Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Drug Des Devel Ther ; 12: 4139-4147, 2018.
Article in English | MEDLINE | ID: mdl-30584278

ABSTRACT

BACKGROUND: The protein tyrosine phosphatases PTP1B and SHP2 are promising drug targets in treatment design for breast cancer. Searching for specific inhibitors of their activity has recently become the challenge of many studies. Previous work has indicated that the promising PTP inhibitors may be small compounds that are able to bind and interact with amino residues from the binding site. PURPOSE: The main goal of our study was to synthesize and analyze the effect of selected small peptide inhibitors on oncogenic PTP1B and SHP2 enzymatic activity and viability of MCF7 breast cancer cells. We also performed computational analysis of peptides binding with allosteric sites of PTP1B and SHP2 phosphatases. METHODS: We measured the inhibitory activity of compounds utilizing recombinant enzymes and MCF7 cell line. Computational analysis involved docking studies of binding conformation and interactions of inhibitors with allosteric sites of phosphatases. RESULTS: The results showed that the tested compounds decrease the enzymatic activity of phosphatases PTP1B and SHP2 with IC50 values in micromolar ranges. We observed higher inhibitory activity of dipeptides than tripeptides. Phe-Asp was the most effective against SHP2 enzymatic activity, with IC50=5.2±0.4 µM. Micromolar concentrations of tested dipeptides also decreased the viability of MCF7 breast cancer cells, with higher inhibitory activity observed for the Phe-Asp peptide. Moreover, the peptides tested were able to bind and interact with allosteric sites of PTP1B and SHP2 phosphatases. CONCLUSION: Our research showed that small peptide compounds can be considered for the design of specific inhibitors of oncogenic protein tyrosine phosphatases.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Drug Design , Enzyme Inhibitors/pharmacology , Molecular Docking Simulation , Peptides/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Allosteric Regulation , Antineoplastic Agents/chemical synthesis , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Female , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Molecular Targeted Therapy , Peptides/chemical synthesis , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
2.
Anticancer Res ; 37(9): 4799-4806, 2017 09.
Article in English | MEDLINE | ID: mdl-28870898

ABSTRACT

BACKGROUND: Rapidly-dividing cancer cells have higher requirement for iron compared to non-transformed cells, making iron chelating a potential anticancer strategy. In the present study we compared the anticancer activity of uncommon iron chelator aurintricarboxylic acid (ATA) with the known deferoxamine (DFO). MATERIALS AND METHODS: We investigated the impact of ATA and DFO on the viability and proliferation of MCF-7 cancer cells. Moreover we performed enzymatic activity assays and computational analysis of the ATA and DFO effects on pro-oncogenic phosphatases PTP1B and SHP2. RESULTS: ATA and DFO decrease the viability and proliferation of breast cancer cells, but only ATA considerably reduces the activity of PTP1B and SHP2 phosphatases. Our studies indicated that ATA strongly inactivates and binds in the PTP1B and SHP2 active site, interacting with arginine residue essential for enzyme activity. CONCLUSION: We confirmed that iron chelating can be considered as a potential strategy for the adjunctive treatment of breast cancer.


Subject(s)
Aurintricarboxylic Acid/pharmacology , Breast Neoplasms/enzymology , Deferoxamine/pharmacology , Iron Chelating Agents/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Aurintricarboxylic Acid/chemistry , Binding Sites , Breast Neoplasms/pathology , Catalase/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Deferoxamine/chemistry , Female , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Molecular Docking Simulation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
3.
World J Microbiol Biotechnol ; 32(10): 163, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27562597

ABSTRACT

Yersinia sp. bacteria owe their viability and pathogenic virulence to the YopH factor, which is a highly active bacterial protein tyrosine phosphatase. Inhibition of YopH phosphatase results in the lack of Yersinia sp. pathogenicity. We have previously described that aurintricarboxylic acid inhibits the activity of YopH at nanomolar concentrations and represents a unique mechanism of YopH inactivation due to a redox process. This work is a continuation of our previous studies. Here we show that modifications of the structure of aurintricarboxylic acid reduce the ability to inactivate YopH and lead to higher cytotoxicity. In the present paper we examine the inhibitory properties of aurintricarboxylic acid analogues, such as eriochrome cyanine R (ECR) and pararosaniline. Computational docking studies we report here indicate that ATA analogues are not precluded to bind in the YopH active site and in all obtained binding conformations ECR and pararosaniline bind to YopH active site. The free binding energy calculations show that ECR has a stronger binding affinity to YopH than pararosaniline, which was confirmed by experimental YopH enzymatic activity studies. We found that ATA analogues can reversibly reduce the enzymatic activity of YopH, but possess weaker inhibitory properties than ATA. The ATA analogues induced inactivation of YopH is probably due to oxidative mechanism, as pretreatment with catalase prevents from inhibition. We also found that ATA analogues significantly decrease the viability of macrophage cells, especially pararosaniline, while ATA reveals only slight effect on cell viability.


Subject(s)
Aurintricarboxylic Acid/analogs & derivatives , Bacterial Outer Membrane Proteins/chemistry , Benzenesulfonates/chemistry , Protein Tyrosine Phosphatases/chemistry , Rosaniline Dyes/chemistry , Toluidines/chemistry , Yersinia/drug effects , Animals , Aurintricarboxylic Acid/chemistry , Aurintricarboxylic Acid/pharmacology , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Benzenesulfonates/pharmacology , Catalytic Domain/drug effects , Cell Line , Cell Survival/drug effects , Mice , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Protein Binding , Protein Tyrosine Phosphatases/antagonists & inhibitors , Rosaniline Dyes/pharmacology , Toluidines/pharmacology , Yersinia/enzymology
4.
Oncotarget ; 7(3): 2229-38, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26735581

ABSTRACT

Chicoric acid (CA) is a phenolic compound present in dietary supplements with a large spectrum of biological properties reported ranging from antioxidant, to antiviral, to immunostimulatory properties. Due to the fact that chicoric acid promotes phagocytic activity and was reported as an allosteric inhibitor of the PTP1B phosphatase, we examined the effect of CA on YopH phosphatase from pathogenic bacteria, which block phagocytic processes of a host cell. We performed computational studies of chicoric acid binding to YopH as well as validation experiments with recombinant enzymes. In addition, we performed similar studies for caffeic and chlorogenic acids to compare the results. Docking experiments demonstrated that, from the tested compounds, only CA binds to both catalytic and secondary binding sites of YopH. Our experimental results showed that CA reduces activity of recombinant YopH phosphatase from Yersinia enterocolitica and human CD45 phosphatase. The inhibition caused by CA was irreversible and did not induce oxidation of catalytic cysteine. We proposed that inactivation of YopH induced by CA is involved with allosteric inhibition by interacting with essential regions responsible for ligand binding.


Subject(s)
Bacterial Outer Membrane Proteins/antagonists & inhibitors , Caffeic Acids/chemistry , Catalytic Domain/drug effects , Protein Tyrosine Phosphatases/antagonists & inhibitors , Succinates/chemistry , Yersinia enterocolitica/enzymology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Binding Sites , Chlorogenic Acid/chemistry , Cysteine/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Virulence Factors , Yersinia enterocolitica/pathogenicity
5.
Oncotarget ; 6(21): 18364-73, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26286963

ABSTRACT

YopH is a bacterial protein tyrosine phosphatase, which is essential for the viability and pathogenic virulence of the plague-causing Yersinia sp. bacteria. Inactivation of YopH activity would lead to the loss of bacterial pathogenicity. We have studied the inhibitory properties of aurintricarboxylic acid (ATA) against YopH phosphatase and found that at nanomolar concentrations ATA reversibly decreases the activity of YopH. Computational docking studies indicated that in all binding poses ATA binds in the YopH active site. Molecular dynamics simulations showed that in the predicted binding pose, ATA binds to the essential Cys403 and Arg409 residues in the active site and has a stronger binding affinity than the natural substrate (pTyr). The cyclic voltammetry experiments suggest that ATA reacts remarkably strongly with molecular oxygen. Additionally, the electrochemical reduction of ATA in the presence of a negative potential from -2.0 to 2.5 V generates a current signal, which is observed for hydrogen peroxide. Here we showed that ATA indicates a unique mechanism of YopH inactivation due to a redox process. We proposed that the potent inhibitory properties of ATA are a result of its strong binding in the YopH active site and in situ generation of hydrogen peroxide near catalytic cysteine residue.


Subject(s)
Aurintricarboxylic Acid/chemistry , Bacterial Outer Membrane Proteins/chemistry , Protein Tyrosine Phosphatases/chemistry , Virulence Factors/chemistry , Algorithms , Aurintricarboxylic Acid/metabolism , Aurintricarboxylic Acid/pharmacology , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Bacterial Outer Membrane Proteins/metabolism , Humans , Kinetics , Molecular Conformation , Molecular Dynamics Simulation , Molecular Structure , Oxidation-Reduction , Plague/microbiology , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/metabolism , Virulence , Virulence Factors/antagonists & inhibitors , Virulence Factors/metabolism , Yersinia pestis/metabolism , Yersinia pestis/pathogenicity , Yersinia pestis/physiology
6.
Theor Biol Med Model ; 11: 52, 2014 Dec 27.
Article in English | MEDLINE | ID: mdl-25542608

ABSTRACT

A variety of topics are reviewed in the area of mathematical and computational modeling in biology, covering the range of scales from populations of organisms to electrons in atoms. The use of maximum entropy as an inference tool in the fields of biology and drug discovery is discussed. Mathematical and computational methods and models in the areas of epidemiology, cell physiology and cancer are surveyed. The technique of molecular dynamics is covered, with special attention to force fields for protein simulations and methods for the calculation of solvation free energies. The utility of quantum mechanical methods in biophysical and biochemical modeling is explored. The field of computational enzymology is examined.


Subject(s)
Computer Simulation , Entropy , Molecular Dynamics Simulation
7.
Curr Top Med Chem ; 13(9): 989-1001, 2013.
Article in English | MEDLINE | ID: mdl-23651479

ABSTRACT

Protein:protein interactions are becoming increasingly significant as potential drug targets; however, the rational identification of small molecule inhibitors of such interactions remains a challenge. Pharmacophore modelling is a popular tool for virtual screening of compound libraries, and has previously been successfully applied to the discovery of enzymatic inhibitors. However, the application of pharmacophore modelling in the field of protein:protein interaction inhibitors has historically been considered more of a challenge and remains limited. In this review, we explore the interaction mimicry by known inhibitors that originate from in vitro screening, demonstrating the validity of pharmacophore mapping in the generation of queries for virtual screening. We discuss the pharmacophore mapping methods that have been successfully employed in the discovery of first-in-class inhibitors. These successful cases demonstrate the usefulness of a "tool kit" of diverse strategies for application across a range of situations depending on the available structural information.


Subject(s)
Drug Discovery , Proteins/antagonists & inhibitors , Proteins/metabolism , High-Throughput Screening Assays , Models, Molecular , Molecular Weight , Protein Binding/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...