Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Acta Crystallogr D Struct Biol ; 80(Pt 2): 123-136, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38289714

ABSTRACT

To identify starting points for therapeutics targeting SARS-CoV-2, the Paul Scherrer Institute and Idorsia decided to collaboratively perform an X-ray crystallographic fragment screen against its main protease. Fragment-based screening was carried out using crystals with a pronounced open conformation of the substrate-binding pocket. Of 631 soaked fragments, a total of 29 hits bound either in the active site (24 hits), a remote binding pocket (three hits) or at crystal-packing interfaces (two hits). Notably, two fragments with a pose that was sterically incompatible with a more occluded crystal form were identified. Two isatin-based electrophilic fragments bound covalently to the catalytic cysteine residue. The structures also revealed a surprisingly strong influence of the crystal form on the binding pose of three published fragments used as positive controls, with implications for fragment screening by crystallography.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Catalytic Domain , Coronavirus 3C Proteases , Crystallography, X-Ray
2.
Biochem Pharmacol ; 206: 115291, 2022 12.
Article in English | MEDLINE | ID: mdl-36306820

ABSTRACT

Selatogrel is a potent inhibitor of adenosine diphosphate (ADP) binding to the P2Y12 receptor, preventing platelet activation. We have previously shown that the P2Y12 receptor constitutively activates Gi- and Go-protein-mediated signaling in human platelets. Here, we report that selatogrel acts as an inverse agonist of the P2Y12 receptor. Specifically, using bioluminescence resonance energy transfer2 (BRET2) probes, selatogrel, ticagrelor, and elinogrel were shown to stabilize the inactive form of the Gαi/o-Gßγ complex in cells with recombinant expression of the P2Y12 receptor. In dose-response experiments, while selatogrel exhibited a maximal efficacy similar to ticagrelor, selatogrel was approximately 100-fold more potent than ticagrelor. Quantification of relative cyclic adenosine monophosphate (cAMP) levels in cells expressing the cAMP BRET1 sensor (CAMYEL probe) confirmed that selatogrel completely abolished the constitutive activity of the P2Y12 receptor. In agreement, selatogrel increased basal cAMP levels in human platelets, confirming inverse agonism on the endogenous human platelet P2Y12 receptor. In agreement with the biochemical phenotype of inverse agonism efficacy of selatogrel, the 2.8 Angstrom resolution cocrystal structure of selatogrel bound to the P2Y12 receptor confirmed that selatogrel stabilizes the inactive, basal state of the receptor. Selatogrel bound to pocket 1, spanning helix III to VII. Furthermore, the binding mode of selatogrel, suggesting steric overlap with the proposed binding site of ADP and the ADP analog 2-methylthioadenosine diphosphate (2MeSADP), agrees with the functional characterization of selatogrel preventing platelet activation by blocking ADP binding to the P2Y12 receptor.


Subject(s)
Platelet Activation , Purinergic P2Y Receptor Antagonists , Humans , Ticagrelor/metabolism , Purinergic P2Y Receptor Antagonists/pharmacology , Purinergic P2Y Receptor Antagonists/metabolism , Blood Platelets , Adenosine Diphosphate/metabolism , Platelet Aggregation
3.
J Med Chem ; 63(11): 5697-5722, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32073845

ABSTRACT

The alternative pathway (AP) of the complement system is a key contributor to the pathogenesis of several human diseases including age-related macular degeneration, paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), and various glomerular diseases. The serine protease factor B (FB) is a key node in the AP and is integral to the formation of C3 and C5 convertase. Despite the prominent role of FB in the AP, selective orally bioavailable inhibitors, beyond our own efforts, have not been reported previously. Herein we describe in more detail our efforts to identify FB inhibitors by high-throughput screening (HTS) and leveraging insights from several X-ray cocrystal structures during optimization efforts. This work culminated in the discovery of LNP023 (41), which is currently being evaluated clinically in several diverse AP mediated indications.


Subject(s)
Benzoic Acid/chemistry , Complement Factor B/antagonists & inhibitors , Indoles/chemistry , Atypical Hemolytic Uremic Syndrome/metabolism , Atypical Hemolytic Uremic Syndrome/pathology , Benzoic Acid/metabolism , Benzoic Acid/pharmacokinetics , Binding Sites , Catalytic Domain , Complement Factor B/metabolism , Crystallography, X-Ray , Drug Evaluation, Preclinical , Half-Life , Humans , Indoles/metabolism , Indoles/pharmacokinetics , Inhibitory Concentration 50 , Macular Degeneration/metabolism , Macular Degeneration/pathology , Molecular Dynamics Simulation , Structure-Activity Relationship
4.
J Med Chem ; 63(1): 66-87, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31804826

ABSTRACT

UDP-3-O-((R)-3-hydroxymyristoyl)-N-glucosamine deacetylase (LpxC) is as an attractive target for the discovery and development of novel antibacterial drugs to address the critical medical need created by multidrug resistant Gram-negative bacteria. By using a scaffold hopping approach on a known family of methylsulfone hydroxamate LpxC inhibitors, several hit series eliciting potent antibacterial activities against Enterobacteriaceae and Pseudomonas aeruginosa were identified. Subsequent hit-to-lead optimization, using cocrystal structures of inhibitors bound to Pseudomonas aeruginosa LpxC as guides, resulted in the discovery of multiple chemical series based on (i) isoindolin-1-ones, (ii) 4,5-dihydro-6H-thieno[2,3-c]pyrrol-6-ones, and (iii) 1,2-dihydro-3H-pyrrolo[1,2-c]imidazole-3-ones. Synthetic methods, antibacterial activities and relative binding affinities, as well as physicochemical properties that allowed compound prioritization are presented. Finally, in vivo properties of lead molecules which belong to the most promising pyrrolo-imidazolone series, such as 18d, are discussed.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anti-Bacterial Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Escherichia coli Infections/drug therapy , Gram-Negative Bacteria/drug effects , Hydroxamic Acids/therapeutic use , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacokinetics , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Escherichia coli/drug effects , Female , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacokinetics , Klebsiella pneumoniae/drug effects , Mice, Inbred ICR , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/enzymology , Pyrroles/chemical synthesis , Pyrroles/pharmacokinetics , Pyrroles/therapeutic use
5.
J Med Chem ; 62(9): 4656-4668, 2019 05 09.
Article in English | MEDLINE | ID: mdl-30995036

ABSTRACT

Complement factor D (FD), a highly specific S1 serine protease, plays a central role in the amplification of the alternative complement pathway (AP) of the innate immune system. Dysregulation of AP activity predisposes individuals to diverse disorders such as age-related macular degeneration, atypical hemolytic uremic syndrome, membranoproliferative glomerulonephritis type II, and paroxysmal nocturnal hemoglobinuria. Previously, we have reported the screening efforts and identification of reversible benzylamine-based FD inhibitors (1 and 2) binding to the open active conformation of FD. In continuation of our drug discovery program, we designed compounds applying structure-based approaches to improve interactions with FD and gain selectivity against S1 serine proteases. We report herein the design, synthesis, and medicinal chemistry optimization of the benzylamine series culminating in the discovery of 12, an orally bioavailable and selective FD inhibitor. 12 demonstrated systemic suppression of AP activation in a lipopolysaccharide-induced AP activation model as well as local ocular suppression in intravitreal injection-induced AP activation model in mice expressing human FD.


Subject(s)
Benzylamines/pharmacology , Complement Pathway, Alternative/drug effects , Serine Proteinase Inhibitors/pharmacology , Animals , Benzylamines/chemical synthesis , Benzylamines/metabolism , Binding Sites , Complement Factor D/antagonists & inhibitors , Complement Factor D/chemistry , Complement Factor D/metabolism , Dogs , Drug Design , Humans , Mice, Inbred C57BL , Mice, Transgenic , Molecular Docking Simulation , Protein Conformation , Rats , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/metabolism
6.
Proc Natl Acad Sci U S A ; 116(16): 7926-7931, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30926668

ABSTRACT

Dysregulation of the alternative complement pathway (AP) predisposes individuals to a number of diseases including paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, and C3 glomerulopathy. Moreover, glomerular Ig deposits can lead to complement-driven nephropathies. Here we describe the discovery of a highly potent, reversible, and selective small-molecule inhibitor of factor B, a serine protease that drives the central amplification loop of the AP. Oral administration of the inhibitor prevents KRN-induced arthritis in mice and is effective upon prophylactic and therapeutic dosing in an experimental model of membranous nephropathy in rats. In addition, inhibition of factor B prevents complement activation in sera from C3 glomerulopathy patients and the hemolysis of human PNH erythrocytes. These data demonstrate the potential therapeutic value of using a factor B inhibitor for systemic treatment of complement-mediated diseases and provide a basis for its clinical development.


Subject(s)
Complement Factor B/antagonists & inhibitors , Complement Pathway, Alternative/drug effects , Drug Discovery/methods , Immunologic Factors/pharmacology , Animals , Disease Models, Animal , Glomerulonephritis, Membranous/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Rats, Sprague-Dawley
7.
PLoS One ; 13(6): e0199639, 2018.
Article in English | MEDLINE | ID: mdl-29949628

ABSTRACT

The use of the tellurium-centered Anderson-Evans polyoxotungstate [TeW6O24]6- (TEW) as a crystallization additive has been described. Here, we present the use of TEW as an additive in the crystallization screening of the nucleotide binding domain (NBD) of HSP70. Crystallization screening of the HSP70 NBD in the absence of TEW using a standard commercial screen resulted in a single crystal form. An identical crystallization screen of the HSP70 NBD in the presence of TEW resulted in both the "TEW free" crystal form and an additional crystal form with a different crystal packing. TEW binding was observed in both crystal forms, either as a well-defined molecule or in overlapping alternate positions suggesting translational disorder. The structures were solved by both molecular replacement and single wavelength anomalous diffraction (SAD) using the anomalous signal of a single bound molecule of TEW. This study adds one more example of TEW binding to a protein and influencing its crystallization behavior.


Subject(s)
HSP70 Heat-Shock Proteins/chemistry , Binding Sites , Crystallization , Escherichia coli , Fluorometry , Molecular Structure , Protein Stability , Protein Unfolding , Temperature
8.
ACS Med Chem Lett ; 9(5): 490-495, 2018 May 10.
Article in English | MEDLINE | ID: mdl-29795765

ABSTRACT

Complement Factor D, a serine protease of the S1 family and key component of the alternative pathway amplification loop, represents a promising target for the treatment of several prevalent and rare diseases linked to the innate immune system. Previously reported FD inhibitors have been shown to bind to the FD active site in its self-inhibited conformation characterized by the presence of a salt bridge at the bottom of the S1 pocket between Asp189 and Arg218. We report herein a new set of small-molecule FD ligands that harbor a basic S1 binding moiety directly binding to the carboxylate of Asp189, thereby displacing the Asp189-Arg218 ionic interaction and significantly changing the conformation of the self-inhibitory loop.

9.
J Med Chem ; 60(13): 5717-5735, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28621538

ABSTRACT

The highly specific S1 serine protease factor D (FD) plays a central role in the amplification of the complement alternative pathway (AP) of the innate immune system. Genetic associations in humans have implicated AP activation in age-related macular degeneration (AMD), and AP dysfunction predisposes individuals to disorders such as paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS). The combination of structure-based hit identification and subsequent optimization of the center (S)-proline-based lead 7 has led to the discovery of noncovalent reversible and selective human factor D (FD) inhibitors with drug-like properties. The orally bioavailable compound 2 exerted excellent potency in 50% human whole blood in vitro and blocked AP activity ex vivo after oral administration to monkeys as demonstrated by inhibition of membrane attack complex (MAC) formation. Inhibitor 2 demonstrated sustained oral and ocular efficacy in a model of lipopolysaccharide (LPS)-induced systemic AP activation in mice expressing human FD.


Subject(s)
Complement Factor D/antagonists & inhibitors , Complement Pathway, Alternative/drug effects , Proline/analogs & derivatives , Proline/pharmacology , Administration, Oral , Animals , Atypical Hemolytic Uremic Syndrome/drug therapy , Atypical Hemolytic Uremic Syndrome/immunology , Complement Factor D/immunology , Complement Membrane Attack Complex/antagonists & inhibitors , Complement Membrane Attack Complex/immunology , Female , Haplorhini , Humans , Macaca fascicularis , Macular Degeneration/drug therapy , Macular Degeneration/immunology , Male , Mice , Proline/administration & dosage , Proline/pharmacokinetics
10.
J Med Chem ; 60(5): 1946-1958, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28157311

ABSTRACT

Chronic dysregulation of alternative complement pathway activation has been associated with diverse clinical disorders including age-related macular degeneration and paroxysmal nocturnal hemoglobinurea. Factor D is a trypsin-like serine protease with a narrow specificity for arginine in the P1 position, which catalyzes the first enzymatic reaction of the amplification loop of the alternative pathway. In this article, we describe two hit finding approaches leading to the discovery of new chemical matter for this pivotal protease of the complement system: in silico active site mapping for hot spot identification to guide rational structure-based design and NMR screening of focused and diverse fragment libraries. The wealth of information gathered by these complementary approaches enabled the identification of ligands binding to different subpockets of the latent Factor D conformation and was instrumental for understanding the binding requirements for the generation of the first known potent noncovalent reversible Factor D inhibitors.


Subject(s)
Protease Inhibitors/pharmacology , Catalytic Domain , Complement Factor D/chemistry , Drug Design , Humans , Magnetic Resonance Spectroscopy , Molecular Structure , Protease Inhibitors/chemistry
11.
Nat Chem Biol ; 12(12): 1105-1110, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27775713

ABSTRACT

Complement is a key component of the innate immune system, recognizing pathogens and promoting their elimination. Complement component 3 (C3) is the central component of the system. Activation of C3 can be initiated by three distinct routes-the classical, the lectin and the alternative pathways-with the alternative pathway also acting as an amplification loop for the other two pathways. The protease factor D (FD) is essential for this amplification process, which, when dysregulated, predisposes individuals to diverse disorders including age-related macular degeneration and paroxysmal nocturnal hemoglobinuria (PNH). Here we describe the identification of potent and selective small-molecule inhibitors of FD. These inhibitors efficiently block alternative pathway (AP) activation and prevent both C3 deposition onto, and lysis of, PNH erythrocytes. Their oral administration inhibited lipopolysaccharide-induced AP activation in FD-humanized mice. These data demonstrate the feasibility of inhibiting the AP with small-molecule antagonists and support the development of FD inhibitors for the treatment of complement-mediated diseases.


Subject(s)
Complement Factor D/antagonists & inhibitors , Complement Pathway, Alternative/drug effects , Enzyme Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Animals , Complement Factor D/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 25(3): 438-43, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25571794

ABSTRACT

Adenoviral infections are associated with a wide range of acute diseases, among which ocular viral conjunctivitis (EKC) and disseminated disease in immunocompromised patients. To date, no approved specific anti-adenoviral drug is available, but there is a growing need for an effective treatment of such infections. The adenoviral protease, adenain, plays a crucial role for the viral lifecycle and thus represents an attractive therapeutic target. Structure-guided design with the objective to depeptidize tetrapeptide nitrile 1 led to the novel chemotype 2. Optimization of scaffold 2 resulted in picomolar adenain inhibitors 3a and 3b. In addition, a complementary series of irreversible vinyl sulfone containing inhibitors were rationally designed, prepared and evaluated against adenoviral protease. High resolution X-ray co-crystal structures of representatives of each series proves the successful design of these inhibitors and provides an excellent basis for future medicinal chemistry optimization of these compounds.


Subject(s)
Adenoviridae/enzymology , Antiviral Agents/chemistry , Cysteine Endopeptidases/chemistry , Drug Design , Protease Inhibitors/chemistry , Viral Proteins/antagonists & inhibitors , Adenoviridae/drug effects , Antiviral Agents/metabolism , Antiviral Agents/toxicity , Binding Sites , Crystallography, X-Ray , Cysteine Endopeptidases/metabolism , HEK293 Cells , Humans , Molecular Docking Simulation , Protease Inhibitors/metabolism , Protease Inhibitors/toxicity , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship , Viral Proteins/metabolism
13.
ACS Med Chem Lett ; 5(8): 937-41, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25147618

ABSTRACT

The cysteine protease adenain is the essential protease of adenovirus and, as such, represents a promising target for the treatment of ocular and other adenoviral infections. Through a concise two-pronged hit discovery approach we identified tetrapeptide nitrile 1 and pyrimidine nitrile 2 as complementary starting points for adenain inhibition. These hits enabled the first high-resolution X-ray cocrystal structures of adenain with inhibitors bound and revealed the binding mode of 1 and 2. The screening hits were optimized by a structure-guided medicinal chemistry strategy into low nanomolar drug-like inhibitors of adenain.

14.
J Mol Biol ; 345(1): 115-27, 2005 Jan 07.
Article in English | MEDLINE | ID: mdl-15567415

ABSTRACT

The key enzyme in the non-mevalonate pathway of isoprenoid biosynthesis, 1-deoxy-D-xylulose 5-phosphate reductoisomerase (DXR) has been shown to be the target enzyme of fosmidomycin, an antimalarial, antibacterial and herbicidal compound. Here we report the crystal structure of selenomethionine-labelled Escherichia coli DXR in a ternary complex with NADPH and fosmidomycin at 2.2 A resolution. The structure reveals a considerable conformational rearrangement upon fosmidomycin binding and provides insights into the slow, tight binding inhibition mode of the inhibitor. Although the inhibitor displays an unusual non-metal mediated mode of inhibition, which is an artefact most likely due to the low metal affinity of DXR at the pH used for crystallization, the structural data add valuable information for the rational design of novel DXR inhibitors. Using this structure together with the published structural data and the 1.9 A crystal structure of DXR in a ternary complex with NADPH and the substrate 1-deoxy-D-xylulose 5-phosphate, a model for the physiologically relevant tight-binding mode of inhibition is proposed. The structure of the substrate complex must be interpreted with caution due to the presence of a second diastereomer in the active site.


Subject(s)
Aldose-Ketose Isomerases/chemistry , Antimalarials/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Fosfomycin/analogs & derivatives , Fosfomycin/chemistry , Multienzyme Complexes/chemistry , NADP/chemistry , Oxidoreductases/chemistry , Protein Conformation , Aldose-Ketose Isomerases/genetics , Aldose-Ketose Isomerases/metabolism , Antimalarials/metabolism , Crystallography, X-Ray , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fosfomycin/metabolism , Macromolecular Substances , Models, Molecular , Molecular Structure , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , NADP/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Protein Binding
15.
J Med Chem ; 47(6): 1325-8, 2004 Mar 11.
Article in English | MEDLINE | ID: mdl-14998322

ABSTRACT

High-resolution crystal structures of Staphylococcus aureus methionine aminopeptidase I in complex with various keto heterocycles and aminoketones were determined, and the intermolecular ligand interactions with the enzyme are reported. The compounds are effective inhibitors of the S. aureus enzyme because of the formation of an uncleavable tetrahedral intermediate upon binding. The electron densities unequivocally show the enzyme-catalyzed transition-state analogue mimicking that for amide bond hydrolysis of substrates.


Subject(s)
Amines/chemistry , Aminopeptidases/antagonists & inhibitors , Aminopeptidases/chemistry , Ketones/chemistry , Pyridines/chemistry , Staphylococcus aureus/enzymology , Thiazoles/chemistry , Binding Sites , Crystallography, X-Ray , Cyclopropanes/chemistry , Methionyl Aminopeptidases , Models, Molecular , Molecular Structure
16.
J Synchrotron Radiat ; 11(Pt 1): 24-6, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14646125

ABSTRACT

The formation of nuclei in a crystallization experiment requires the interaction of protein molecules until a critical size of aggregate is created. In many crystallization screens sufficiently high levels of saturation are never reached to allow this critical nucleation event to occur. There are at least two possibilities to change this situation. The first is to increase the concentration of the protein and precipitating agent during the experiment to levels where spontaneous nucleation will occur. The second is to influence the nucleation event so that crystals can form at lower concentrations. The use of a modified microbatch method has made the first strategy possible and the use of heterogeneous seeding can be used to influence the second.


Subject(s)
Crystallization/methods , Crystallography/methods , Microchemistry/methods , Proteins/chemistry , Solvents/chemistry , Crystallization/instrumentation , Dimerization , Macromolecular Substances , Protein Binding , Protein Conformation , Proteins/chemical synthesis
17.
J Mol Biol ; 332(1): 13-21, 2003 Sep 05.
Article in English | MEDLINE | ID: mdl-12946343

ABSTRACT

Methionyl aminopeptidases (MetAPs) represent a unique class of protease that are responsible for removing the N-terminal methionine residue from proteins and peptides. There are two major classes of MetAPs (type I and type II) described and each class can be subdivided into two subclasses. Eukaryotes contain both the type I and type II MetAPs, whereas prokaryotes possess only the type I enzyme. Due to the physiological importance of these enzymes there is considerable interest in inhibitors to be used as antiangiogenic and antimicrobial agents. Here, we describe the 1.15A crystal structure of the Staphylococcus aureus MetAP-I as an apo-enzyme and its complexes with various 1,2,4-triazole-based derivatives at high-resolution. The protein has a typical "pita-bread" fold as observed for the other MetAP structures. The inhibitors bind in the active site with the N1 and N2 atoms of the triazole moiety complexing two divalent ions. The 1,2,4-triazols represent a novel class of potent non-peptidic inhibitors for the MetAP-Is.


Subject(s)
Aminopeptidases/chemistry , Enzyme Inhibitors/chemistry , Staphylococcus aureus/enzymology , Triazoles/chemistry , Amino Acid Sequence , Aminopeptidases/genetics , Aminopeptidases/metabolism , Apoenzymes , Binding Sites , Crystallography, X-Ray , Humans , Macromolecular Substances , Methionyl Aminopeptidases , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Conformation , Protein Folding , Protein Structure, Tertiary , Sequence Alignment
18.
Acta Crystallogr D Biol Crystallogr ; 59(Pt 7): 1206-12, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12832764

ABSTRACT

Dipeptidyl peptidase IV is a multifunctional type II transmembrane serine protease glycoprotein. The high-resolution crystal structure of the homodimeric human apo dipeptidyl peptidase IV has been determined at 1.9 A resolution. In addition, the structure of the binary complex with 1-[([2-[(5-iodopyridin-2-yl)amino]-ethyl]amino)-acetyl]-2-cyano-(S)-pyrrolidine has been solved, revealing the nature of the covalent interaction with the active-site serine.


Subject(s)
Aminopyridines/chemistry , Dipeptidyl Peptidase 4/chemistry , Pyrrolidines/chemistry , Apoenzymes/chemistry , Binding Sites , Crystallization , Crystallography, X-Ray , Dipeptidyl Peptidase 4/isolation & purification , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Protein Binding , Protein Conformation
19.
Acta Crystallogr D Biol Crystallogr ; 59(Pt 7): 1343-6, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12832806

ABSTRACT

The nucleation event in protein crystallization is a part of the process that is poorly controlled. It is generally accepted that the protein should be in the metastable phase for crystal growth, but for nucleation higher levels of saturation are needed. Formation of nuclei in bulk solvent requires interaction of protein molecules until a critical size of aggregate is created. In many crystallization experiments sufficiently high levels of saturation are not reached to allow this critical nucleation event to occur. If an environment can be created that favours a higher local concentration of macromolecules, the energy barrier for nucleation may be lowered. When seeds are introduced at lower levels of saturation in a crystallization experiment, nucleation may be facilitated and crystal growth initiated. In this study, the use of natural materials as stable seeds for nucleation has been investigated. The method makes it possible to introduce seeds into crystallization trials at any stage of the experiment using both microbatch and vapour-diffusion methods.


Subject(s)
Crystallization/methods , Proteins/chemistry , Aldose-Ketose Isomerases/chemistry , Hair , Muramidase/chemistry , Trypsin/chemistry
20.
Mol Cell ; 11(2): 543-9, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12620240

ABSTRACT

In the death receptor induced apoptotic pathway, caspase-8 autocatalytically cleaves itself at specific cleavage sites. To better understand the regulatory mechanisms behind caspase-8 activation, we compared active wild-type caspase-8 (wtC8) and an uncleavable form of procaspase-8 (uncleavable C8). We demonstrate that wtC8 predominantly exists as a monomer and dimerizes in a concentration and inhibitor binding-dependent fashion. The K(D) for dimeric wtC8 is approximately 50 micro M and decreases when inhibitor bound. Uncleavable C8 is mainly monomeric, but a small amount that dimerizes is as active as wtC8. Inhibitor binding does not favor dimerization but induces active site rearrangements in uncleavable C8. Our findings suggest that dimerization is the crucial factor for caspase-8 activation.


Subject(s)
Caspases/metabolism , Amino Acid Chloromethyl Ketones/metabolism , Caspase 8 , Caspase 9 , Caspases/chemistry , Caspases/genetics , Catalytic Domain , Cysteine Proteinase Inhibitors/metabolism , Dimerization , Enzyme Activation , In Vitro Techniques , Kinetics , Models, Biological , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL