Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Immunotherapy ; 12(1): 53-62, 2020 01.
Article in English | MEDLINE | ID: mdl-31910695

ABSTRACT

Aim: Evaluate the effectiveness and safety of immunotherapy with Acarovac Plus® in a 1-year prospective multicentered real-life study. Methods: A total of 118 adults with allergic rhinitis sensitized to Dermatophagoides received subcutaneous immunotherapy with Acarovac Plus. Treatment outcomes were evaluated at baseline, 6 months and 1 year after treatment initiation. Primary end point was the evolution of the combined symptom and medication score. Secondary end points included other effectiveness outcomes and measurement of product tolerability. Results: Acarovac Plus induced significant improvements in primary and secondary end points after 6 months compared with baseline. These differences persisted after 1 year of treatment (p < 0.001; baseline vs 1 year): combined symptom and medication score (1.60 vs 0.79). No serious adverse events were recorded. Conclusion: Acarovac Plus for 1 year was effective and well tolerated in a real-life setting.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antigens, Dermatophagoides/immunology , Desensitization, Immunologic/methods , Rhinitis, Allergic/therapy , Tyrosine/therapeutic use , Adolescent , Adult , Aged , Animals , Antigens, Dermatophagoides/therapeutic use , Female , Humans , Injections, Subcutaneous , Male , Middle Aged , Novobiocin/chemistry , Prospective Studies , Pyroglyphidae , Rhinitis, Allergic/immunology , Treatment Outcome , Tyrosine/analogs & derivatives , Tyrosine/chemistry , Young Adult
2.
J Recept Signal Transduct Res ; 40(1): 77-88, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31971048

ABSTRACT

Adenosine deaminase (ADA) is an enzyme present in purine metabolic pathway. Its inhibitors are considered to be potent drug lead compounds against inflammatory and malignant diseases. This study aimed to test ADA inhibitory activity of some Streptomyces secondary metabolites by using computational and in vitro methods. The in silico screening of the inhibitory properties has been carried out using pharmacophore modeling, docking, and molecular dynamics studies. The in vitro validation of the selected antibiotics has been carried out by enzyme kinetics and fluorescent spectroscopic studies. The results indicated that novobiocin, an aminocoumarin antibiotic from Streptomyces niveus, has significant inhibition on ADA activity. Hence, the antibiotic can be used as a lead compound for the development of potential ADA inhibitors.


Subject(s)
Adenosine Deaminase Inhibitors/pharmacology , Adenosine Deaminase/metabolism , Anti-Bacterial Agents/pharmacology , Drug Repositioning , Molecular Docking Simulation , Molecular Dynamics Simulation , Streptomyces/chemistry , Adenosine Deaminase Inhibitors/chemistry , Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Catalytic Domain , Drug Evaluation, Preclinical , Enzyme Assays , Humans , Least-Squares Analysis , Ligands , Novobiocin/chemistry , Novobiocin/pharmacology , Quantitative Structure-Activity Relationship , Spectrometry, Fluorescence
3.
Bioorg Med Chem Lett ; 21(19): 5697-700, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21871799

ABSTRACT

Synthetic derivatives of the natural product antibiotic novobiocin were synthesized in order to improve their physiochemical properties. A Mannich reaction was used to introduce new side chains at a solvent-exposed position of the molecule, and a diverse panel of functional groups was evaluated at this position. Novobiocin and the new derivatives were tested for their binding to gyrase B and their antibacterial activities against Staphylococcus aureus, Mycobacterium tuberculosis, Francisella tularensis and Escherichia coli. While the new derivatives still bound the gyrase B protein potently (0.07-1.8 µM, IC(50)), they had significantly less antibacterial activity. Two compounds were identified with increased antibacterial activity against M. tuberculosis, with a minimum inhibitory concentration of 2.5 µg/ml.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Drug Design , Drug Discovery , Mannich Bases/chemistry , Novobiocin/analogs & derivatives , Novobiocin/chemistry , Topoisomerase II Inhibitors , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Cell Membrane/metabolism , Drug Evaluation, Preclinical , Escherichia coli/drug effects , Escherichia coli/metabolism , Microbial Sensitivity Tests , Molecular Structure , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Novobiocin/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Structure-Activity Relationship
4.
Anal Biochem ; 365(1): 132-43, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17395144

ABSTRACT

The rise in bacterial resistance to antibiotics demonstrates the medical need for new antibacterial agents. One approach to this problem is to identify new antibacterials that act through validated drug targets such as bacterial DNA gyrase. DNA gyrase uses the energy of ATP hydrolysis to introduce negative supercoils into plasmid and chromosomal DNA and is essential for DNA replication. Inhibition of the ATPase activity of DNA gyrase is the mechanism by which coumarin-class antibiotics such as novobiocin inhibit bacterial growth. Although ATPase inhibitors exhibit potent antibacterial activity against gram-positive pathogens, no gyrase ATPase activity from a gram-positive organism is described in the literature. To address this, we developed and optimized an enzyme-coupled phosphate assay and used this assay to characterize the ATPase kinetics of Streptococcus pneumoniae gyrase. The S. pneumoniae enzyme exhibits cooperativity with ATP and requires organic potassium salts. We also studied inhibition of the enzyme by novobiocin. Apparent inhibition constants for novobiocin increased linearly with ATP concentration, indicative of an ATP-competitive mechanism. Similar binding affinities were measured by isothermal titration calorimetry. These results reveal unique features of the S. pneumoniae DNA gyrase ATPase and demonstrate the utility of the assay for screening and kinetic characterization of ATPase inhibitors.


Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Drug Evaluation, Preclinical/methods , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/enzymology , Topoisomerase II Inhibitors , Adenosine Triphosphatases/drug effects , Adenosine Triphosphatases/isolation & purification , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cloning, Molecular , Coumarins/chemistry , Coumarins/pharmacology , DNA Gyrase/drug effects , DNA Gyrase/isolation & purification , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Escherichia coli/metabolism , Kinetics , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Novobiocin/chemistry , Novobiocin/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/genetics
5.
J Biol Chem ; 281(11): 7161-71, 2006 Mar 17.
Article in English | MEDLINE | ID: mdl-16421106

ABSTRACT

The C-terminal domain of Hsp90 displays independent chaperone activity, mediates dimerization, and contains the MEEVD motif essential for interaction with tetratricopeptide repeat-containing immunophilin cochaperones assembled in mature steroid receptor complexes. An alpha-helical region, upstream of the MEEVD peptide, helps form the dimerization interface and includes a hydrophobic microdomain that contributes to the Hsp90 interaction with the immunophilin cochaperones and corresponds to the binding site for novobiocin, a coumarin-related Hsp90 inhibitor. Mutation of selected residues within the hydrophobic microdomain significantly impacted the chaperone function of a recombinant C-terminal Hsp90 fragment and novobiocin inhibited wild-type chaperone activity. Prior incubation of the Hsp90 fragment with novobiocin led to a direct blockade of immunophilin cochaperone binding. However, the drug had little influence on the pre-formed Hsp90-immunophilin complex, suggesting that bound cochaperones mask the novobiocin-binding site. We observed a differential effect of the drug on Hsp90-immunophilin interaction, suggesting that the immunophilins make distinct contacts within the C-terminal domain to specifically modulate Hsp90 function. Novobiocin also precluded the interaction of full-length Hsp90 with the p50(cdc37) cochaperone, which targets the N-terminal nucleotide-binding domain, and is prevalent in Hsp90 complexes with protein kinase substrates. Novobiocin therefore acts locally and allosterically to induce conformational changes within multiple regions of the Hsp90 protein. We provide evidence that coumermycin A1, a coumarin structurally related to novobiocin, interferes with dimerization of the Hsp90 C-terminal domain. Coumarin-based inhibitors then may antagonize Hsp90 function by inducing a conformation favoring separation of the C-terminal domains and release of substrate.


Subject(s)
Anti-Bacterial Agents/chemistry , Coumarins/chemistry , HSP90 Heat-Shock Proteins/chemistry , Molecular Chaperones/chemistry , Novobiocin/chemistry , Alanine/chemistry , Amino Acid Sequence , Aminocoumarins/chemistry , Binding Sites , DNA, Complementary/metabolism , Dimerization , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme-Linked Immunosorbent Assay , Glutathione Transferase/metabolism , HeLa Cells , Humans , Immunophilins/chemistry , Molecular Conformation , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Plasmids/metabolism , Point Mutation , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Steroids/chemistry , Substrate Specificity , Tacrolimus Binding Proteins/chemistry , Time Factors
6.
Chembiochem ; 6(5): 920-6, 2005 May.
Article in English | MEDLINE | ID: mdl-15812853

ABSTRACT

Topoisomerase II is the only enzyme able to cleave and religate double-stranded DNA; this makes it essential for many vital functions during normal cell growth. Increased expression of topoisomerase II is a common occurrence in neoplasia, and different topoisomerase II inhibitors have indeed been proven to be powerful anticancer drugs. For this reason, the topoisomerase II catalytic cycle has attracted strong interest, but only a few techniques contributing to studies in this field have emerged. All of the currently used conventional methods to elucidate the action and inhibition of topoisomerase II require separation steps and are therefore unsatisfactory in terms of sensitivity, speed, and throughput. Here, for the first time, we present an assay that works in homogenous solution. The assay is based on dual-color fluorescence cross-correlation spectroscopy (DC-FCCS) and allows monitoring of topoisomerase II action and, especially, detection and discrimination of different topoisomerase II inhibitor classes. The effectiveness of our new assay was confirmed by measuring the effects of a catalytic inhibitor (novobiocin) and a topoisomerase poison (m-AMSA) with bacteriophage T4 topoisomerase as a model system, thus showing the strategy to be easy, fast, and extremely sensitive. Further development of the DC-FCCS-based assay and subsequent application in high-throughput drug screening of new anticancer drugs is proposed and discussed.


Subject(s)
DNA Topoisomerases, Type II/metabolism , Enzyme Inhibitors/pharmacology , Topoisomerase II Inhibitors , Catalysis , DNA/metabolism , Diffusion , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Models, Biological , Novobiocin/chemistry , Novobiocin/pharmacology , Time Factors
7.
J Pept Sci ; 10(9): 566-77, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15473264

ABSTRACT

The coumarin antibiotics are potent inhibitors of DNA replication whose target is the enzyme DNA gyrase, an ATP-dependent bacterial type II topoisomerase. The coumarin drugs inhibit gyrase action by competitive binding to the ATP-binding site of DNA gyrase B protein. The production of new biologically active products has stimulated additional studies on coumarin-gyrase interactions. In this regard, a 4.2 kDa peptide mimic of DNA gyrase B protein from Escherichia coli has been designed and synthesized. The peptide sequence includes the natural fragment 131-146 (coumarin resistance-determining region) and a segment containing the gyrase-DNA interaction region (positions 753-770). The peptide mimic binds to novobiocin (Ka = 1.4+/-0.3 x 10(5) M(-1)), plasmid (Ka = 1.6+/-0.5 x 10(6) M(-1)) and ATP (Ka = 1.9+/-50.4 x 10(3) M(-1)), results previously found with the intact B protein. On the other hand, the binding to novobiocin was reduced when a mutation of Arg-136 to Leu-136 was introduced, a change previously found in the DNA gyrase B protein from several coumarin-resistant clinical isolates of Escherichia coli In contrast, the binding to plasmid and to ATP was not altered. These results suggest that synthetic peptides designed in a similar way to that described here could be used as mimics of DNA gyrase in studies which seek a better understanding of the ATP, as well as coumarin, binding to the gyrase and also the mechanism of action of this class of antibacterial drugs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Coumarins/pharmacology , DNA Gyrase/chemistry , Peptides/chemistry , Adenosine Triphosphate/chemistry , Anti-Bacterial Agents/chemistry , Binding, Competitive , Coumarins/chemistry , DNA/chemistry , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Molecular Probes , Novobiocin/chemistry , Peptides/chemical synthesis , Topoisomerase II Inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL