Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Appl Microbiol Biotechnol ; 107(23): 7269-7285, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37741938

ABSTRACT

Pseudomonas aeruginosa is an emerging threat for hospitalized and cystic fibrosis patients. Biofilm, a microbial community embedded in extracellular polymeric substance, fortifies bacteria against the immune system. In biofilms, the expression of functional amyloids is linked with highly aggregative, multi-resistant strains, and chronic infections. Serrapeptase (SPT), a protease possessing similar or superior anti-microbial properties with many antibiotics, presents anti-amyloid potential. However, studies on the employment of SPT against Pseudomonas biofilms and Fap amyloid, or the possible mechanisms of action are scarce. Here, SPT inhibited biofilm formation of P. aeruginosa ATCC 27853 on both plastic and glass surfaces, with an IC50 of 11.26 µg/mL and 0.27 µg/mL, respectively. The inhibitory effect of SPT on biofilm was also verified with optical microscopy of crystal violet-stained biofilms and with confocal microscopy. Additionally, SPT caused a dose-dependent decrease of bacterial viability (IC50 of 3.07 µg/mL) as demonstrated by MTT assay. Reduction of bacterial functional amyloids was also demonstrated, employing both fluorescence microscopy with thioflavin T and photometrical determination of Congo-red-positive compounds. Both viability and functional amyloids correlated significantly with biofilm inhibition. Finally, in silico molecular docking studies provided a mechanistic insight into the interaction of SPT with FapC or FapD, proving that both peptides are possible targets of SPT. These results offer new insights into the biofilm formation of P. aeruginosa and potentiate the involvement of SPT in the prevention and eradication of Pseudomonas biofilms. KEY POINTS: • Serrapeptase inhibits biofilm formation of P. aeruginosa on plastic and glass. • Biofilm inhibition correlated with reduced viability and functional amyloid levels. • In silico studies indicated that serrapeptase may target FapC and FapD peptides.


Subject(s)
Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Extracellular Polymeric Substance Matrix/metabolism , Molecular Docking Simulation , Biofilms , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Peptide Hydrolases/metabolism , Peptides/metabolism , Pseudomonas Infections/microbiology , Microbial Sensitivity Tests
2.
Molecules ; 27(21)2022 Nov 04.
Article in English | MEDLINE | ID: mdl-36364398

ABSTRACT

Since there is an urgent need for novel treatments to combat the current coronavirus disease 2019 (COVID-19) pandemic, in silico molecular docking studies were implemented as an attempt to explore the ability of selected bioactive constituents of extra virgin olive oil (EVOO) to act as potent SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) antiviral compounds, aiming to explore their ability to interact with SARS-CoV-2 Spike key therapeutic target protein. Our results suggest that EVOO constituents display substantial capacity for binding and interfering with Spike (S) protein, both wild-type and mutant, via the receptor-binding domain (RBD) of Spike, or other binding targets such as angiotensin-converting enzyme 2 (ACE2) or the RBD-ACE2 protein complex, inhibiting the interaction of the virus with host cells. This in silico study provides useful insights for the understanding of the mechanism of action of the studied compounds at a molecular level. From the present study, it could be suggested that the studied active phytochemicals could potentially inhibit the Spike protein, contributing thus to the understanding of the role that they can play in future drug designing and the development of anti-COVID-19 therapeutics.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Humans , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Olive Oil , Molecular Docking Simulation , Peptidyl-Dipeptidase A/metabolism , Binding Sites , Protein Binding
3.
J Inorg Biochem ; 231: 111805, 2022 06.
Article in English | MEDLINE | ID: mdl-35334392

ABSTRACT

In silico molecular docking studies, in vitro toxicity and in silico predictions on the biological activity profile, pharmacokinetic properties, drug-likeness, ADMET (absorption, distribution, metabolism, excretion, and toxicity) physicochemical pharmacokinetic data, and target proteins and toxicity predictions were performed on six copper(II) complexes with the non-steroidal anti-inflammatory drugs ibuprofen, loxoprofen, fenoprofen and clonixin as ligands, in order to investigate the ability of these complexes to interact with the key therapeutic target proteins of SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) 3C-like cysteine main protease (3CLpro/Mpro), viral papain-like protease (PLpro), RNA-dependent RNA polymerase (RdRp), and non-structural proteins (Nsps) Nsp16-Nsp10 2'-O-methyltransferase complex, and their capacity to act as antiviral agents, contributing thus to understanding the role they can play in the context of coronavirus 2019 (COVID-19) pandemic. Cytotoxic activity against five human cancer and normal cell lines were also evaluated.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Anti-Inflammatory Agents , Antiviral Agents/chemistry , Copper , Humans , Molecular Docking Simulation , SARS-CoV-2
4.
J Inorg Biochem ; 228: 111695, 2022 03.
Article in English | MEDLINE | ID: mdl-35007963

ABSTRACT

A series of heteroleptic Ag(I) complexes bearing 4,6-dimethyl-2-pyrimidinethiol (dmp2SH), i.e., [AgCl(dmp2SH)(PPh3)2] (1), [Ag(dmp2SH)(PPh3)2]NO3 (2), [Ag(dmp2SΗ)(xantphos)]NO3 (3), [Ag(µ-dmp2S)(PPh3)]2 (4), [Ag(dmp2S)(xantphos)] (5), [Ag(µ-dmp2S)(DPEphos)]2 (6) (xantphos = 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene and DPEPhos = bis[(2-diphenylphosphino)phenyl]ether) were synthesized. The complexes display systematic variation of particular structural characteristics which were proved to have a significant impact on their in vitro cytotoxicity and antimicrobial properties. A moderate-to-high potential for bacteria growth inhibition was observed for all complexes, with 2, 3 and 5 being particularly effective against Gram-(+) bacteria (IC50 = 1.6-4.5 µM). The three complexes exhibit high in vitro cytotoxicity against HeLa and MCF-7 cancer cells (IC50 = 0.32-3.00 µΜ), suggesting the importance of coordination unsaturation and cationic charge for effective bioactivity. A very low cytotoxicity against HDFa normal cells was observed, revealing a high degree of selectivity (selectivity index ~10) and, hence, biocompatibility. Fluorescence microscopy using 2 showed effective targeting on the membrane of the HeLa cancer cells, subsequently inducing cell death. Binding of the complexes to serum albumin proteins is reasonably strong for potential uptake and subsequent release to target sites. A moderate in vitro antioxidant capacity for free radicals scavenging was observed and a low potential to destroy the double-strand structure of calf-thymus DNA by intercalation, suggesting likely implication of these properties in the bioactivity mechanisms of these complexes. Further insight into possible mechanisms of bioactivity was obtained by molecular modeling calculations, by exploring their ability to act as potential inhibitors of DNA-gyrase, human estrogen receptor alpha, human cyclin-dependent kinase 6, and human papillomavirus E6 oncoprotein.


Subject(s)
Anti-Infective Agents/pharmacology , Coordination Complexes/chemistry , Silver/chemistry , Thioamides/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/chemistry , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Bacteria/drug effects , Cyclin-Dependent Kinase 6/metabolism , DNA/metabolism , DNA Gyrase/metabolism , HeLa Cells , Humans , Ligands , MCF-7 Cells , Microbial Sensitivity Tests/methods , Models, Molecular , Molecular Docking Simulation/methods , Phosphines/chemistry , Silver/pharmacology , Thioamides/pharmacology , Xanthenes/chemistry
5.
Exp Gerontol ; 156: 111621, 2021 12.
Article in English | MEDLINE | ID: mdl-34748951

ABSTRACT

Oxidative/nitrative stress that results from the unbalance of the overproduction/clearance of reactive oxygen/nitrogen species (ROS/NOS), originated from a variety of endo- and/or exo-genous sources, can have detrimental effects on DNA and is involved in Alzheimer's disease (AD) pathology. An excellent marker of oxidative DNA lesions is 8-hydroxy-2'-deoxyguanosine (8-OHdG) while of nitrative stress the enzyme NOS2 (Nitric oxide synthase 2). Under massive oxidative stress, poly(ADP-ribose)polymerase 1 (PARP-1) enzyme activity, responsible for restoration of DNA damage, is augmented, DNA repair enzymes are recruited, and cell survival/or death is ensued through PARP-1 activation, which is correlated positively with neurodegenerative diseases. In this biochemical study the levels of PARP-1, 8-oxo-dG, and NOS2, Aß1-42, and p-tau in their sera determined using Enzyme-Linked Immunosorbent Assay (ELISA). Patients diagnosed with Mild Cognitive Impairment participated in MICOIL clinical trial, were daily administered with 50 ml Extra Virgin Olive Oil (EVOO) for one year. All MCI patients' biomarkers that had consumed EVOO were tantamount to those of healthy participants, contrary to MCI patients who were not administered. EVOO administration in MCI patients resulted in the restoration of DNA damage and of the well-established "hallmarks" AD biomarkers, thanks probably to its antioxidant properties exhibiting a therapeutic potentiality against AD. Molecular docking simulations of the EVOO constituents on the crystal structure of PARP-1 and NOS-2 target enzymes were also employed, to study in silico the ability of the compounds to bind to these enzymes and explain the observed in vitro activity. In silico analysis has proved the binding of EVOO constituents on PARP-1and NOS-2 enzymes and their interaction with crucial amino acids of the active sites. CLINICAL TRIAL REGISTRATION: https://clinicaltrials.gov/ct2/show/NCT03362996. MICOIL GOV IDENTIFIER: NCT03362996.


Subject(s)
Cognitive Dysfunction , Poly(ADP-ribose) Polymerase Inhibitors , DNA Damage , Humans , Molecular Docking Simulation , Olive Oil/pharmacology , Oxidative Stress , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
6.
J Inorg Biochem ; 224: 111563, 2021 11.
Article in English | MEDLINE | ID: mdl-34399232

ABSTRACT

Six novel copper(II) complexes with the non-steroidal anti-inflammatory drugs ibuprofen, loxoprofen, fenoprofen and clonixin as ligands were synthesized and characterized by diverse techniques including single-crystal X-ray crystallography. The in vitro scavenging activity of the complexes against 1,1-diphenyl-picrylhydrazyl and 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) free radicals and the ability to reduce H2O2 were studied in the context of the antioxidant activity studies. The complexes may interact with calf-thymus DNA via intercalation as revealed by the techniques employed. The affinity of the complexes for bovine and human serum albumins was evaluated by fluorescence emission spectroscopy and the corresponding binding constants were determined. Molecular docking simulations on the crystal structure of calf-thymus DNA, human and bovine serum albumins were also employed in order to study in silico the ability of the studied compounds to bind to these target biomacromolecules, in terms of impairment of DNA and transportation through serum albumins, to explain the observed in vitro activity and to establish a possible mechanism of action.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Coordination Complexes/chemistry , Copper/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/chemistry , Clonixin/chemistry , Coordination Complexes/pharmacology , Copper/pharmacology , Crystallography, X-Ray/methods , DNA/chemistry , Fenoprofen/chemistry , Free Radical Scavengers/chemistry , Humans , Hydrogen Peroxide/chemistry , Ibuprofen/chemistry , Intercalating Agents/chemistry , Molecular Docking Simulation/methods , Phenylpropionates/chemistry , Serum Albumin, Bovine/chemistry , Serum Albumin, Human/chemistry
7.
J Inorg Biochem ; 222: 111507, 2021 09.
Article in English | MEDLINE | ID: mdl-34139455

ABSTRACT

Five novel nickel(II) complexes with the non-steroidal anti-inflammatory drug sodium meclofenamate (Na-mclf) have been synthesized and characterized in the absence or co-existence of the nitrogen-donors imidazole (Himi), 2,2'-bipyridylamine (bipyam), 2,2'-bipyridylketoxime (Hpko) and 2,9-dimethyl-1,10-phenanthroline (neoc); namely [Ni(mclf-O)2(Himi)2(MeOH)2], [Ni(mclf-O)2(MeOH)4], [Ni(mclf-O)(mclf-O,O')(bipyam)(MeOH)]·0.25MeOH, [Ni(mclf-O,O')2(neoc)] and [Ni(mclf-O)2(Hpko-N,N')2]·MeOH·0.5H2O. The affinity of the complexes for calf-thymus (CT) DNA was investigated by various techniques and intercalation is suggested as the most possible interaction mode. The interaction of the complexes for bovine and human serum albumins was also investigated in order to determine the binding constants, concluding that the complexes bind reversibly to albumins for the transportation towards their target cells or tissues and their release upon arrival at biotargets. The antioxidant activity of the compounds was evaluated via their ability to scavenge 1,1-diphenyl-picrylhydrazyl and 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) free radicals and to reduce H2O2. For the determination of the anticholinergic ability of the complexes the in vitro inhibitory activity against the enzymes acetylcholinesterase and butyrylcholinesterase was evaluated and presented promising results. The in silico molecular modeling calculations employed provide useful insights for the understanding of the mechanism of action of the studied complexes at a molecular level. This applies on both the impairment of DNA by its binding with the studied complexes and transportation through serum albumins, as well as the ability of these compounds to act as anticholinergic agents.


Subject(s)
Coordination Complexes/chemistry , DNA/metabolism , Meclofenamic Acid/analogs & derivatives , Serum Albumin, Bovine/metabolism , Serum Albumin, Human/metabolism , Acetylcholinesterase/chemistry , Animals , Butyrylcholinesterase/chemistry , Cattle , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/metabolism , Coordination Complexes/chemical synthesis , Coordination Complexes/metabolism , Free Radical Scavengers/chemical synthesis , Free Radical Scavengers/chemistry , Free Radical Scavengers/metabolism , Humans , Meclofenamic Acid/chemical synthesis , Meclofenamic Acid/metabolism , Molecular Docking Simulation , Molecular Structure , Nickel/chemistry
8.
J Inorg Biochem ; 221: 111402, 2021 08.
Article in English | MEDLINE | ID: mdl-33975249

ABSTRACT

An oxovanadium(IV) - curcumin based complex, viz. [VO(cur)(2,2´-bipy)(H2O)] where cur is curcumin and bipy is bipyridine, previously synthesized, has been studied for interaction with albumin and DNA. Fluorescence emission spectroscopy was used to evaluate the interaction of the complex with bovine serum albumin (BSA) and the BSA-binding constant (Kb) was calculated to be 2.56 x 105 M-1, whereas a single great-affinity binding site was revealed. Moreover, the hemocompatibility test demonstrated that the complex presented low hemolytic fraction (mostly below 1%), in all concentrations tested (0-250 µΜ of complex, 5% DMSO) assuring a safe application in interaction with blood. The binding of the complex to DNA was also investigated using absorption, fluorescence, and viscometry methods indicating a binding through a minor groove mode. From competitive studies with ethidium bromide the apparent binding constant value to DNA was estimated to be 4.82 x 106 M-1. Stern-Volmer quenching phenomenon gave a ΚSV constant [1.92 (± 0.05) x 104 M-1] and kq constant [8.33 (± 0.2) x 1011 M-1s-1]. Molecular docking simulations on the crystal structure of BSA, calf thymus DNA, and DNA gyrase, as well as pharmacophore analysis for BSA target, were also employed to study in silico the ability of [VO(cur)(2,2´-bipy)(H2O)] to bind to these target bio-macromolecules and explain the observed in vitro activity.


Subject(s)
Coordination Complexes/metabolism , Curcumin/metabolism , DNA Gyrase/metabolism , DNA/metabolism , Serum Albumin, Bovine/metabolism , Animals , Binding Sites , Cattle , Coordination Complexes/chemistry , Coordination Complexes/toxicity , Curcumin/analogs & derivatives , Curcumin/toxicity , DNA/chemistry , DNA Gyrase/chemistry , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Hemolysis/drug effects , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Serum Albumin, Bovine/chemistry , Vanadium/chemistry , Vanadium/toxicity , Viscosity/drug effects
9.
J Inorg Biochem ; 212: 111213, 2020 11.
Article in English | MEDLINE | ID: mdl-32889129

ABSTRACT

The interaction of cobalt chloride with the non-steroidal anti-inflammatory drug indomethacin (Hindo) led to the formation of the polymeric complex [Co(indo-O)2(H2O)2(µ-Cl)]n·n(MeOH·H2O) bearing one chlorido bridge between the cobalt atoms. The presence of the nitrogen-donor co-ligands 2,2'-bipyridine (bipy), 2,2'-bipyridylamine (bipyam), 1,10-phenanthroline (phen) or 1H-imidazole (Himi) resulted in the isolation of complexes [Co2(µ-indo-O,O')2(indo-O)2(bipy)2(µ-H2O)]·3.3MeOH, [Co(indo-O,O')2(bipyam)]·0.9MeOH·0.2H2O, [Co(indo-O,O')2(phen)] (4) and [Co(indo-O)2(Himi)2] (5), respectively, where the indomethacin ligands were coordinated in diverse manners. The study of the affinity of the complexes for calf-thymus DNA revealed their intercalation between the DNA-bases. The binding of the complexes to albumins was also examined and the corresponding binding constants and binding subdomain were determined. The free radical scavenging activity of the compounds was evaluated towards 1,1-diphenyl-picrylhydrazyl and 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid). Molecular modeling calculations may usually provide a molecular basis for the understanding of both the impairment of DNA by its binding with the studied complexes and the ability of these compounds to transportation through serum albumin proteins. This study can provide information for the elucidation of the mechanism of action of the compounds in a molecular level.


Subject(s)
Cobalt/chemistry , Coordination Complexes/chemistry , Indomethacin/chemistry , Computer Simulation , In Vitro Techniques
10.
J Inorg Biochem ; 208: 111085, 2020 07.
Article in English | MEDLINE | ID: mdl-32454249

ABSTRACT

The interaction of Cu(NO3)2·3H2O with the sulfonyl o-pyridine carboxamidoxime N'-(4-nitrophenylsulfonyloxy)picolinimidamide (L) resulted in the mononuclear complex [Cu(L1)2](L2)2 (1), where L1 = pyridine-2-carboxamidine ligand and (L2)- = 4-nitrobenzenesulfonate anion derived from the homolytic cleavage of the NO bond of L. The complex was characterized by diverse techniques including single-crystal X-ray crystallography. From the antimicrobial tests performed, complex 1 seems to be active against gram-negative bacterial strains. The complex binds tightly and reversibly to serum albumins and tightly to calf-thymus DNA via an intercalative mode and also via electrostatic interactions (as expected due to its cationic nature). Additionally, it interacts with (pBluescriptSK(+)) plasmid DNA in a concentration-dependent manner. The results from the present in silico molecular modeling simulations provide useful complementary insights for the elucidation of the mechanism of action of the studied complex at a molecular level. Molecular modeling calculations provide a molecular basis for the understanding of both the impairment of DNA by its binding with the studied complex and the ability of this compound to act as an antibacterial agent, most probably by its activity against DNA-gyrase, as well as for transportation through serum albumins and possible interaction with other protein targets involved in various diseases.


Subject(s)
Anti-Bacterial Agents , Bacteria/growth & development , Coordination Complexes , Copper , Intercalating Agents , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Copper/chemistry , Copper/pharmacology , DNA/chemistry , Intercalating Agents/chemical synthesis , Intercalating Agents/chemistry , Intercalating Agents/pharmacology , Molecular Docking Simulation , Pyridines/chemistry , Pyridines/pharmacology , Serum Albumin, Bovine/chemistry
11.
J Inorg Biochem ; 199: 110792, 2019 10.
Article in English | MEDLINE | ID: mdl-31365891

ABSTRACT

The synthesis and characterization of the Pd(II) complex of the formula [Pd(L)2] 1 with the Schiff base 4-chloro-2-(N-ethyliminomethyl)-phenol (HL) as derived in situ via the condensation reaction of 5-chloro-salicylaldehyde and ethylamine was undertaken. The structure of 1 was verified by single-crystal X-ray crystallography. The ability of 1 to interact with calf-thymus (CT) DNA was studied by UV-vis and viscosity experiments, and its ability to displace ethidium bromide (EB) from the DNA-EB conjugate was revealed by fluorescence spectroscopy. It was found that intercalation is the most possible mode of interaction with CT DNA. Additionally, DNA electrophoretic mobility experiments showed that 1 interacts with the plasmid pBluescript SK(+) (pDNA) as proved by the formation of unusual mobility DNA bands and degradation of relaxed pDNA at concentration of 5 mM. The interaction of 1 with human (HSA) and bovine serum albumin (BSA) was monitored revealing its reversible binding to albumins. The complex showed noteworthy antimicrobial activity against one (Bacillus subtilis) of the five tested bacteria. In order to explain the described in vitro activity of the compound, we adopted molecular docking studies on the crystal structure of HSA, BSA, CT DNA and DNA-gyrase. Furthermore, in silico predictive tools have been employed to study the properties of the complex. The in silico studies are adopted on a multitude of proteins involved in cancer growth, as well as prediction of drug-induced changes of gene expression profile, protein- and mRNA-based prediction results, prediction of sites of metabolism, cytotoxicity for cancer cell lines, etc.


Subject(s)
DNA/chemistry , DNA/pharmacology , Ethidium/analogs & derivatives , Palladium/chemistry , Phenol/chemistry , Schiff Bases/chemistry , Serum Albumin/chemistry , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Bacillus subtilis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , DNA Gyrase/metabolism , Ethidium/chemistry , Ethidium/pharmacology , Humans , MCF-7 Cells , Molecular Docking Simulation , Molecular Structure , Protein Binding , Serum Albumin, Bovine/chemistry , Serum Albumin, Human/chemistry
12.
J Inorg Biochem ; 194: 85-96, 2019 05.
Article in English | MEDLINE | ID: mdl-30844610

ABSTRACT

The synthesis and characterization of four palladium(II) complexes with substituted salicylaldehydes (X-saloH) having the general formula [Pd(X-salo)2] was undertaken. The complexes are formulated as [Pd(3-OCH3-salo)2] 1, [Pd(5-NO2-salo)2] 2, [Pd(5-Cl-salo)2] 3, and [Pd(5-Br-salo)2] 4. The structure of complex 1 was verified by single-crystal X-ray crystallography. Spectroscopic (UV-vis), and physicochemical (viscosity measurements) techniques were employed in order to study the binding of the complexes with calf-thymus (CT) DNA, while ethidium bromide (EB) displacement studies, performed by fluorescence emission spectroscopy, revealed the ability of the complexes to displace the DNA-bound EB. Intercalation is the most possible mode of interaction of the complexes with CT DNA. The interaction of the complexes with bovine (BSA) and human (HSA) serum albumin proteins was studied by fluorescence emission spectroscopy and the relatively high binding constants revealed the reversible binding of the complexes to the albumins. Molecular docking simulations on the crystal structure of HSA, BSA and CT DNA were employed in order to study in silico the ability of the studied complexes 1-4 to bind to these target macromolecules.


Subject(s)
Aldehydes/metabolism , Benzaldehydes/metabolism , Coordination Complexes/metabolism , DNA/metabolism , Intercalating Agents/metabolism , Serum Albumin, Bovine/metabolism , Serum Albumin, Human/metabolism , Aldehydes/chemical synthesis , Aldehydes/chemistry , Animals , Benzaldehydes/chemical synthesis , Benzaldehydes/chemistry , Cattle , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Humans , Intercalating Agents/chemical synthesis , Intercalating Agents/chemistry , Ligands , Molecular Docking Simulation , Molecular Structure , Palladium/chemistry , Protein Binding
13.
J Inorg Biochem ; 190: 1-14, 2019 01.
Article in English | MEDLINE | ID: mdl-30312777

ABSTRACT

The in vitro and in silico biological properties of two manganese complexes with the non-steroidal anti-inflammatory drug mefenamic acid (Hmef) in the presence or absence of salicylaldoxime (Η2sao), i.e. [Μn6(O)2(mef)2(sao)6(CH3OH)4] 1, and [Μn(mef)2(CH3OH)4] 2, respectively, are presented in the present contribution. More specifically, the in vitro biological activity of the complexes was investigated by studying their affinity to calf-thymus DNA (by diverse spectroscopic and physicochemical techniques) and their binding towards bovine (BSA) or human serum albumin (HSA) (by fluorescence emission spectroscopy). Molecular docking simulations on the crystal structures of HSA and DNA, exploring in silico the ability of the complexes to bind to these macromolecules, were also employed in order to explain the described in vitro activity of the compounds. Furthermore, in silico predictive tools have been employed to study the properties of the most active complex 2 to act as anticancer agent, in continuation of the previously reported cytotoxic activity. It is adopted in silico studies on a multitude of proteins involved in cancer growth, as well as prediction of drug-induced changes of gene expression profile, protein- and mRNA-based prediction results, prediction of sites of metabolism, quantitative prediction of antitarget interaction profiles etc.


Subject(s)
Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Manganese/chemistry , Mefenamic Acid/chemistry , Animals , Cattle , Computer Simulation , Coordination Complexes/metabolism , DNA/metabolism , Drug Evaluation, Preclinical , Humans , In Vitro Techniques , Molecular Docking Simulation , Molecular Structure , Protein Binding , Serum Albumin, Human/metabolism , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet
14.
J Inorg Biochem ; 187: 41-55, 2018 10.
Article in English | MEDLINE | ID: mdl-30055395

ABSTRACT

In the present contribution, the biological properties of four manganese complexes with the non-steroidal anti-inflammatory drugs sodium diclofenac (Nadicl) or indomethacin (Hindo) in the presence or absence of salicylaldoxime (Η2sao), i.e. [Μn6(O)2(dicl)2(sao)6(CH3OH)6] 1, [Μn6(O)2(indo)2(sao)6(H2O)4], 2, [Μn(dicl)2(CH3OH)4], 3, and [Μn(indo)2(CH3OH)4], 4 are presented. More specifically, the in vitro cytotoxic effects of the complexes were evaluated against three cancer cell lines (HeLa, MCF-7 and A549 cells) as well as their combinatory activity with the well-known chemotherapeutic drugs irinotecan, cisplatin, paclitaxel and 5-fluorouracil. The biological activity of the complexes was investigated in vitro by studying their affinity to calf-thymus DNA and their binding towards bovine or human serum albumin (HSA). Molecular docking simulations on the crystal structure of HSA and human estrogen receptor alpha (hERa) were employed in order to study in silico the ability of the studied complexes to bind to these proteins.


Subject(s)
Coordination Complexes , Cytotoxins , Diclofenac , Indomethacin , Manganese , Molecular Docking Simulation , A549 Cells , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Cytotoxins/chemical synthesis , Cytotoxins/chemistry , Cytotoxins/pharmacology , Diclofenac/chemistry , Diclofenac/pharmacology , Estrogen Receptor alpha/chemistry , HeLa Cells , Humans , Indomethacin/chemistry , Indomethacin/pharmacology , MCF-7 Cells , Manganese/chemistry , Manganese/pharmacology , Molecular Structure , Serum Albumin, Human/chemistry
15.
Amino Acids ; 50(2): 279-291, 2018 02.
Article in English | MEDLINE | ID: mdl-29185031

ABSTRACT

Quercetin is a flavonoid presenting cytotoxicity against different cancer cell lines. We hypothesized that its core could serve as a scaffold for generating more potent compounds. A quercetin-alanine bioconjugate was synthesized, its cellular internalization was monitored through confocal microscopy and its cytotoxic activity was explored against ten different cell lines. The bioconjugate consistently illustrated enhanced cytotoxic activity with respect to the parent compound. A threefold enhancement in its cytotoxicity was revealed for HeLa, A549, MCF-7 and LNCaP cells. In silico studies suggested that quercetin-alanine possesses enhanced binding affinity to human estrogen receptor alpha corroborating to its activity to MCF-7, overexpressing this receptor. Spectrofluorimetric, calorimetric and in silico studies revealed that quercetin-alanine binds primarily to Sudlow site I of serum albumin mainly through hydrogen bonding. Through this array of experiments we discovered that the specific compound bears a more refined pharmaceutical profile in contrast to quercetin in terms of cytotoxicity, while at the same time preserves its affinity to serum albumin. Natural products could thus offer a potent scaffold to develop bioconjugates with amplified therapeutic window.


Subject(s)
Antineoplastic Agents/pharmacology , Quercetin/analogs & derivatives , Quercetin/pharmacology , Alanine/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Biological Products/chemistry , Biological Products/metabolism , Biological Products/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Flavonoids/chemistry , Flavonoids/metabolism , Flavonoids/pharmacology , Humans , Inhibitory Concentration 50 , Mice , Molecular Docking Simulation , Protein Binding/drug effects , Quercetin/chemistry , Quercetin/metabolism , Serum Albumin/metabolism , Structure-Activity Relationship
16.
Org Biomol Chem ; 15(37): 7956-7976, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28902204

ABSTRACT

Anti-apoptotic proteins, like the Bcl-2 family proteins, present an important therapeutic cancer drug target. Their activity is orchestrated through neutralization upon interaction of pro-apoptotic protein counterparts that leads to immortality of cancer cells. Therefore, generating compounds targeting these proteins is of immense therapeutic importance. Herein, Induced Fit Docking (IFD) and Molecular Dynamics (MD) simulations were performed to rationally design quercetin analogues that bind in the BH3 site of the Bcl-xL protein. IFD calculations determined their binding cavity while Molecular Mechanics Poisson Boltzmann Surface Area (MM-PBSA) and Molecular Mechanics Generalised Born Surface Area (MM-GBSA) calculations provided an insight into the binding enthalpies of the analogues. The quercetin analogues were synthesized and their binding to Bcl-xL was verified with fluorescence spectroscopy. The binding affinity and the thermodynamic parameters between Bcl-xL and quercetin-glutamic acid were estimated through Isothermal Titration Calorimetry. 2D 1H-15N HSQC NMR chemical shift perturbation mapping was used to chart the binding site of the quercetin analogues in the Bcl-xL that overlapped with the predicted poses generated by both IFD and MD calculations. Furthermore, evaluation of the four conjugates against the prostate DU-145 and PC-3 cancer cell lines, revealed quercetin-glutamic acid and quercetin-alanine as the most potent conjugates bearing the higher cytostatic activity. This pinpoints that the chemical space of natural products can be tailored to exploit new hits for difficult tractable targets such as protein-protein interactions.


Subject(s)
Amino Acids/pharmacology , Antineoplastic Agents/pharmacology , Cytostatic Agents/pharmacology , Drug Design , Quercetin/pharmacology , bcl-X Protein/antagonists & inhibitors , Amino Acids/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cytostatic Agents/chemical synthesis , Cytostatic Agents/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Structure , Quercetin/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
17.
Anticancer Drugs ; 28(5): 489-502, 2017 06.
Article in English | MEDLINE | ID: mdl-28272098

ABSTRACT

The aim of this study was to evaluate whether the palliative treatment for metastatic disease with dexamethasone (DEX) plus octreotide (OCT) can improve the anticancer effects of the standard treatment with adriamycin (ADR) on a 4T1 metastatic breast cancer (MBC) model. 4T1 cells were first characterized for the expression of the somatostatin receptors 1-5 and were then inoculated onto the femur of BALB/C mice. Investigation protocols used 4T1 cell proliferation and invasion assays, analysis of radiographic images of the bone metastatic lesions, and overall survival of the diseased animals. The triple combination treatment regime (ADR+OCT+DEX) was ineffective for growth inhibition and showed an antagonistic effect on ADR activity in the 4T1 cell line in both proliferation and invasion assays. ADR treatment following the administration of the DEX+OCT regimen decreased the anticancer activity of ADR both on the grading of the bone metastatic lesions and on the overall survival of diseased animals. Moreover, the palliation treatment with OCT+DEX and in combination with ADR rather caused disease progression of the metastatic disease and bone lesions in a 4T1 MBC model in vivo. These results suggest that the administration of the DEX+OCT regimen, although may preserve palliative effects, neutralizes or reverses the anticancer effects of ADR on a 4T1 MBC model in vitro and in vivo. The simultaneous use of these drugs should be considered carefully in clinical practice.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Mammary Neoplasms, Experimental/drug therapy , Animals , Biomarkers, Tumor/blood , Bone Neoplasms/blood , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Interactions , Female , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Octreotide/administration & dosage , Octreotide/pharmacology , Random Allocation , Receptors, Somatostatin/metabolism
18.
J BUON ; 21(4): 764-779, 2016.
Article in English | MEDLINE | ID: mdl-27685895

ABSTRACT

Conventional drug design embraces the "one gene, one drug, one disease" philosophy. Nowadays, new generation of anti- cancer drugs, able to inhibit more than one pathway, is believed to play a major role in contemporary anticancer drug research. In this way, polypharmacology, focusing on multi-target drugs, has emerged as a new paradigm in drug discovery. A number of recent successful drugs have in part or in whole emerged from a structure-based research approach. Many advances including crystallography and informatics are behind these successes. Increasing insight into the genetics and molecular biology of cancer has resulted in the identification of an increasing number of potential molecular targets, for anticancer drug discovery and development. These targets can be approached through exploitation of emerging structural biology, "rational" drug design, screening of chemical libraries, or a combination of these methods. The result is the rapid discovery of new anticancer drugs. In this article we discuss the application of molecular modeling, molecular docking and virtual high-throughput screening to multi-targeted anticancer drug discovery. Efforts have been made to employ in silico methods for facilitating the search and design of selective multi-target agents. These computer aided molecular design methods have shown promising potential in facilitating drug discovery directed at selective multiple targets and is expected to contribute to intelligent lead anticancer drugs.


Subject(s)
Antineoplastic Agents/chemistry , Drug Discovery/methods , Molecular Docking Simulation/methods , Small Molecule Libraries/chemistry , Antineoplastic Agents/therapeutic use , Drug Design , High-Throughput Screening Assays , Humans , Models, Molecular , Neoplasms/drug therapy , Small Molecule Libraries/therapeutic use
19.
J BUON ; 21(6): 1337-1358, 2016.
Article in English | MEDLINE | ID: mdl-28039691

ABSTRACT

Conventional drug design embraces the "one gene, one drug, one disease" philosophy. Nowadays, new generation of anticancer drugs, able to inhibit more than one pathway, is believed to play a major role in contemporary anticancer drug research. In this way, polypharmacology, focusing on multi-target drugs, has emerged as a new paradigm in drug discovery. A number of recent successful drugs have in part or in whole emerged from a structure-based research approach. Many advances including crystallography and informatics are behind these successes. In this part II we will review the role and methodology of ligand-, structure- and fragment-based computer-aided drug design computer aided drug desing (CADD), virtual high throughput screening (vHTS), de novo drug design, fragment-based design and structure-based molecular docking, homology modeling, combinatorial chemistry and library design, pharmacophore model chemistry and informatics in modern drug discovery.


Subject(s)
Antineoplastic Agents/pharmacology , Combinatorial Chemistry Techniques , Computer-Aided Design , Drug Design , High-Throughput Screening Assays , Molecular Targeted Therapy , Neoplasms/drug therapy , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Binding Sites , Humans , Ligands , Molecular Docking Simulation , Molecular Structure , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , Protein Conformation , Signal Transduction/drug effects , Structure-Activity Relationship
20.
Food Chem Toxicol ; 74: 45-50, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25239662

ABSTRACT

Crocin, a main constituent of Crocus sativus L (saffron), has been found to inhibit the growth of K-562 human chronic myelogenous leukemia (CML) cells expressing Bcr-Abl protein tyrosine kinase activity. The aim of our study is to investigate the ability of the bioactive saffron's constituents, crocin (CRC) and safranal (SFR), to inhibit the Bcr-Abl protein activity employing an in silico approach, as well as the in vitro effect of these compounds on K-562 growth and gene expression of Bcr-Abl. In silico molecular docking studies revealed that mostly SFR can be attached to Bcr-Abl protein, positioned inside the protein's binding cavity at the same place with the drug used in the treatment of CML, imatinib mesylate (IM). The predicted polar interactions and hydrophobic contacts constructing a hydrophobic cavity inside the active site, explain the observed inhibitory activity. Cytotoxicity experiments showed that SFR and CRC mediate cytotoxic response to K562 cells. In vitro studies on the expression of Bcr-Abl gene revealed that SFR and in a lesser degree IM inhibited the expression of the gene, while in contrast CRC induced an increase. The ultimate goal was to evaluate the existence of a potential antitumor activity of saffron's constituents SFR and CRC.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Cyclohexenes/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Terpenes/therapeutic use , Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Carotenoids/therapeutic use , Cell Line, Tumor , Computer Simulation , Crocus/metabolism , Fusion Proteins, bcr-abl/biosynthesis , Fusion Proteins, bcr-abl/drug effects , Gene Expression/drug effects , Humans , Imatinib Mesylate , Molecular Docking Simulation , Piperazines/therapeutic use , Pyrimidines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...