Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Article in English | MEDLINE | ID: mdl-28584149

ABSTRACT

Cadazolid (CDZ) is a new antibiotic currently in clinical development for the treatment of Clostridium difficile infections. CDZ interferes with the bacterial protein synthesis machinery. The aim of the present study was to identify resistance mechanisms for CDZ and compare the results to those obtained for linezolid (LZD) in C. difficile by whole-genome sequencing (WGS) of strains generated by in vitro passages and to those obtained for LZD-resistant clinical isolates. Clones of C. difficile 630 selected with CDZ during 46 passages had a maximally 4-fold increase in CDZ MIC, while the LZD MIC for clones selected with LZD increased up to 16-fold. CDZ cross-resistance with LZD was maximally 4-fold, and no cross-resistance with other antibiotics tested was observed. Our data suggest that there are different resistance mechanisms for CDZ and LZD in C. difficile Mutations after passages with CDZ were found in rplD (ribosomal protein L4) as well as in tra and rmt, whereas similar experiments with LZD showed mutations in rplC (ribosomal protein L3), reg, and tpr, indicating different resistance mechanisms. Although high degrees of variation between the sequenced genomes of the clinical isolates were observed, the same mutation in rplC was found in two clinical isolates with high LZD MICs. No mutations were found in the 23S rRNA genes, and attempts to isolate the cfr gene from resistant clinical isolates were unsuccessful. Analysis of 50% inhibitory concentrations (IC50s) determined in in vitro transcription/translation assays performed with C. difficile cell extracts from passaged clones correlated well with the MIC values for all antibiotics tested, indicating that the ribosomal mutations are causing the resistant phenotype.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , Drug Resistance, Bacterial/genetics , Linezolid/pharmacology , Oxazolidinones/pharmacology , Protein Synthesis Inhibitors/pharmacology , Ribosomal Proteins/genetics , Amino Acid Sequence , Clostridioides difficile/isolation & purification , Drug Resistance, Bacterial/physiology , Genome, Bacterial/genetics , Humans , Microbial Sensitivity Tests , RNA, Ribosomal, 23S/genetics , Ribosomal Protein L3 , Sequence Analysis, DNA
2.
Appl Microbiol Biotechnol ; 101(18): 6951-6968, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28770303

ABSTRACT

The potential of using a synthetic cardosin-based rennet in cheese manufacturing was recently demonstrated with the development and optimization of production of a recombinant form of cardosin B in Kluyveromyces lactis. With the goal of providing a more detailed characterization of this rennet, we herein evaluate the impact of the plant-specific insert (PSI) on cardosin B secretion in this yeast, and provide a thorough analysis of the specificity requirements as well as the biochemical and structural properties of the isolated recombinant protease. We demonstrate that the PSI domain can be substituted by different linker sequences without substantially affecting protein secretion and milk clotting activity. However, the presence of small portions of the PSI results in dramatic reductions of secretion yields in this heterologous system. Kinetic characterization and specificity profiling results clearly suggest that synthetic cardosin B displays lower catalytic efficiency and is more sequence selective than native cardosin B. Elucidation of the structure of synthetic cardosin B confirms the canonical fold of an aspartic protease with the presence of two high mannose-type, N-linked glycan structures; however, there are some differences in the conformation of the flap region when compared to cardosin A. These subtle variations in catalytic properties and the more stringent substrate specificity of synthetic cardosin B help to explain the observed suitability of this rennet for cheese production.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Chymosin/metabolism , Kluyveromyces/metabolism , Plants/enzymology , Animals , Aspartic Acid Endopeptidases/genetics , Cheese , Chymosin/genetics , Glycosylation , Kluyveromyces/genetics , Milk/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Plants/genetics , Protein Transport
3.
Hum Mol Genet ; 20(2): 301-11, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21036942

ABSTRACT

Renal tubular dysgenesis (RTD) is a recessive autosomal disease characterized by persistent fetal anuria and perinatal death. During the systematic screening of mutations of the different genes of the renin-angiotensin system associated with RTD, two missense mutations in the renin gene were previously identified, the first affects one of the two catalytic aspartates (D38N) of renin, and the second, S69Y, is located upstream of the 'flap', a mobile ß-hairpin structure which covers the substrate-binding site of renin. Here we report a novel renin mutation leading to the duplication of the tyrosine residue Y15dup, homologous to Y9 in some other aspartyl proteases, which seems to play a crucial role along the activation pathway. The biochemical and cellular mechanisms underlying renin inactivation were investigated. We expressed prorenin constructs harboring the identified point mutations in two established cell lines, able (AtT-20 cells) or unable (CHO cells) to process prorenin to renin and we evaluated the cellular localization of renin mutants and their functional properties. All three mutants were misfolded to different levels, were enzymatically inactive and exhibited abnormal intracellular trafficking. We suggest a misfolding of Y15dup renin, a partial misfolding of D38N prorenin and a misfolding of S69Y prorenin leading to complete absence of secretion. The structural consequences of the renin mutations were estimated by molecular modeling, which suggested some important structural alterations. This is the first characterization of the mechanisms underlying loss of renin function in RTD.


Subject(s)
Point Mutation , Protein Transport , Renin/genetics , Renin/metabolism , Urogenital Abnormalities/genetics , Urogenital Abnormalities/pathology , Amino Acid Sequence , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Female , Humans , Kidney Tubules, Proximal/abnormalities , Kidney Tubules, Proximal/pathology , Models, Molecular , Molecular Sequence Data , Pregnancy , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Renin/analysis , Renin/chemistry , Sequence Alignment
4.
Chemistry ; 19(1): 155-64, 2013 Jan 02.
Article in English | MEDLINE | ID: mdl-23161835

ABSTRACT

The increasing prevalence of multidrug-resistant strains of the malarial parasite Plasmodium falciparum requires the urgent development of new therapeutic agents with novel modes of action. The vacuolar malarial aspartic proteases plasmepsin (PM) I, II, and IV are involved in hemoglobin degradation and play a central role in the growth and maturation of the parasite in the human host. We report the structure-based design, synthesis, and in vitro evaluation of a new generation of PM inhibitors featuring a highly decorated 7-azabicyclo[2.2.1]heptane core. While this protonated central core addresses the catalytic Asp dyad, three substituents bind to the flap, the S1/S3, and the S1' pockets of the enzymes. A hydroformylation reaction is the key synthetic step for the introduction of the new vector reaching into the S1' pocket. The configuration of the racemic ligands was confirmed by extensive NMR and X-ray crystallographic analysis. In vitro biological assays revealed high potency of the new inhibitors against the three plasmepsins (IC(50) values down to 6 nM) and good selectivity towards the closely related human cathepsins D and E. The occupancy of the S1' pocket makes an essential contribution to the gain in binding affinity and selectivity, which is particularly large in the case of the PM IV enzyme. Designing non-peptidic ligands for PM II is a valid route to generate compounds that inhibit the entire family of vacuolar plasmepsins.


Subject(s)
Antimalarials/chemistry , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aza Compounds/chemical synthesis , Bridged Bicyclo Compounds/chemical synthesis , Formaldehyde/chemistry , Heptanes/chemical synthesis , Plasmodium falciparum/enzymology , Protease Inhibitors/chemistry , Antimalarials/chemical synthesis , Antimalarials/metabolism , Antimalarials/pharmacology , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/metabolism , Aza Compounds/chemistry , Aza Compounds/pharmacology , Bridged Bicyclo Compounds/chemistry , Bridged Bicyclo Compounds/pharmacology , Heptanes/chemistry , Heptanes/pharmacology , Humans , Models, Molecular , Plasmodium falciparum/drug effects , Protease Inhibitors/chemical synthesis , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacology , Stereoisomerism
5.
Biochem J ; 443(3): 769-78, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22332999

ABSTRACT

TTR (transthyretin) was found recently to possess proteolytic competency besides its well-known transport capabilities. It was described as a cryptic serine peptidase cleaving multiple natural substrates (including ß-amyloid and apolipoprotein A-I) involved in diseases such as Alzheimer's disease and atherosclerosis. In the present study, we aimed to elucidate the catalytic machinery of TTR. All attempts to identify a catalytic serine residue were unsuccessful. However, metal chelators abolished TTR activity. Proteolytic inhibition by EDTA or 1,10-phenanthroline could be reversed with Zn2+ and Mn2+. These observations, supported by analysis of three-dimensional structures of TTR complexed with Zn2+, led to the hypothesis that TTR is a metallopeptidase. Site-directed mutagenesis of selected amino acids unambiguously confirmed this hypothesis. The TTR active site is inducible and constituted via a protein rearrangement resulting in ~7% of proteolytically active TTR at pH 7.4. The side chain of His88 is shifted near His90 and Glu92 establishing a Zn2+-chelating pattern HXHXE not found previously in any metallopeptidase and only conserved in TTR of humans and some other primates. Point mutations of these three residues yielded proteins devoid of proteolytic activity. Glu72 was identified as the general base involved in activation of the catalytic water. Our results unveil TTR as a metallopeptidase and define its catalytic machinery.


Subject(s)
Metalloproteases/metabolism , Prealbumin/metabolism , Catalytic Domain , Chromatography, Gel , Hydrogen-Ion Concentration , Kinetics , Prealbumin/chemistry , Protein Conformation , Proteolysis
6.
Bioorg Med Chem Lett ; 22(21): 6705-11, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23006603

ABSTRACT

A series of 2-amino-[1,8]-naphthyridine-3-carboxamides (ANCs) with potent inhibition of bacterial NAD(+)-dependent DNA ligases (LigAs) evolved from a 2,4-diaminopteridine derivative discovered by HTS. The design was guided by several highly resolved X-ray structures of our inhibitors in complex with either Streptococcus pneumoniae or Escherichia coli LigA. The structure-activity-relationship based on the ANC scaffold is discussed. The in-depth characterization of 2-amino-6-bromo-7-(trifluoromethyl)-[1,8]-naphthyridine-3-carboxamide, which displayed promising in vitro (MIC Staphylococcus aureus 1 mg/L) and in vivo anti-staphylococcal activity, is presented.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , DNA Ligases/antagonists & inhibitors , Drug Design , Staphylococcus/drug effects , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Crystallography, X-Ray , DNA, Bacterial/antagonists & inhibitors , Inhibitory Concentration 50 , Mice , Microbial Sensitivity Tests , Molecular Structure , Rats , Staphylococcal Infections/drug therapy , Structure-Activity Relationship
7.
Bioorg Med Chem Lett ; 20(21): 6286-90, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20843686

ABSTRACT

The discovery of a new series of piperidine-based renin inhibitors is described herein. SAR optimization upon the P3 renin sub-pocket is described, leading to the discovery of 9 and 41, two bioavailable renin inhibitors orally active at low doses in a transgenic rat model of hypertension.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Piperidines/chemical synthesis , Piperidines/pharmacology , Renin/antagonists & inhibitors , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Drug Design , Models, Molecular , Piperidines/chemistry , Protein Conformation , Rats , Structure-Activity Relationship , X-Ray Diffraction
8.
Bioorg Med Chem Lett ; 20(21): 6291-6, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20843690

ABSTRACT

The optimization of the 4-position of recently described new 3,4-disubstituted piperidine-based renin inhibitors is reported herein. The synthesis and characterization of compounds leading to the discovery of 11 (ACT-178882, MK-1597), a renin inhibitor with a suitable profile for development is described.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Piperidines/chemical synthesis , Piperidines/pharmacology , Renin/antagonists & inhibitors , Angiotensinogen/genetics , Animals , Animals, Genetically Modified , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/chemistry , Humans , Indicators and Reagents , Models, Molecular , Piperidines/chemistry , Rats , Renin/genetics , Stereoisomerism , Structure-Activity Relationship
10.
Org Biomol Chem ; 7(19): 3947-57, 2009 Oct 07.
Article in English | MEDLINE | ID: mdl-19763297

ABSTRACT

The development of new therapeutic agents against malaria has become urgent during the past few decades, due to an increased prevalence of drug-resistant strains of malaria-causing Plasmodium parasites. Possible targets are the hemoglobin-degrading aspartic proteases, the plasmepsins. While acyclic alpha,alpha-difluoroketone hydrates have been introduced into peptidomimetics to bind to the catalytic Asp dyad of aspartic proteases, alicyclic derivatives were unknown. This paper describes a versatile synthesis of hydrated alicyclic alpha,alpha-difluoro-cyclopentanones and -cyclohexanones, decorated with appropriate substituents to fill the S1/S3 and the "flap-open" pocket at the enzyme active sites. Their biological activity was tested against plasmepsin II and IV, revealing an IC(50) value (concentration of an inhibitor at which 50% maximum initial velocity is observed) of 7 microM for the best ligand. Reference inhibitors with a protonated secondary ammonium centre to address the catalytic dyad showed similar binding affinities. The X-ray crystal structure of a cyclic alpha,alpha-difluoroketone hydrate revealed the ability of these novel building blocks to participate in H-bonding networks. The hydration of difluoroketones was also investigated in solution. An exemplary study showed that the equilibrium constants for the hydration of alpha,alpha-difluorinated cyclohexanones are much higher than those for the corresponding cyclopentanones.


Subject(s)
Aspartic Acid Endopeptidases/antagonists & inhibitors , Fluorine/chemistry , Ketones/chemistry , Ketones/pharmacology , Malaria/enzymology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Antimalarials/chemical synthesis , Antimalarials/chemistry , Antimalarials/metabolism , Antimalarials/pharmacology , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/metabolism , Crystallography, X-Ray , Humans , Ketones/chemical synthesis , Ketones/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Protease Inhibitors/chemical synthesis , Protease Inhibitors/metabolism , Water/chemistry
11.
Sci Rep ; 9(1): 5634, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30948752

ABSTRACT

Oxazolidinones are synthetic antibiotics used for treatment of infections caused by Gram-positive bacteria. They target the bacterial protein synthesis machinery by binding to the peptidyl transferase centre (PTC) of the ribosome and interfering with the peptidyl transferase reaction. Cadazolid is the first member of quinoxolidinone antibiotics, which are characterized by combining the pharmacophores of oxazolidinones and fluoroquinolones, and it is evaluated for treatment of Clostridium difficile gastrointestinal infections that frequently occur in hospitalized patients. In vitro protein synthesis inhibition by cadazolid was shown in Escherichia coli and Staphylococcus aureus, including an isolate resistant against linezolid, the prototypical oxazolidinone antibiotic. To better understand the mechanism of inhibition, we determined a 3.0 Å cryo-electron microscopy structure of cadazolid bound to the E. coli ribosome in complex with mRNA and initiator tRNA. Here we show that cadazolid binds with its oxazolidinone moiety in a binding pocket in close vicinity of the PTC as observed previously for linezolid, and that it extends its unique fluoroquinolone moiety towards the A-site of the PTC. In this position, the drug inhibits protein synthesis by interfering with the binding of tRNA to the A-site, suggesting that its chemical features also can enable the inhibition of linezolid-resistant strains.


Subject(s)
Oxazolidinones/metabolism , Oxazolidinones/pharmacology , Protein Synthesis Inhibitors/pharmacology , Acetamides/pharmacology , Anti-Bacterial Agents/pharmacology , Clostridium Infections/drug therapy , Cryoelectron Microscopy/methods , Escherichia coli/metabolism , Fluoroquinolones/pharmacology , Humans , Microbial Sensitivity Tests , Peptidyl Transferases/antagonists & inhibitors , RNA, Transfer, Met/metabolism , Ribosomes/metabolism , Staphylococcus aureus/metabolism
12.
FEBS J ; 274(14): 3685-3694, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17608726

ABSTRACT

Yeast IA(3) aspartic proteinase inhibitor operates through an unprecedented mechanism and exhibits a remarkable specificity for one target enzyme, saccharopepsin. Even aspartic proteinases that are very closely similar to saccharopepsin (e.g. the vacuolar enzyme from Pichia pastoris) are not susceptible to significant inhibition. The Pichia proteinase was selected as the target for initial attempts to engineer IA(3) to re-design the specificity. The IA(3) polypeptides from Saccharomyces cerevisiae and Saccharomyces castellii differ considerably in sequence. Alterations made by deletion or exchange of the residues in the C-terminal segment of these polypeptides had only minor effects. By contrast, extension of each of these wild-type and chimaeric polypeptides at its N-terminus by an MK(H)(7)MQ sequence generated inhibitors that displayed subnanomolar potency towards the Pichia enzyme. This gain-in-function was completely reversed upon removal of the extension sequence by exopeptidase trimming. Capture of the potentially positively charged aromatic histidine residues of the extension by remote, negatively charged side-chains, which were identified in the Pichia enzyme by modelling, may increase the local IA(3) concentration and create an anchor that enables the N-terminal segment residues to be harboured in closer proximity to the enzyme active site, thus promoting their interaction. In saccharopepsin, some of the counterpart residues are different and, consistent with this, the N-terminal extension of each IA(3) polypeptide was without major effect on the potency of interaction with saccharopepsin. In this way, it is possible to convert IA(3) polypeptides that display little affinity for the Pichia enzyme into potent inhibitors of this proteinase and thus broaden the target selectivity of this remarkable small protein.


Subject(s)
Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Alternative Splicing/genetics , Amino Acid Sequence , Antigens, Protozoan , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/metabolism , Hydrogen-Ion Concentration , Membrane Proteins , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Protozoan Proteins , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sensitivity and Specificity , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
13.
J Med Chem ; 60(9): 3755-3775, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28406299

ABSTRACT

Our strategy to combat resistant bacteria consisted of targeting the GyrB/ParE ATP-binding sites located on bacterial DNA gyrase and topoisomerase IV and not utilized by marketed antibiotics. Screening around the minimal ethyl urea binding motif led to the identification of isoquinoline ethyl urea 13 as a promising starting point for fragment evolution. The optimization was guided by structure-based design and focused on antibacterial activity in vitro and in vivo, culminating in the discovery of unprecedented substituents able to interact with conserved residues within the ATP-binding site. A detailed characterization of the lead compound highlighted the potential for treatment of the problematic fluoroquinolone-resistant MRSA, VRE, and S. pneumoniae, and the possibility to offer patients an intravenous-to-oral switch therapy was supported by the identification of a suitable prodrug concept. Eventually, hERG K-channel block was identified as the main limitation of this chemical series, and efforts toward its minimization are reported.


Subject(s)
Anti-Bacterial Agents/pharmacology , Isoquinolines/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Area Under Curve , Drug Discovery , Gram-Negative Bacteria/drug effects , Half-Life , Hydrogen Bonding , Isoquinolines/chemistry , Isoquinolines/pharmacokinetics , Isoquinolines/therapeutic use , Microbial Sensitivity Tests , Potassium Channels/drug effects , Rats , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/microbiology , Solubility , Urea/chemistry
14.
J Med Chem ; 60(9): 3776-3794, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28406300

ABSTRACT

There is an urgent unmet medical need for novel antibiotics that are effective against a broad range of bacterial species, especially multidrug resistant ones. Tetrahydropyran-based inhibitors of bacterial type II topoisomerases (DNA gyrase and topoisomerase IV) display potent activity against Gram-positive pathogens and no target-mediated cross-resistance with fluoroquinolones. We report our research efforts aimed at expanding the antibacterial spectrum of this class of molecules toward difficult-to-treat Gram-negative pathogens. Physicochemical properties (polarity and basicity) were considered to guide the design process. Dibasic tetrahydropyran-based compounds such as 6 and 21 are potent inhibitors of both DNA gyrase and topoisomerase IV, displaying antibacterial activities against Gram-positive and Gram-negative pathogens (Staphylococcus aureus, Enterobacteriaceae, Pseudomonas aeruginosa, and Acinetobacter baumannii). Compounds 6 and 21 are efficacious in clinically relevant murine infection models.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria/drug effects , Pyrans/pharmacology , Topoisomerase Inhibitors/chemical synthesis , Topoisomerase Inhibitors/pharmacology , Animals , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/chemical synthesis , Guinea Pigs , Humans , Microbial Sensitivity Tests , Myocytes, Cardiac/drug effects , Pyrans/adverse effects , Pyrans/chemical synthesis , Topoisomerase Inhibitors/adverse effects
15.
Sci Rep ; 6: 23869, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27029611

ABSTRACT

The widespread presence of pepsin-like enzymes in eukaryotes together with their relevance in the control of multiple biological processes is reflected in the large number of studies published so far for this family of enzymes. By contrast, pepsin homologs from bacteria have only recently started to be characterized. The work with recombinant shewasin A from Shewanella amazonensis provided the first documentation of this activity in prokaryotes. Here we extend our studies to shewasin D, the pepsin homolog from Shewanella denitrificans, to gain further insight into this group of bacterial peptidases that likely represent ancestral versions of modern eukaryotic pepsin-like enzymes. We demonstrate that the enzymatic properties of recombinant shewasin D are strongly reminiscent of eukaryotic pepsin homologues. We determined the specificity preferences of both shewasin D and shewasin A using proteome-derived peptide libraries and observed remarkable similarities between both shewasins and eukaryotic pepsins, in particular with BACE-1, thereby confirming their phylogenetic proximity. Moreover, we provide first evidence of expression of active shewasin D in S. denitrificans cells, confirming its activity at acidic pH and inhibition by pepstatin. Finally, our results revealed an unprecedented localization for a family A1 member by demonstrating that native shewasin D accumulates preferentially in the cytoplasm.


Subject(s)
Bacterial Proteins/metabolism , Cytoplasm/enzymology , Pepsin A/metabolism , Shewanella/enzymology , Amino Acid Sequence , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biological Evolution , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Conserved Sequence , Cytoplasm/ultrastructure , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Hydrogen-Ion Concentration , Kinetics , Pepsin A/antagonists & inhibitors , Pepsin A/chemistry , Pepsin A/genetics , Pepstatins/pharmacology , Peptide Library , Proteolysis , Proteome/genetics , Proteome/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Shewanella/drug effects , Shewanella/genetics , Shewanella/ultrastructure , Substrate Specificity
16.
FEBS J ; 272(22): 5786-98, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16279943

ABSTRACT

Here we report the identification of phospholipase Dalpha as a cardosin A-binding protein. The interaction was confirmed by coimmunoprecipitation studies and pull-down assays. To investigate the structural and molecular determinants involved in the interaction, pull-down assays with cardosin A and various glutathione S-transferase-fused phospholipase Dalpha constructs were performed. Results revealed that the C2 domain of phospholipase Dalpha contains the cardosin A-binding activity. Further assays with mutated recombinant forms of cardosin A showed that the RGD motif as well as the unprecedented KGE motif, which is structurally and charge-wise very similar to RGD, are indispensable for the interaction. Taken together our results indicate that the C2 domain of plant phospholipase Dalpha can act as a cardosin A-binding domain and suggest that plant C2 domains may have an additional role as RGD/KGE-recognition domains.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Cynara/enzymology , Phospholipase D/metabolism , Plant Proteins/metabolism , Pollen/enzymology , Amino Acid Sequence , Base Sequence , Blotting, Western , Chromatography, Affinity , Cloning, Molecular , Conserved Sequence , Glutathione Transferase/metabolism , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Molecular Weight , Mutagenesis, Site-Directed , Mutation , Oligopeptides , Phospholipase D/chemistry , Phospholipase D/genetics , Phospholipase D/isolation & purification , Precipitin Tests , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid
17.
J Med Chem ; 58(2): 927-42, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25494934

ABSTRACT

Novel antibacterial drugs that are effective against infections caused by multidrug resistant pathogens are urgently needed. In a previous report, we have shown that tetrahydropyran-based inhibitors of bacterial type II topoisomerases (DNA gyrase and topoisomerase IV) display potent antibacterial activity and exhibit no target-mediated cross-resistance with fluoroquinolones. During the course of our optimization program, lead compound 5 was deprioritized due to adverse findings in cardiovascular safety studies. In the effort of mitigating these findings and optimizing further the pharmacological profile of this class of compounds, we have identified a subseries of tetrahydropyran-based molecules that are potent DNA gyrase and topoisomerase IV inhibitors and display excellent antibacterial activity against Gram positive pathogens, including clinically relevant resistant isolates. One representative of this class, compound 32d, elicited only weak inhibition of hERG K(+) channels and hNaV1.5 Na(+) channels, and no effects were observed on cardiovascular parameters in anesthetized guinea pigs. In vivo efficacy in animal infection models has been demonstrated against Staphylococcus aureus and Streptococcus pneumoniae strains.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Gram-Positive Bacteria/drug effects , Pyrans/chemical synthesis , Topoisomerase II Inhibitors/chemical synthesis , Animals , Anti-Bacterial Agents/pharmacology , Guinea Pigs , Hemodynamics/drug effects , Humans , Male , Mice , Microbial Sensitivity Tests , Pyrans/pharmacology , Rats , Rats, Wistar , Structure-Activity Relationship , Topoisomerase II Inhibitors/pharmacology
18.
J Med Chem ; 47(11): 2776-95, 2004 May 20.
Article in English | MEDLINE | ID: mdl-15139756

ABSTRACT

Since its discovery in 1988 by Yanagisawa et al., endothelin (ET), a potent vasoconstrictor, has been widely implicated in the pathophysiology of cardiovascular, cerebrovascular, and renal diseases. Many research groups have embarked on the discovery and development of ET receptor antagonists for the treatment of such diseases. While several compounds, e.g., ambrisentan 2, are in late clinical trials for various indications, one compound (bosentan, Tracleer) is being marketed to treat pulmonary arterial hypertension. Inspired by the structure of ambrisentan 2, we designed a novel class of ET receptor antagonists based on a 1,3,4,5-tetrahydro-1H-benzo[e][1,4]diazepin-2-one scaffold. Here, we report on the preparation as well as the in vitro and in vivo structure-activity relationships of these derivatives. Potent dual ET(A)/ET(B) receptor antagonists with affinities in the low nanomolar range have been identified. In addition, several compounds efficiently reduced arterial blood pressure after oral administration to Dahl salt sensitive rats. In this animal model, the efficacy of the benzo[e][1,4]diazepin-2-one derivative rac-39au was superior to that of racemic ambrisentan, rac-2.


Subject(s)
Benzodiazepines/chemical synthesis , Endothelin A Receptor Antagonists , Endothelin B Receptor Antagonists , Animals , Antihypertensive Agents/chemical synthesis , Antihypertensive Agents/chemistry , Antihypertensive Agents/pharmacology , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Biological Availability , Blood Pressure/drug effects , Cytochrome P-450 Enzyme Inhibitors , Hepatocytes/metabolism , Humans , In Vitro Techniques , Male , Microsomes, Liver/metabolism , Models, Molecular , Rats , Rats, Inbred Dahl , Rats, Wistar , Stereoisomerism , Structure-Activity Relationship
19.
Protein Pept Lett ; 10(1): 35-42, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12625824

ABSTRACT

The newly-discovered human aspartic proteinase, napsin A was not susceptible to protein inhibitors from potato, squash or yeast but was weakly inhibited by the 17 kDa polypeptide from Ascaris lumbricoides and potently by isovaleryl and lactoyl-pepstatins. A series of synthetic inhibitors was also investigated which contained in the P(1)-P(1)' positions the dipeptide analogue statine or its phenylalanine or cyclohexylalanine homologues and in which the residues occupying P(4)-P(3)' were varied systematically. On this basis, the active site of napsin A can be readily distinguished from other human aspartic proteinases.


Subject(s)
Aspartic Acid Endopeptidases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Amino Acid Sequence , Amino Acids/chemistry , Amino Acids/pharmacology , Animals , Ascaris lumbricoides/chemistry , Binding Sites , Cells, Cultured , Enzyme Inhibitors/chemistry , Humans , Kinetics , Models, Molecular , Pepstatins/chemistry , Pepstatins/pharmacology , Peptides/pharmacology , Recombinant Proteins/antagonists & inhibitors
20.
PLoS One ; 9(9): e107809, 2014.
Article in English | MEDLINE | ID: mdl-25226600

ABSTRACT

The competitive endothelin receptor antagonists (ERA) bosentan and ambrisentan, which have long been approved for the treatment of pulmonary arterial hypertension, are characterized by very short (1 min) occupancy half-lives at the ET(A) receptor. The novel ERA macitentan, displays a 20-fold increased receptor occupancy half-life, causing insurmountable antagonism of ET-1-induced signaling in pulmonary arterial smooth muscle cells. We show here that the slow ET(A) receptor dissociation rate of macitentan was shared with a set of structural analogs, whereas compounds structurally related to bosentan displayed fast dissociation kinetics. NMR analysis showed that macitentan adopts a compact structure in aqueous solution and molecular modeling suggests that this conformation tightly fits into a well-defined ET(A) receptor binding pocket. In contrast the structurally different and negatively charged bosentan-type molecules only partially filled this pocket and expanded into an extended endothelin binding site. To further investigate these different ET(A) receptor-antagonist interaction modes, we performed functional studies using ET(A) receptor variants harboring amino acid point mutations in the presumed ERA interaction site. Three ET(A) receptor residues significantly and differentially affected ERA activity: Mutation R326Q did not affect the antagonist activity of macitentan, however the potencies of bosentan and ambrisentan were significantly reduced; mutation L322A rendered macitentan less potent, whereas bosentan and ambrisentan were unaffected; mutation I355A significantly reduced bosentan potency, but not ambrisentan and macitentan potencies. This suggests that--in contrast to bosentan and ambrisentan--macitentan-ET(A) receptor binding is not dependent on strong charge-charge interactions, but depends predominantly on hydrophobic interactions. This different binding mode could be the reason for macitentan's sustained target occupancy and insurmountable antagonism.


Subject(s)
Endothelin Receptor Antagonists/metabolism , Pyrimidines/metabolism , Receptors, Endothelin/metabolism , Sulfonamides/metabolism , Binding Sites , Bosentan , Catalytic Domain , Cell Line , Endothelin Receptor Antagonists/chemistry , Endothelin Receptor Antagonists/pharmacology , Humans , Inhibitory Concentration 50 , Kinetics , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Conformation , Pyrimidines/chemistry , Pyrimidines/pharmacology , Receptors, Endothelin/chemistry , Receptors, Endothelin/genetics , Sulfonamides/chemistry , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL