Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Acta Pharm Sin B ; 14(5): 2349-2357, 2024 May.
Article in English | MEDLINE | ID: mdl-38799620

ABSTRACT

A titrant for the SARS-CoV-2 main protease (Mpro) was developed that enables, for the first time, the exact determination of the concentration of the enzymatically active Mpro by active-site titration. The covalent binding mode of the tetrapeptidic titrant was elucidated by the determination of the crystal structure of the enzyme-titrant complex. Four fluorogenic substrates of Mpro, including a prototypical, internally quenched Dabcyl-EDANS peptide, were compared in terms of solubility under typical assay conditions. By exploiting the new titrant, key kinetic parameters for the Mpro-catalyzed cleavage of these substrates were determined.

2.
ACS Pharmacol Transl Sci ; 5(11): 1097-1108, 2022 Nov 11.
Article in English | MEDLINE | ID: mdl-36407962

ABSTRACT

The multifactorial nature of Alzheimer's disease necessitates the development of agents able to interfere with different relevant targets. A series of 22 tailored chromanones was conceptualized, synthesized, and subjected to biological evaluation. We identified one representative bearing a linker-connected azepane moiety (compound 19) with balanced pharmacological properties. Compound 19 exhibited inhibitory activities against human acetyl-, butyrylcholinesterase and monoamine oxidase-B, as well as high affinity to both the σ1 and σ2 receptors. Our study provides a framework for the development of further chromanone-based multineurotarget agents.

3.
J Enzyme Inhib Med Chem ; 37(1): 515-526, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35144520

ABSTRACT

Cathepsin K (CatK) is a target for the treatment of osteoporosis, arthritis, and bone metastasis. Peptidomimetics with a cyanohydrazide warhead represent a new class of highly potent CatK inhibitors; however, their binding mechanism is unknown. We investigated two model cyanohydrazide inhibitors with differently positioned warheads: an azadipeptide nitrile Gü1303 and a 3-cyano-3-aza-ß-amino acid Gü2602. Crystal structures of their covalent complexes were determined with mature CatK as well as a zymogen-like activation intermediate of CatK. Binding mode analysis, together with quantum chemical calculations, revealed that the extraordinary picomolar potency of Gü2602 is entropically favoured by its conformational flexibility at the nonprimed-primed subsites boundary. Furthermore, we demonstrated by live cell imaging that cyanohydrazides effectively target mature CatK in osteosarcoma cells. Cyanohydrazides also suppressed the maturation of CatK by inhibiting the autoactivation of the CatK zymogen. Our results provide structural insights for the rational design of cyanohydrazide inhibitors of CatK as potential drugs.


Subject(s)
Cathepsin K/antagonists & inhibitors , Hydrazines/pharmacology , Nitriles/pharmacology , Protease Inhibitors/pharmacology , Cathepsin K/metabolism , Dose-Response Relationship, Drug , Humans , Hydrazines/chemistry , Models, Molecular , Molecular Structure , Nitriles/chemistry , Protease Inhibitors/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
4.
J Med Chem ; 64(18): 13793-13806, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34473502

ABSTRACT

The cysteine protease cathepsin K is a target for the treatment of diseases associated with high bone turnover. Cathepsin K is mainly expressed in osteoclasts and responsible for the destruction of the proteinaceous components of the bone matrix. We designed various fluorescent activity-based probes (ABPs) and their precursors that bind to and inactivate cathepsin K. ABP 25 exhibited extraordinary potency (kinac/Ki = 35,300 M-1s-1) and selectivity for human cathepsin K. Crystal structures of cathepsin K in complex with ABP 25 and its nonfluorescent precursor 21 were determined to characterize the binding mode of this new type of acrylamide-based Michael acceptor with the particular orientation of the dibenzylamine moiety to the primed subsite region. The cyanine-5 containing probe 25 allowed for sensitive detection of cathepsin K, selective visualization in complex proteomes, and live cell imaging of a human osteosarcoma cell line, underlining its applicability in a pathophysiological environment.


Subject(s)
Acrylamides/chemistry , Cathepsin K/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemistry , Fluorescent Dyes/chemistry , Acrylamides/chemical synthesis , Acrylamides/metabolism , Catalytic Domain , Cathepsin K/chemistry , Cathepsin K/metabolism , Cell Line, Tumor , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/metabolism , Drug Design , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Protein Binding
5.
Bioorg Med Chem Lett ; 30(18): 127420, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32763808

ABSTRACT

A library of cathepsin S inhibitors of the dipeptide nitrile chemotype, bearing a bioisosteric sulfonamide moiety, was synthesized. Kinetic investigations were performed at four human cysteine proteases, i.e. cathepsins S, B, K and L. Compound 12 with a terminal 3-biphenyl sulfonamide substituent was the most potent (Ki = 4.02 nM; selectivity ratio cathepsin S/K = 5.8; S/L = 67) and 24 with a 4'-fluoro-4-biphenyl sulfonamide substituent the most selective cathepsin S inhibitor (Ki = 35.5 nM; selectivity ratio cathepsin S/K = 57; S/L = 31). In silico design and biochemical evaluation emphasized the impact of the sulfonamide linkage on selectivity and a possible switch of P2 and P3 substituents with respect to the occupation of the corresponding binding sites of cathepsin S.


Subject(s)
Cathepsins/antagonists & inhibitors , Dipeptides/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Nitriles/chemical synthesis , Sulfonamides/chemistry , Amino Acid Sequence , Binding Sites , Cathepsin K/metabolism , Cathepsin L/metabolism , Computer Simulation , Cysteine Proteases/metabolism , Humans , Kinetics , Protein Binding , Structure-Activity Relationship
6.
Eur J Pharmacol ; 883: 173183, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32534072

ABSTRACT

Although agonists and antagonists of muscarinic receptors have been known for long time, there is renewed interest in compounds (such as allosteric or bitopic ligands, or biased agonists) able to differently and selectively modulate these receptors. As a continuation of our previous research, we designed a new series of dimers of the well-known cholinergic agonist carbachol. The new compounds were tested on the five cloned human muscarinic receptors (hM1-5) expressed in CHO cells by means of equilibrium binding experiments, showing a dependence of the binding affinity on the length and position of the linker connecting the two monomers. Kinetic binding studies revealed that some of the tested compounds were able to slow the rate of NMS dissociation, suggesting allosteric behavior, also supported by docking simulations. Assessment of ERK1/2 phosphorylation on hM1, hM2 and hM3 activation showed that the new compounds are endowed with muscarinic antagonist properties. At hM2 receptors, some compounds were able to stimulate GTPγS binding but not cAMP accumulation, suggesting a biased behavior. Classification, Molecular and cellular pharmacology.


Subject(s)
Carbachol/pharmacology , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Receptors, Muscarinic/drug effects , Animals , CHO Cells , Carbachol/chemistry , Carbachol/metabolism , Cricetulus , Cyclic AMP/metabolism , Dimerization , Extracellular Signal-Regulated MAP Kinases/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Kinetics , Molecular Docking Simulation , Molecular Structure , Muscarinic Agonists/chemistry , Muscarinic Agonists/metabolism , Muscarinic Antagonists/chemistry , Muscarinic Antagonists/metabolism , Phosphorylation , Protein Binding , Receptors, Muscarinic/genetics , Receptors, Muscarinic/metabolism , Signal Transduction , Structure-Activity Relationship
7.
Biochim Biophys Acta Proteins Proteom ; 1868(9): 140445, 2020 09.
Article in English | MEDLINE | ID: mdl-32405284

ABSTRACT

Coumarins represent well-established structures to introduce fluorescence into tool compounds for biochemical investigations. They are valued for their small size, chemical stability and accessibility as well as their tunable photochemical properties. As components of fluorophore/quencher pairs or FRET donor/acceptor pairs, coumarins have frequently been applied in substrate mapping approaches for serine and cysteine proteases. This review also focuses on the incorporation of coumarins into the side chain of amino acids and the exploitation of the resulting fluorescent amino acids for the positional profiling of protease substrates. The protease-inhibiting properties of certain coumarin derivatives and the utilization of coumarin moieties to assemble activity-based probes for serine and cysteine proteases are discussed as well.


Subject(s)
Coumarins/chemistry , Coumarins/metabolism , Cysteine Proteases/metabolism , Serine Proteases/metabolism , Catalytic Domain , Coumarins/pharmacology , Cysteine Proteases/drug effects , Fluorescence , Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes/chemistry , Serine/metabolism , Serine Proteases/drug effects , Substrate Specificity
8.
ACS Omega ; 4(26): 22161-22168, 2019 Dec 24.
Article in English | MEDLINE | ID: mdl-31891098

ABSTRACT

The complex nature of multifactorial diseases, such as Morbus Alzheimer, has produced a strong need to design multitarget-directed ligands to address the involved complementary pathways. We performed a purposive structural modification of a tetratarget small-molecule, that is contilisant, and generated a combinatorial library of 28 substituted chromen-4-ones. The compounds comprise a basic moiety which is linker-connected to the 6-position of the heterocyclic chromenone core. The syntheses were accomplished by Mitsunobu- or Williamson-type ether formations. The resulting library members were evaluated at a panel of seven human enzymes, all of which being involved in the pathophysiology of neurodegeneration. A concomitant inhibition of human acetylcholinesterase and human monoamine oxidase B, with IC50 values of 5.58 and 7.20 µM, respectively, was achieved with the dual-target 6-(4-(piperidin-1-yl)butoxy)-4H-chromen-4-one (7).

9.
Bioorg Med Chem ; 27(1): 1-15, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30473362

ABSTRACT

The potential of papain-like cysteine proteases, such as cathepsin B, as drug discovery targets for systemic human diseases has prevailed over the past years. The development of potent and selective low-molecular cathepsin B inhibitors relies on the detailed expertise on preferred amino acid and inhibitor residues interacting with the corresponding specificity pockets of cathepsin B. Such knowledge might be obtained by mapping the active site of the protease with combinatorial libraries of peptidic substrates and peptidomimetic inhibitors. This review, for the first time, summarizes a wide spectrum of active site mapping approaches. It considers relevant X-ray crystallographic data and discloses propensities towards favorable protein-ligand interactions in case of the therapeutically relevant protease cathepsin B.


Subject(s)
Cathepsin B/chemistry , Cysteine Proteinase Inhibitors/chemistry , Peptides/chemistry , Animals , Catalytic Domain , Crystallography, X-Ray , Humans , Kinetics , Ligands , Substrate Specificity
10.
Bioorg Med Chem Lett ; 28(11): 2008-2012, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29748052

ABSTRACT

A series of inhibitors targeting human cathepsins have been designed and synthesized following a combinatorial approach. The compounds bear an α,ß-unsaturated phenyl vinyl sulfone or ethyl acrylate warhead and a peptidomimetic portion aligned to the non-primed binding region. Biochemical evaluation toward four human cathepsins was carried out and the kinetic characterization confirmed an irreversible mode of inhibition. Compound 6c combining the most advantageous building blocks for cathepsin S inhibition was identified as a potent cathepsin S inactivator exhibiting a second-order rate constant of 30600 M-1 s-1.


Subject(s)
Acrylates/pharmacology , Cathepsins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Sulfones/pharmacology , Acrylates/chemical synthesis , Acrylates/chemistry , Cathepsins/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Kinetics , Molecular Structure , Structure-Activity Relationship , Sulfones/chemical synthesis , Sulfones/chemistry
11.
ACS Med Chem Lett ; 7(3): 211-6, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26985300

ABSTRACT

An active site mapping of human cathepsin B with dipeptide nitrile inhibitors was performed for a combinatorial approach by introducing several points of diversity and stepwise optimizing the inhibitor structure. To address the occluding loop of cathepsin B by a carboxylate moiety, click chemistry to generate linker-connected molecules was applied. Inhibitor 17 exhibited K i values of 41.3 nM, 27.3 nM, or 19.2 nM, depending on the substrate and pH of the assay. Kinetic data were discussed with respect to the conformational selection and induced fit models.

12.
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
13.
ChemMedChem ; 10(8): 1365-77, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26119278

ABSTRACT

Cleavage of the invariant chain is the key event in the trafficking pathway of major histocompatibility complex class II. Cathepsin S is the major processing enzyme of the invariant chain, but cathepsin F acts in macrophages as its functional synergist which is as potent as cathepsin S in invariant chain cleavage. Dedicated low-molecular-weight inhibitors for cathepsin F have not yet been developed. An active site mapping with 52 dipeptide nitriles, reacting as covalent-reversible inhibitors, was performed to draw structure-activity relationships for the non-primed binding region of human cathepsin F. In a stepwise process, new compounds with optimized fragment combinations were designed and synthesized. These dipeptide nitriles were evaluated on human cysteine cathepsins F, B, L, K and S. Compounds 10 (N-(4-phenylbenzoyl)-leucylglycine nitrile) and 12 (N-(4-phenylbenzoyl)leucylmethionine nitrile) were found to be potent inhibitors of human cathepsin F, with Ki values <10 nM. With all dipeptide nitriles from our study, a 3D activity landscape was generated to visualize structure-activity relationships for this series of cathepsin F inhibitors.


Subject(s)
Cathepsin F/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemistry , Dipeptides/chemistry , Nitriles/chemistry , Binding Sites , Catalytic Domain , Cathepsin F/metabolism , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/metabolism , Humans , Molecular Docking Simulation , Nitriles/chemical synthesis , Nitriles/metabolism , Protein Binding , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Structure-Activity Relationship
14.
ACS Med Chem Lett ; 5(10): 1076-81, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25313316

ABSTRACT

Nitrile-type inhibitors are known to interact with cysteine proteases in a covalent-reversible manner. The chemotype of 3-cyano-3-aza-ß-amino acid derivatives was designed in which the N-cyano group is centrally arranged in the molecule to allow for interactions with the nonprimed and primed binding regions of the target enzymes. These compounds were evaluated as inhibitors of the human cysteine cathepsins K, S, B, and L. They exhibited slow-binding behavior and were found to be exceptionally potent, in particular toward cathepsin K, with second-order rate constants up to 52 900 × 10(3) M(-1) s(-1).

15.
Steroids ; 72(6-7): 545-51, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17509632

ABSTRACT

In human sports doping control analysis most of the steroids are analyzed after enzymatic hydrolysis of the glucuronides as per-trimethylsilyl (TMS) derivatives applying gas chromatography-mass spectrometry (GC-MS). According to the recommendations of the World Anti-Doping Agency the identification of analytes should be based on retention time and on mass spectrometric characterization. This study shows that the bis-TMS derivatives of 16 specific C19 steroids, namely the stereoisomers of 5xi-androst-1-ene-3xi,17xi-diol (8 isomers), androst-4-ene-3xi,17xi-diol (4 isomers), and 17xi-hydroxy-5xi-androstan-3-one (4 isomers), reveal very similar mass spectra. As a rule, when taking the retention times, which are provided as Kovac indices for all these isomers, into account, a restriction to two or three possible isomers is possible. Reliable identification should additionally include a comparison of the retention times of the analytes with the reference compounds measured concomitantly. In some cases standard addition may be appropriate. Due to the limited availability, the above mentioned isomers were synthesized by reduction of the corresponding alpha,beta-unsaturated oxo steroids either with K-Selectride or by catalytic hydrogenation (Pd/C as catalyst). The products of the reactions were identified by means of nuclear magnetic resonance (NMR) characterization and by further reduction to the corresponding 5xi-androstane-3xi,17xi-diols and GC-MS comparison with commercially available reference standards.


Subject(s)
Androstanes/chemistry , Gas Chromatography-Mass Spectrometry , Isomerism , Magnetic Resonance Spectroscopy
SELECTION OF CITATIONS
SEARCH DETAIL
...