influence of the modifications Gly1Lys, Ile10Arg, and Phe12His, as well as the introduction of Narg in P1 or P1 and P4 positions were examined. Selected peptides were further analyzed, together with previously reported peptides, for their inhibitory activity against related human proteases, that are, matriptase-1, plasmin, thrombin and trypsin. A highly potent inhibitor of matriptase-2, the bicycylic [Arg5 , Arg10 , His12 ]SFTI-1, with a Ki value of 15 nm was obtained.


Subject(s)
Drug Design , Helianthus/chemistry , Membrane Proteins/antagonists & inhibitors , Peptides, Cyclic/chemistry , Serine Proteinase Inhibitors/chemical synthesis , Trypsin Inhibitors/chemistry , Amino Acid Sequence , Helianthus/metabolism , Humans , Kinetics , Membrane Proteins/metabolism , Peptides, Cyclic/blood , Protein Stability , Seeds/chemistry , Seeds/metabolism , Sequence Alignment , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/metabolism , Thrombin/antagonists & inhibitors , Thrombin/metabolism , Trypsin/chemistry , Trypsin/metabolism
4.
Chemistry ; 23(22): 5205-5209, 2017 Apr 19.
Article in English | MEDLINE | ID: mdl-28370501

ABSTRACT

Activity-based probes are compounds that exclusively form covalent bonds with active enzymes. They can be utilized to profile enzyme activities in vivo, to identify target enzymes and to characterize their function. The design of a new activity-based probe for matriptase, a member of the type II transmembrane serine proteases, is based on linker-connected bis-benzguanidines. An amino acid, introduced as linker, bears the coumarin fluorophore. Moreover, an incorporated phosphonate allows for a covalent interaction with the active-site serine. The resulting irreversible mode of action was demonstrated, leading to enzyme inactivation and, simultaneously, to a fluorescence labeling of matriptase. The ten-step synthetic approach to a coumarin-labeled bis-benzguanidine and its evaluation as activity-based probe for matriptase based on in-gel fluorescence and fluorescence HPLC is reported. HPLC fluorescence detection as a new application for activity-based probes for proteases is demonstrated herein for the first time.


Subject(s)
Fluorescent Dyes/chemistry , Serine Endopeptidases/chemistry , Serine Proteases/chemistry , Serine Proteases/metabolism , Catalytic Domain , Serine Endopeptidases/metabolism
5.
FASEB J ; 31(3): 1226-1237, 2017 03.
Article in English | MEDLINE | ID: mdl-28003343

ABSTRACT

The adhesion molecule CD99 is essential for the transendothelial migration of leukocytes. In this study, we used biochemical and cellular assays to show that CD99 undergoes ectodomain shedding by the metalloprotease meprin ß and subsequent intramembrane proteolysis by γ-secretase. The cleavage site in CD99 was identified by mass spectrometry within an acidic region highly conserved through different vertebrate species. This finding fits perfectly to the unique cleavage specificity of meprin ß with a strong preference for aspartate residues and suggests coevolution of protease and substrate. We hypothesized that limited CD99 cleavage by meprin ß would alter cellular transendothelial migration (TEM) behavior in tissue remodeling processes, such as inflammation and cancer. Indeed, meprin ß induced cell migration of Lewis lung carcinoma cells in an in vitro TEM assay. Accordingly, deficiency of meprin ß in Mep1b-/- mice resulted in significantly increased CD99 protein levels in the lung. Therefore, meprin ß could serve as a therapeutic target, given that in a proof-of-concept approach we showed accumulation of CD99 protein in lungs of meprin ß inhibitor-treated mice.-Bedau, T., Peters, F., Prox, J., Arnold, P., Schmidt, F., Finkernagel, M., Köllmann, S., Wichert, R., Otte, A., Ohler, A., Stirnberg, M., Lucius, R., Koudelka, T., Tholey, A., Biasin, V., Pietrzik, C. U., Kwapiszewska, G., Becker-Pauly, C. Ectodomain shedding of CD99 within highly conserved regions is mediated by the metalloprotease meprin ß and promotes transendothelial cell migration.


Subject(s)
12E7 Antigen/metabolism , Conserved Sequence , Metalloendopeptidases/metabolism , Proteolysis , Transendothelial and Transepithelial Migration , 12E7 Antigen/chemistry , Animals , Carcinoma, Lewis Lung/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred C57BL
6.
Bioorg Med Chem Lett ; 26(15): 3741-5, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27287367

ABSTRACT

The serine protease matriptase-2 has attracted much attention as a potential target for the treatment of iron overload diseases. In this study, a series of 27 symmetric, achiral bisbenzamidines was evaluated for inhibitory activity against human matriptase-2, against the closely related enzyme human matriptase, as well as against human thrombin, bovine factor Xa and human trypsin. The conformationally restricted piperazine derivative 19 and the oxamide-derived bisbenzamidine 1 were identified as the most potent inhibitors of this series for matriptase-2 and matriptase, respectively.


Subject(s)
Benzamidines/pharmacology , Membrane Proteins/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Animals , Benzamidines/chemical synthesis , Benzamidines/chemistry , Cattle , Dose-Response Relationship, Drug , Factor Xa/metabolism , Humans , Membrane Proteins/metabolism , Molecular Structure , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship , Thrombin/antagonists & inhibitors , Thrombin/metabolism , Trypsin/metabolism
7.
Chemistry ; 22(25): 8525-35, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27214780

ABSTRACT

Matriptase-2, a type II transmembrane serine protease, plays a key role in human iron homeostasis. Inhibition of matriptase-2 is considered as an attractive strategy for the treatment of iron-overload diseases, such as hemochromatosis and ß-thalassemia. In the present study, synthetic routes to nine dipeptidomimetic inactivators were developed. Five active compounds (41-45) were identified and characterized kinetically as irreversible inhibitors of matriptase-2. In addition to a phosphonate warhead, these dipeptides possess two benzguanidine moieties as arginine mimetics to provide affinity for matriptase-2 by binding to the S1 and S3/S4 subpockets, respectively. This binding mode was strongly supported by covalent docking analysis. Compounds 41-45 were obtained as mixtures of two diastereomers and were therefore separated into the single epimers. Compound 45 A, with S configuration at the N-terminal amino acid and R configuration at the phosphonate carbon atom, was the most potent matriptase-2 inactivator with a rate constant of inactivation of 2790 m(-1) s(-1) and abolished the activity of membrane-bound matriptase-2 on the surface of intact cells. Based on the chemotyp of phosphono bisbenzguanidines, the design and synthesis of a fluorescent probe (51 A) by insertion of a coumarin label is described. The in-gel fluorescence detection of matriptase-2 was demonstrated by applying 51 A as the first activity-based probe for this enzyme.


Subject(s)
Guanidines/chemistry , Membrane Proteins/antagonists & inhibitors , Serine Proteinase Inhibitors/chemistry , Animals , Binding Sites , Catalytic Domain , Cattle , Coumarins/chemistry , Factor Xa/chemistry , Factor Xa/metabolism , Fluorescent Dyes/chemistry , Guanidines/chemical synthesis , Guanidines/metabolism , Humans , Kinetics , Membrane Proteins/metabolism , Molecular Docking Simulation , Peptidomimetics , Phosphorus/chemistry , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/metabolism , Stereoisomerism , Trypsin/chemistry , Trypsin/metabolism
8.
Biol Chem ; 397(8): 777-90, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27078672

ABSTRACT

Proteolytic processing of the amyloid precursor protein (APP) leads to amyloid-ß (Aß) peptides. So far, the mechanism of APP processing is insufficiently characterized at the molecular level. Whereas the knowledge of Aß generation by several proteases has been expanded, the contribution of the Kunitz-type protease inhibitor domain (KPI) present in two major APP isoforms to the complex proteolytic processing of APP is poorly understood. In this study, we have identified KPI-containing APP as a very potent, slow-binding inhibitor for the membrane-bound proteolytic regulator of iron homeostasis matriptase-2 by forming stable complexes with its target protease in HEK cells. Inhibition and complex formation depend on the intact KPI domain. By inhibiting matriptase-2, KPI-containing APP is protected from matriptase-2-mediated proteolysis within the Aß region, thus preventing the generation of N-terminally truncated Aß.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Membrane Proteins/antagonists & inhibitors , Amino Acid Sequence , Amyloid beta-Protein Precursor/analysis , Cells, Cultured , HEK293 Cells , Humans , Kinetics , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism
9.
Chembiochem ; 17(7): 595-604, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26762582

ABSTRACT

The cell-surface serine protease matriptase-2 is a critical stimulator of iron absorption by negatively regulating hepcidin, the key hormone of iron homeostasis. Thus, it has attracted much attention as a target in primary and secondary iron overload diseases. Here, we have characterised Kunitz-type inhibitors hepatocyte growth factor activator inhibitor 1 (HAI-1) and HAI-2 as powerful, slow-binding matriptase-2 inhibitors. The binding modes of the matriptase-2-HAI complexes were suggested by molecular modelling. Different assays, including cell-free and cell-based measurements of matriptase-2 activity, determination of inhibition constants and evaluation of matriptase-2 inhibition by analysis of downstream effects in human liver cells, demonstrated that matriptase-2 is an excellent target for Kunitz inhibitors. In particular, HAI-2 is considered a promising scaffold for the design of potent and selective matriptase-2 inhibitors.


Subject(s)
Drug Delivery Systems , Enzyme Inhibitors/therapeutic use , Iron Overload/drug therapy , Membrane Proteins/antagonists & inhibitors , Cell Line , Down-Regulation , Enzyme Activation , Humans , Iron Overload/enzymology , Liver/enzymology , Membrane Proteins/genetics , Models, Molecular , Protein Domains/genetics , Proteinase Inhibitory Proteins, Secretory/antagonists & inhibitors , Proteinase Inhibitory Proteins, Secretory/chemistry , Serine Endopeptidases/genetics
10.
Pharmaceuticals (Basel) ; 9(1)2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26771619

ABSTRACT

The liver enzyme matriptase-2 is a multi-domain, transmembrane serine protease with an extracellular, C-terminal catalytic domain. Synthetic low-molecular weight inhibitors of matriptase-2 have potential as therapeutics to treat iron overload syndromes, in particular in patients with ß-thalassemia. A sub-library of 64 compounds was screened for matriptase-2 inhibition and several active compounds were identified. (S)-Ethyl 2-(benzyl(3-((4-carbamidoylphenoxy)methyl)-2,3-dihydrobenzo[b][1,4]dioxin-6-yl)amino)-2-oxoacetate ((S)-12) showed an IC50 value of less than 10 µM. Structure-activity relationships were discussed and proposals to design new matriptase-2 inhibitors were made.

11.
Chemistry ; 22(2): 610-25, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26625703

ABSTRACT

In the absence of X-ray data, the exploration of compound binding modes continues to be a challenging task. For structure-based design, specific features of active sites in different targets play a major role in rationalizing ligand binding characteristics. For example, dibasic compounds have been reported as potent inhibitors of various trypsin-like serine proteases, the active sites of which contain several binding pockets that can be targeted by cationic moieties. This results in several possible orientations within the active site, complicating the binding mode prediction of such compounds by docking tools. Therefore, we introduced symmetry in bi- and tribasic compounds to reduce conformational space in docking calculations and to simplify binding mode selection by limiting the number of possible pocket occupations. Asymmetric bisbenzamidines were used as starting points for a multistage and structure-guided optimization. A series of 24 final compounds with either two or three benzamidine substructures was ultimately synthesized and evaluated as inhibitors of five serine proteases, leading to potent symmetric inhibitors for the pharmaceutical drug targets matriptase, matriptase-2, thrombin and factor Xa. This study underlines the relevance of ligand symmetry for chemical biology.


Subject(s)
Membrane Proteins/chemistry , Peptidomimetics/chemistry , Serine Endopeptidases/chemistry , Serine Proteinase Inhibitors/chemistry , Thrombin/chemistry , Benzamidines/chemical synthesis , Benzamidines/chemistry , Binding Sites , Crystallography, X-Ray , Ligands , Models, Molecular , Protein Binding , Serine Endopeptidases/metabolism
12.
ChemMedChem ; 10(10): 1641-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26306030

ABSTRACT

A hybrid approach was applied for the design of an inhibitor of trypsin-like serine proteases. Compound 16 [(R,R)- and (R,S)-diphenyl (4-(1-(4-amidinobenzylamino)-1-oxo-3-phenylpropan-2-ylcarbamoyl)phenylamino)(4-amidinophenyl)methylphosphonate hydrochloride], prepared in a convergent synthetic procedure, possesses a phosphonate warhead prone to react with the active site serine residue in a covalent, irreversible manner. Each of the two benzamidine moieties of 16 can potentially be accommodated in the S1 pocket of the target enzyme, but only the benzamidine close to the phosphonate group would then promote an irreversible interaction. The Janus-faced inhibitor 16 was evaluated against several serine proteases and caused a pronounced inactivation of human thrombin with a second-order rate constant (kinac /Ki) of 59 500 M(-1) s(-1). With human matriptase, 16 showed preference for a reversible mode of inhibition (IC50 =2.6 µM) as indicated by linear progress curves and enzyme reactivation.


Subject(s)
Benzamidines/pharmacology , Organophosphonates/pharmacology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Benzamidines/chemistry , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Humans , Molecular Structure , Organophosphonates/chemistry , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship , Thrombin/antagonists & inhibitors , Thrombin/metabolism
13.
Org Biomol Chem ; 13(41): 10310-23, 2015 Nov 07.
Article in English | MEDLINE | ID: mdl-26307943

ABSTRACT

Besides their extracellular activity crucial for several pathophysiological conditions, human cysteine cathepsins, in particular cathepsins K and S, represent important intracellular targets for drug development. In the present study, a prototypic dipeptide nitrile inhibitor structure was equipped with a coumarin moiety to function as a fluorescent reporter group. In a second inhibitor, a PEG linker was introduced between the dipeptide nitrile and the fluorophore. These tool compounds 6 and 7 were characterized by kinetic investigations as covalent reversible inhibitors of human cathepsins L, S, K and B. Probe 6 showed a pronounced inhibitory activity against cathepsins K and S, which was corroborated by modeling of inhibition modes. Probe 7 was highly potent (Ki = 93 nM) and selective for cathepsin S. To examine the ability of both probes to enter living cells, human embryonic kidney 293 cells were targeted. At a concentration of 10 µM, cellular uptake of probe 6 was demonstrated by fluorescence measurement after an incubation time of 30 min and 3 h, respectively. The probe's concentration in cell lysates was ascertained on the basis of the emission at 492 nm upon excitation at 450 nm, and the results were expressed as concentrations of probe 6 relative to the protein concentration originating from the lysate. After incubation of 10 µM of probe 6 for 3 h, the cellular uptake was confirmed by fluorescence microscopy. HPLC was used to assess the probes' lipophilicity, and the obtained


Subject(s)
Cathepsins/antagonists & inhibitors , Cells/metabolism , Cysteine Proteinase Inhibitors/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Drug Design , Fluorescent Dyes/chemistry , Cathepsins/metabolism , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/chemistry , Dose-Response Relationship, Drug , Fluorescent Dyes/analysis , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship
14.
Biochem J ; 470(1): 91-103, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26251449

ABSTRACT

Increased expression of metalloprotease meprin ß is associated with fibrotic syndromes and Alzheimer's disease (AD). Hence, regulation of meprin activity might be a suitable strategy for the treatment of these conditions. Meprin ß is a type 1 transmembrane protein, but can be released from the cell surface by ectodomain shedding. The protease is expressed as an inactive zymogen and requires proteolytic maturation by tryptic serine proteases. In the present study, we demonstrate, for the first time, the differences in the activation of soluble and membrane bound meprin ß and suggest transmembrane serine protease 6 [TMPRSS6 or matriptase-2 (MT2)] as a new potent activator, cleaving off the propeptide of meprin ß between Arg(61) and Asn(62) as determined by MS. We show that MT2, but not TMPRSS4 or pancreatic trypsin, is capable of activating full-length meprin ß at the cell surface, analysed by specific fluorogenic peptide cleavage assay, Western blotting and confocal laser scanning microscopy (CLSM). Maturation of full-length meprin ß is required for its activity as a cell surface sheddase, releasing the ectodomains of transmembrane proteins, as previously shown for the amyloid precursor protein (APP).


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cell Membrane/metabolism , Metalloendopeptidases/metabolism , Serine Endopeptidases/metabolism , Amino Acid Sequence , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Animals , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Molecular Sequence Data , Protein Structure, Secondary , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Swine
15.
Chembiochem ; 16(11): 1601-7, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-25999208

ABSTRACT

A series of 17 new analogues of trypsin inhibitor SFTI-1 were designed and synthesized to obtain matriptase-2 inhibitors. A number of the modified bicyclic peptides displayed much higher affinity towards matriptase-2 than towards the highly homologous matriptase-1. Replacement of Lys5 by Arg in the wild-type SFTI-1 led to an 11-fold increase in the matriptase-2 inhibitory activity. Replacement of Arg2 by its enantiomer (D-arginine) slightly lowered the inhibition of matriptase-2, but almost completely abolished the affinity towards matriptase-1, thus yielding the most selective matriptase-2 inhibitor. This is the first report describing inhibitors of the recently discovered matriptase-2 based on the SFTI-1 structure. The results showed that SFTI-1 is a promising scaffold for the design of potent and selective inhibitors of this enzyme.


Subject(s)
Membrane Proteins/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Trypsin Inhibitors/pharmacology , Amino Acid Sequence , HEK293 Cells , Humans , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Serine Endopeptidases , Trypsin Inhibitors/chemical synthesis , Trypsin Inhibitors/chemistry
16.
Biol Chem ; 396(1): 81-93, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25205713

ABSTRACT

Matriptase-2 is a type II transmembrane serine protease controlling the expression of hepcidin, the key regulator of iron homeostasis. By cleaving hemojuvelin, matriptase-2 suppresses bone morphogenetic protein/sons of mothers against decapentaplegic signaling. So far, the only known putative substrates of matriptase-2 are hemojuvelin and matriptase-2 itself. In this study, fetuin-A (α2-Heremans-Schmid glycoprotein) was identified in vitro as a substrate of matriptase-2. The protease-substrate interaction was validated by isolating matriptase-2 via the affinity to fetuin-A. Fetuin-A is a liver-derived plasma protein with multiple functions, which is proteolytically processed to yield a disulfide-linked two-chain form. In co-transfected cells, a matriptase-2-dependent conversion of unprocessed fetuin-A into a two-chain form was detected. Conversely, downregulation of endogenously expressed matriptase-2 stabilized fetuin-A. Arg and Lys residues located within the 40 residue spanning connecting peptide of fetuin-A were identified as cleavage sites for matriptase-2. Analysis of hepcidin expression revealed an inductive effect of fetuin-A, which was abolished by matriptase-2. Fetuin-A deficiency in mice resulted in decreased hepcidin mRNA levels. These findings implicate a role of fetuin-A in iron homeostasis and provide new insights into the mechanism of how matriptase-2 might modulate hepcidin expression.


Subject(s)
Fetuins/metabolism , Hepcidins/metabolism , Membrane Proteins/genetics , Serine Endopeptidases/genetics , Animals , Fetuins/genetics , Hepcidins/genetics , Mice , Serine Proteases , Signal Transduction
17.
Org Biomol Chem ; 11(35): 5913-21, 2013 Sep 21.
Article in English | MEDLINE | ID: mdl-23912233

ABSTRACT

An activity-based probe, containing an irreversibly locked GFP-like fluorophore, was synthesized and evaluated as an inhibitor of human cathepsins and, as exemplified with cathepsin K, it proved to be suitable for ex vivo imaging and quantification of cysteine cathepsins by SDS-PAGE.


Subject(s)
Cathepsins/analysis , Cysteine/analysis , Electrophoresis, Polyacrylamide Gel/methods , Fluorescent Dyes/analysis , Green Fluorescent Proteins/analysis , Cathepsin K/analysis , Fluorescence , HEK293 Cells , Humans
18.
Biochem J ; 450(3): 583-93, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23293962

ABSTRACT

Matriptase-2, a recently identified cell surface protease, is the key enzyme of iron homoeostasis modulating the expression of the liver peptide hormone hepcidin. HAI (hepatocyte growth factor activator inhibitor) types 1 and 2 (HAI-1 and HAI-2 respectively) have been shown to inhibit the close homologue, i.e. matriptase. By co-expressing matriptase-2 and the inhibitor HAI-2 we have identified HAI-2 displaying high inhibitory potential against matriptase-2 at the cell surface as well as in conditioned medium. Accordingly, complex formation between matriptase-2 and HAI-2 was demonstrated by isolation of the complex via immobilizing either HAI-2 or matriptase-2 from lysates and conditioned medium of co-expressing cells. Furthermore, HAI-2 indirectly influences the expression of the hepcidin-encoding gene HAMP. The inhibitor abrogates the matriptase-2-mediated suppression of HAMP expression, presumably by inhibiting the supposed potential of matriptase-2 to cleave membrane-bound HJV (haemojuvelin). Taken together, the results of the present study have characterized HAI-2 as an inhibitor of matriptase-2 that modulates the synthesis of hepcidin and provides new insights into the regulatory mechanism of iron homoeostasis, with clinical importance for a treatment of iron overload diseases.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Membrane Glycoproteins/physiology , Membrane Proteins/antagonists & inhibitors , Antimicrobial Cationic Peptides/metabolism , Biomimetics , Cell Hypoxia/physiology , Gene Expression Regulation/drug effects , HEK293 Cells , Hep G2 Cells , Hepcidins , Humans , Iron/pharmacology , Iron Deficiencies , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , RNA, Small Interfering/pharmacology , Serine Endopeptidases/metabolism , Serine Proteases/metabolism , Serine Proteinase Inhibitors/genetics , Serine Proteinase Inhibitors/metabolism , Serine Proteinase Inhibitors/physiology , Tumor Cells, Cultured
19.
Curr Pharm Des ; 19(6): 1052-61, 2013.
Article in English | MEDLINE | ID: mdl-23016685

ABSTRACT

Matriptase-2 is a cell surface serine protease with a modular structure. The exploration of its function in iron homeostasis was of significance for the understanding of the regulation of hepcidin expression, the master protein in iron control. Mutations in matriptase- 2 cause iron-refractory iron deficiency anemia (IRIDA), an iron deficiency disorder where the level of hepcidin is inappropriately high. Matriptase-2 controls hepcidin expression through the suppression of bone morphogenetic protein (BMP)/sons of mothers against decapentaplegic homologue protein (SMAD) signaling, probably by cleaving the BMP co-receptor hemojuvelin. Since prospective studies revealed that genetic inactivation of matriptase-2 reduces iron loading in different mouse models, matriptase-2 becomes highly attractive as a novel target for the design of low-molecular weight inhibitors. The first synthetic peptidomimetic matriptase-2 inhibitors have been reported. A computational model of the active site of matriptase-2 based on the X-ray crystal structure of the close homologue matriptase was generated and mutational studies were performed in order to identify critical amino acids specifying the preferred recognition site of matriptase-2. So far, the only known putative natural substrates of matriptase-2 are hemojuvelin and matriptase-2 itself, as this protease undergoes complex auto-processing during zymogen activation. Cleavage sites within both natural substrates were identified.


Subject(s)
Homeostasis/physiology , Iron/metabolism , Membrane Proteins/metabolism , Protease Inhibitors/pharmacology , Serine Endopeptidases/metabolism , Amino Acid Sequence , Animals , Humans , Membrane Proteins/chemistry , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid , Serine Endopeptidases/chemistry , Substrate Specificity
20.
Bioorg Med Chem ; 20(21): 6489-505, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23026080

ABSTRACT

The benzamidine moiety, a well-known arginine mimetic, has been introduced in a variety of ligands, including peptidomimetic inhibitors of trypsin-like serine proteases. According to their primary substrate specificity, the benzamidine residue interacts with the negatively charged aspartate at the bottom of the S1 pocket of such enzymes. Six series of benzamidine derivatives (1-73) were synthesized and evaluated as inhibitors of two prototype serine proteases, that is, bovine trypsin and human thrombin. As a further target, human matriptase-2, a recently discovered type II transmembrane serine protease, was investigated. Matriptase-2 represents an important regulatory protease in iron homeostasis by down-regulation of the hepcidin expression. Compounds 1-73 were designed to contain a fixed sulfamoyl benzamidine moiety as arginine mimetic and a linker-connected additional substructure, such as a tert-butyl ester, carboxylate or second benzamidine functionality. A systematic mapping approach was performed with these inhibitors to scan the active site of the three target proteases. In particular, bisbenzamidines, able to interact with both the S1 and S3/S4 binding sites, showed notable affinity. In branched bisbenzamidines 66-73 containing a third hydrophobic residue, opposite effects of the stereochemistry on trypsin and thrombin inhibition were observed.


Subject(s)
Benzamidines/pharmacology , Enzyme Inhibitors/pharmacology , Membrane Proteins/antagonists & inhibitors , Thrombin/antagonists & inhibitors , Trypsin/metabolism , Animals , Benzamidines/chemical synthesis , Benzamidines/chemistry , Catalytic Domain/drug effects , Cattle , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Membrane Proteins/metabolism , Molecular Structure , Serine Endopeptidases/metabolism , Structure-Activity Relationship , Thrombin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...