Page 246 - Read Online
P. 246

Page 8 of 10          Raevsky et al. Neuroimmunol Neuroinflammation 2018;5:33  I  http://dx.doi.org/10.20517/2347-8659.2018.34


               Table 2. Scoring values of ligands docked in the active site of cathepsin K
                Sample                  Total score  Crash score  Internal strain  Complex energy  Fragment RMSD *
                CF 3 P(CH 2 CH 3 ) 3  triethylphosphine  1.3065   -0.4537   0.0682   369.7      0.694
                CF 3  Au-thiomalate       1.984       -0.3104       0.034         317.7         0.705
                                          2.4274      -0.4531       0.0663        390.3         0.528
                CF 3 P(CH 2 CH 3 )(C 6 H 5 ) 2
                                          3.4247      -1.4016       4.5           451.1         0.901
                CF 3 P(C 6 H 5 ) 3
               *Fragment root-mean-square deviation

               compound optimization and docking, a limited set of seven substituted phosphines were synthesized from a
               total of 15 designed structures and used for in vitro studies. The best way of comparing the overall calculated
               binding affinities of ligands is a comparison of their total score values. However, all other scoring functions
               can be useful in specific cases. For example, as can be seen from Table 1, the best experimental and docking
               results were obtained for P(C H NH )(C H )  compound. It is evident that high total scores and low internal
                                              +
                                                 6
                                             3
                                        6
                                                   5 2
                                          4
               strain scores are preferable for the newly developed compounds. Therefore, these parameters were used for
               the further design of the improved novel drug-like compounds. Further optimization and improvement of
               each drug-like compound were based on their spatial location in the enzymatic pocket of cathepsin B and
               compliance to the physical features of the active site, including anchoring with the CF  group and localiza-
                                                                                        3
               tion of phosphine groups in the hydrophobic/hydrophilic regions of the enzymatic pocket.
               DISCUSSION
               Our data show that the described docking model is a practical tool for identification and optimization of
               novel compounds, which have been designed based on their phosphine core structure. We also focused
               our attention on the shape features of the cathepsin B binding site and achieved a good trend in selectivity
               of the inhibitors towards this enzyme, by avoiding their binding to cathepsin K. The inverse correlation
               (-0.71) between docking scores of compounds and their IC  values as well as significant selectivity towards
                                                                 50
               cathepsin B and not cathepsin K [Tables 1 and 2] supported subsequent docking analysis and selection of
               compounds for in vitro testing. Thus, by using the molecular modeling described in this study we were
               able to design and create structurally novel derivatives of the clinically available anti-rheumatic drug
               auranofin, which inhibited the enzymatic activity of cathepsin B more effectively than their parent drug.
               It has been established that the clinical anti-inflammatory activity of auranofin depends on its ability
               to affect multiple cellular and molecular targets . Even though clinical effectiveness of auranofin in
                                                          [41]
               neurodegenerative disorders has not been studied, its anti-inflammatory activity may be beneficial for
               slowing down neuroinflammation accompanying such pathologies as Alzheimer’s disease, Parkinson’s
                                                 [42]
               disease and amyotrophic lateral sclerosis . Optimizing the activity and selectivity of auranofin molecule as
               an inhibitor of cathepsin B through structural modifications may lead to additional benefits of such novel
                                                       [43]
               compounds in Alzheimer’s disease in particular . Further in vivo studies will be required to determine the
               pharmacokinetics and pharmacodynamics of these novel derivatives of auranofin, as well as their clinical
               suitability as anti-neuroinflammatory drugs and cathepsin B inhibitors.



               DECLARATIONS
               Authors’ contributions
               Conceived the study and wrote the manuscript: Raevsky AV, Sharifi M, Pinchuk V, Klegeris A
               Conducted modeling experiments and analyzed the data: Raevsky AV, Sharifi M

               Availability of data and materials
               Data in this study were obtained by experimentation and through in silico modeling, and are original. All
               primary data used to construct the figures and summary tables are available by contacting the authors of
               this study.
   241   242   243   244   245   246   247   248   249   250   251