Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 229
Filter
Add more filters











Publication year range
1.
Org Biomol Chem ; 22(30): 6189-6197, 2024 07 31.
Article in English | MEDLINE | ID: mdl-39027944

ABSTRACT

A series of chromone-deferiprone hybrids were designed, synthesized, and evaluated as inhibitors of human monoamine oxidase B (hMAO-B) with iron-chelating activity for the treatment of Alzheimer's disease (AD). The majority exhibited moderate inhibitory activity towards hMAO-B and potent iron-chelating properties. Particularly, compound 25c demonstrated remarkable selectivity against hMAO-B with an IC50 value of 1.58 µM and potent iron-chelating ability (pFe3+ = 18.79) comparable to that of deferiprone (pFe3+ = 17.90). Molecular modeling and kinetic studies showed that 25c functions as a non-competitive hMAO-B inhibitor. According to the predicted results, compound 25c can penetrate the blood-brain barrier (BBB). Additionally, it has been proved to display significant antioxidant activity and the ability to inhibit neuronal ferroptosis. More importantly, compound 25c reduced the cognitive impairment induced by scopolamine and showed significant non-toxicity in short-term toxicity assays. In summary, compound 25c was identified as a potential anti-AD agent with hMAO-B inhibitory, iron-chelating and anti-ferroptosis activities.


Subject(s)
Alzheimer Disease , Chromones , Deferiprone , Iron Chelating Agents , Monoamine Oxidase Inhibitors , Monoamine Oxidase , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/chemical synthesis , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Iron Chelating Agents/pharmacology , Iron Chelating Agents/chemistry , Iron Chelating Agents/chemical synthesis , Deferiprone/pharmacology , Deferiprone/chemistry , Monoamine Oxidase/metabolism , Humans , Chromones/chemistry , Chromones/pharmacology , Chromones/chemical synthesis , Structure-Activity Relationship , Animals , Antioxidants/pharmacology , Antioxidants/chemistry , Antioxidants/chemical synthesis , Ferroptosis/drug effects , Molecular Structure , Molecular Docking Simulation , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Dose-Response Relationship, Drug
2.
J Med Chem ; 67(11): 8630-8641, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38747630

ABSTRACT

A novel Fe(III) complex, Fe-tBPCDTA, was synthesized and explored as a potential contrast agent for MRI. Compared to established agents like Fe-EDTA and Fe-tCDTA, Fe-tBPCDTA exhibited moderate relaxivity (r1 = 1.17 s-1·mmol-1) due to its enhanced second-sphere mechanism. It also displayed improved kinetic inertness, lower cytotoxicity, and enhanced redox stability. In vivo studies demonstrated its function as an extracellular fluid agent, providing tumor contrast comparable to that of Gd-DTPA at a higher dosage. Complete renal clearance occurred within 24 h. These findings suggest Fe-tBPCDTA as a promising candidate for further development as a safe and effective extracellular MRI contrast agent.


Subject(s)
Contrast Media , Iron Chelating Agents , Magnetic Resonance Imaging , Organophosphonates , Contrast Media/chemistry , Contrast Media/chemical synthesis , Magnetic Resonance Imaging/methods , Animals , Humans , Organophosphonates/chemistry , Organophosphonates/chemical synthesis , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacology , Iron Chelating Agents/chemical synthesis , Ferric Compounds/chemistry , Mice , Cell Line, Tumor , Chelating Agents/chemistry , Chelating Agents/chemical synthesis
3.
Bioorg Chem ; 113: 105013, 2021 08.
Article in English | MEDLINE | ID: mdl-34062405

ABSTRACT

AD is a progressive brain disorder. Because of the lack of remarkable single-target drugs against neurodegenerative disorders, the multitarget-directed ligand strategy has received attention as a promising therapeutic approach. Herein, we rationally designed twenty-nine hybrids of N-propargylamine-hydroxypyridinone. The designed hybrids possessed excellent iron-chelating activity (pFe3+ = 17.09-22.02) and potent monoamine oxidase B inhibitory effects. Various biological evaluations of the optimal compound 6b were performed step by step, including inhibition screening of monoamine oxidase (hMAO-B IC50 = 0.083 ± 0.001 µM, hMAO-A IC50 = 6.11 ± 0.08 µM; SI = 73.5), prediction of blood-brain barrier permeability and mouse behavioral research. All of these favorable results proved that the N-propargylamine-hydroxypyridinone scaffold is a promising structure for the discovery of multitargeted ligands for AD therapy.


Subject(s)
Monoamine Oxidase Inhibitors/chemistry , Pargyline/analogs & derivatives , Propylamines/chemistry , Pyridines/chemistry , Alzheimer Disease/drug therapy , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Disease Models, Animal , Drug Design , Drug Stability , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , Iron Chelating Agents/chemical synthesis , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Maze Learning/drug effects , Mice , Mice, Inbred ICR , Monoamine Oxidase/chemistry , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/therapeutic use , Pargyline/chemistry , Structure-Activity Relationship
4.
J Biol Inorg Chem ; 26(4): 467-478, 2021 06.
Article in English | MEDLINE | ID: mdl-33963933

ABSTRACT

The blood-brain barrier (BBB) permeability of molecules needs to meet stringent requirements of Lipinski's rule, which pose a difficulty for the rational design of efficient chelating agents for Parkinson's disease chelation therapy. Therefore, the iron chelators employed N-aliphatic alcohols modification of deferiprone were reasonably designed in this work. The chelators not only meet Lipinski's rule for BBB permeability, but also ensure the iron affinity. The results of solution thermodynamics demonstrated that the pFe3+ value of N-hydroxyalkyl substituted deferiprone is between 19.20 and 19.36, which is comparable to that of clinical deferiprone. The results of 2,2-diphenyl-1-picrylhydrazyl radical scavenging assays indicated that the N-hydroxyalkyl substituted deferiprone also possesses similar radical scavenging ability in comparison to deferiprone. Meanwhile, the Cell Counting Kit-8 assays of neuron-like rat pheochromocytoma cell-line demonstrated that the N-hydroxyalkyl substituted deferiprone exhibits extremely low cytotoxicity and excellent H2O2-induced oxidative stress protection effect. These results indicated that N-hydroxyalkyl substituted deferiprone has potential application prospects as chelating agents for Parkinson's disease chelation therapy strategy.


Subject(s)
Deferiprone/analogs & derivatives , Deferiprone/chemical synthesis , Iron Chelating Agents/chemical synthesis , Iron Chelating Agents/pharmacology , Parkinson Disease/drug therapy , Animals , Calcium , Cell Survival/drug effects , Deferiprone/pharmacology , Drug Design , Hydrogen Peroxide/toxicity , Oxidative Stress/drug effects , PC12 Cells , Rats , Zinc
5.
Biometals ; 34(2): 259-275, 2021 04.
Article in English | MEDLINE | ID: mdl-33389339

ABSTRACT

Alzheimer's Disease (AD) is a complex neurodegenerative disorder associated in some instances with dyshomeostasis of redox-active metal ions, such as copper and iron. In this work, we investigated whether the conjugation of various aromatic amines would improve the pharmacological efficacy of the iron chelator desferrioxamine (DFO). Conjugates of DFO with aniline (DFOANI), benzosulfanylamide (DFOBAN), 2-naphthalenamine (DFONAF) and 6-quinolinamine (DFOQUN) were obtained and their properties examined. DFOQUN had good chelating activity, promoted a significant increase in the inhibition of ß-amyloid peptide aggregation when compared to DFO, and also inhibited acetylcholinesterase (AChE) activity both in vitro and in vivo (Caenorhabditis elegans). These data indicate that the covalent conjugation of a strong iron chelator to an AChE inhibitor offers a powerful approach for the amelioration of iron-induced neurotoxicity symptoms.


Subject(s)
Amines/pharmacology , Antioxidants/pharmacology , Caenorhabditis elegans/drug effects , Cholinesterase Inhibitors/pharmacology , Deferoxamine/pharmacology , Iron Chelating Agents/pharmacology , Acetylcholinesterase/metabolism , Amines/chemistry , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Antioxidants/chemical synthesis , Antioxidants/chemistry , Biphenyl Compounds/antagonists & inhibitors , Caenorhabditis elegans/enzymology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Deferoxamine/chemistry , Humans , Iron Chelating Agents/chemical synthesis , Iron Chelating Agents/chemistry , Molecular Structure , Picrates/antagonists & inhibitors , Protein Aggregates/drug effects
6.
J Biol Inorg Chem ; 26(1): 29-41, 2021 02.
Article in English | MEDLINE | ID: mdl-33156416

ABSTRACT

In the current study, the synthesis of a theranostic platform composed of superparamagnetic iron oxide nanoparticles (SPION)-deferasirox conjugates targeted with AS1411 DNA aptamer was reported. In this regard, SPION was amine-functionalized by (3-aminopropyl)trimethoxysilane (ATPMS), and then deferasirox was covalently conjugated onto its surface. Finally, to provide guided drug delivery to cancerous tissue, AS1411 aptamer was conjugated to the complex of SPION-deferasirox. The cellular toxicity assay on CHO, C-26 and AGS cell lines verified higher cellular toxicity of targeted complex in comparison with non-targeted one. The evaluation of in vivo tumor growth inhibitory effect in C26 tumor-bearing mice illustrated that the aptamer-targeted complex significantly enhanced the therapeutic outcome in comparison with both non-targeted complex and free drug. The diagnostic capability of the prepared platform was also evaluated implementing C26-tumor-bearing mice. Obtained data confirmed higher tumor accumulation and higher tumor residence time for targeted complex through MRI imaging due to the existence of SPION as a contrast agent in the core of the prepared complex. The prepared multimodal theranostic system provides a safe and effective platform for fighting against cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Contrast Media/therapeutic use , Deferasirox/therapeutic use , Iron Chelating Agents/therapeutic use , Magnetite Nanoparticles/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Aptamers, Nucleotide/chemistry , CHO Cells , Cell Line, Tumor , Contrast Media/chemical synthesis , Cricetulus , Deferasirox/chemistry , Female , Humans , Immobilized Nucleic Acids/chemistry , Immobilized Nucleic Acids/therapeutic use , Iron Chelating Agents/chemical synthesis , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Magnetite Nanoparticles/chemistry , Mice, Inbred BALB C , Precision Medicine , Propylamines/chemistry , Silanes/chemistry
7.
Bioorg Chem ; 108: 104564, 2021 03.
Article in English | MEDLINE | ID: mdl-33353806

ABSTRACT

A series of active hybrids combining 3-hydroxypyridin-4(1H)-one and coumarin pharmacophores were designed and synthesized as potential agents for the treatment of Alzheimer's disease (AD). All the compounds exhibited excellent iron-chelating activities (pFe3+ = 14.8-19.2) and showed favorable monoamine oxidase B (MAO-B) inhibitory effects compared to the reference drug Pargyline (IC50 = 86.9 nM). Among them, compound 11 g displayed the best MAO-B inhibitory activity with an IC50 value of 99.3 nM. Molecular docking analysis showed that compound 11 g could enter the entrance cavity and substrate cavity of MAO-B. Furthermore, the compound 11 g had an excellent antioxidant effect and was capable of protecting from the amyloid-ß1-42 (Aß1-42) induced PC12 cell damage. In silico tools were applied for predicting the blood-brain barrier (BBB) penetration and compound 11 g was proved to overcome the brain exposure challenge. In the mice behavioral study, compound 11 g significantly ameliorated cognitive impairment induced by Scopolamine. More importantly, compound 11 g displayed favorable pharmacokinetic profiles in a rat model. In summary, compound 11 g, with both anti-MAO-B and iron-chelating ability, was proved to be a promising potential anti-AD agent for further optimization.


Subject(s)
Alzheimer Disease/drug therapy , Drug Design , Iron Chelating Agents/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Alzheimer Disease/metabolism , Animals , Dose-Response Relationship, Drug , Humans , Iron Chelating Agents/chemical synthesis , Iron Chelating Agents/chemistry , Maze Learning/drug effects , Mice , Mice, Inbred ICR , Models, Molecular , Molecular Structure , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , PC12 Cells , Rats , Structure-Activity Relationship
8.
Int J Mol Sci ; 21(20)2020 Oct 12.
Article in English | MEDLINE | ID: mdl-33053658

ABSTRACT

Siderophores are iron-complexing compounds synthesized by bacteria and fungi. They are low molecular weight compounds (500-1500 Daltons) possessing high affinity for iron(III). Since 1970 a large number of siderophores have been characterized, the majority using hydroxamate or catecholate as functional groups. The biosynthesis of siderophores is typically regulated by the iron levels of the environment where the organism is located. Because of their exclusive affinity and specificity for iron(III), natural siderophores and their synthetic derivatives have been exploited in the treatment of human iron-overload diseases, as both diagnostic and therapeutic agents. Here, solid-phase approach for the preparation of hexadentate, peptide-based tricatecholato containing peptides is described. The versatility of the synthetic method allows for the design of a common scaffolding structure whereby diverse ligands can be conjugated. With so many possibilities, a computational approach has been developed which will facilitate the identification of those peptides which are capable of providing a high affinity iron(III) binding site. This study reports an integrated computational/synthetic approach towards a rational development of peptide-based siderophores.


Subject(s)
Iron Chelating Agents/chemistry , Iron/chemistry , Siderophores/chemistry , Solid-Phase Synthesis Techniques , Binding Sites , Ferric Compounds/chemistry , Humans , Iron Chelating Agents/chemical synthesis , Ligands , Molecular Structure
9.
Biometals ; 33(4-5): 255-267, 2020 10.
Article in English | MEDLINE | ID: mdl-32979113

ABSTRACT

The nematode Caenorhabditis elegans (C. elegans) is a convenient tool to evaluate iron metabolism as it shares great orthology with human proteins involved in iron transport, in addition to being transparent and readily available. In this work, we describe how wild-type (N2) C. elegans nematodes in the first larval stage can be loaded with acetomethoxycalcein (CAL-AM) and study it as a whole-organism model for both iron speciation and chelator permeability of the labile iron pool (LIP). This model may be relevant for high throughput assessment of molecules intended for chelation therapy of iron overload diseases.


Subject(s)
Fluorometry , Iron Chelating Agents/chemistry , Animals , Caenorhabditis elegans , Iron Chelating Agents/chemical synthesis , Molecular Structure
10.
ACS Appl Mater Interfaces ; 12(34): 37834-37844, 2020 Aug 26.
Article in English | MEDLINE | ID: mdl-32639137

ABSTRACT

Iron is an essential micronutrient for life. Its redox activity is a key component in a plethora of vital enzymatic reactions that take place in processes such as drug metabolism, DNA synthesis, steroid synthesis, gene regulation, and cellular respiration (oxygen transport and the electron transport chain). Bacteria are highly dependent on iron for their survival and growth and have specific mechanisms to acquire iron. Limiting the availability of iron to bacteria, thereby preventing their growth, provides new opportunities to treat infection in the era of the persistent rise of antibiotic-resistant bacteria. In this work, we have developed macromolecular iron chelators, conjugates of a high-affinity iron chelator (HBEDS) with polyglycerol, in an attempt to sequester iron uptake by bacteria to limit their growth in order to enhance antibiotic activity. The new macromolecular chelators are successful in slowing the growth of Staphylococcus aureus and worked as an efficient bacteriostatic against S. aureus. Further, these cytocompatible macrochelators acted as effective adjuvants to prevent bacterial growth when used in conjunction with antibiotics. The adjuvant activity of the macrochelators depends on their molecular weight and the chelator density on these molecules. These selective macro iron(III) chelators are highly efficient in growth inhibition and killing of methicillin-resistant S. aureus in conjunction with a low concentration of rifampicin.


Subject(s)
Anti-Bacterial Agents/chemistry , Glycerol/chemistry , Iron Chelating Agents/chemistry , Polymers/chemistry , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Line , Cell Survival/drug effects , Humans , Iron Chelating Agents/chemical synthesis , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Staphylococcus aureus/drug effects
11.
Bioorg Med Chem ; 28(12): 115550, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32503694

ABSTRACT

A series of (3-hydroxypyridin-4-one)-coumarin hybrids were developed and investigated as potential multitargeting candidates for the treatment of Alzheimer's disease (AD) through the incorporation of iron-chelating and monoamine oxidase B (MAO-B) inhibition. This combination endowed the hybrids with good capacity to inhibit MAO-B as well as excellent iron-chelating effects. The pFe3+ values of the compounds were ranging from 16.91 to 20.16, comparable to more potent than the reference drug deferiprone (DFP). Among them, compound 18d exhibited the most promising activity against MAO-B, with an IC50 value of 87.9 nM. Moreover, compound 18d exerted favorable antioxidant activity, significantly reversed the amyloid-ß1-42 (Aß1-42) induced PC12 cell damage. More importantly, 18d remarkably ameliorated the cognitive dysfunction in a scopolamine-induced mice AD model. In brief, a series of hybrids with potential anti-AD effect were successfully obtained, indicating that the design of iron chelators with MAO-B inhibitory and antioxidant activities is an attractive strategy against AD progression.


Subject(s)
Antioxidants/chemistry , Drug Design , Iron Chelating Agents/chemical synthesis , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase/metabolism , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Animals , Behavior, Animal/drug effects , Binding Sites , Cell Survival/drug effects , Coumarins/chemistry , Disease Models, Animal , Humans , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Mice , Mice, Inbred ICR , Molecular Docking Simulation , Monoamine Oxidase/chemistry , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/therapeutic use , PC12 Cells , Peptide Fragments/pharmacology , Rats , Structure-Activity Relationship
12.
Eur J Med Chem ; 198: 112350, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32380385

ABSTRACT

In order to obtain multi-functional molecules for Alzheimer's disease, a series of deferiprone derivatives has been synthesized and evaluated in vitro with the hypothesis that they can restore the cholinergic tone and attenuate the dyshomeostasis of the metals mainly involved in the pathology. These compounds were designed as dual binding site AChE inhibitors: they possess an arylalkylamine moiety connected via an alkyl chain to a 3-hydroxy-4-pyridone fragment, to allow the simultaneous interaction with catalytic active site (CAS) and peripheral anionic site (PAS) of the enzyme. Deferiprone moiety and 2-aminopyridine, 2-aminopyrimidine or 2,4-diaminopyrimidine groups have been incorporated into these compounds, in order to obtain molecules potentially able to chelate bio-metals colocalized in Aß plaques and involved in the generation of radical species. Synthesized compounds were tested by enzymatic inhibition studies towards EeAChE and eqBChE using Ellman's method. The most potent EeAChE inhibitor is compound 5a, with a Ki of 788 ± 51 nM, while the most potent eqBChE inhibitors are compounds 12 and 19, with Ki values of 182 ± 18 nM and 258 ± 25 nM respectively. Selected compounds, among the most potent cholinesterases inhibitors, were able to form complex with iron and in some cases with copper and zinc. Moreover, these compounds were characterized by low toxicity on U-87 MG Cell Line from human brain (glioblastoma astrocytoma).


Subject(s)
Acetylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/chemical synthesis , Deferiprone/chemical synthesis , Iron Chelating Agents/chemical synthesis , Amines/chemistry , Amino Acid Sequence , Aminopyridines/chemistry , Catalytic Domain , Cholinesterase Inhibitors/pharmacology , Coordination Complexes/chemistry , Deferiprone/pharmacology , Drug Design , Humans , Iron Chelating Agents/pharmacology , Molecular Docking Simulation , Pyrimidines/chemistry , Structure-Activity Relationship
13.
Bioorg Chem ; 95: 103465, 2020 01.
Article in English | MEDLINE | ID: mdl-31855824

ABSTRACT

Resistance of pathogens to antimicrobials is a major current healthcare concern. In a series of linked studies, we have investigated synthetic iron chelators based on hydroxy-pyridinone ligands as novel bacteriostatic agents. Herein we describe our synthesis of several useful building blocks based on the 1-hydroxy-2(1H)-pyridinone moiety, including a novel formyl derivative, which were combined with a tris(2-aminoethyl)amine core to obtain a series of new high-affinity hexadentate Fe(III) chelators. The design principle examined by this series is the size and flexibility of the linker between the core and the metal ligands. Measurement of the pKa and stability constants (Fe3+ and Cu2+) of representative coordinating groups was performed to help rationalise the biological activity of the chelators. The novel chelators were tested on a panel of representative microorganisms with some effectively inhibiting microbial growth. We demonstrate that the nature and position of the linker between the hydroxypyridinone and the tris(2-aminoethyl)amine core has considerable impact upon microbial growth inhibition and that both amide or amine linkages can give efficacious chelators.


Subject(s)
Amines/pharmacology , Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Iron Chelating Agents/pharmacology , Pyridones/pharmacology , Acinetobacter baumannii/drug effects , Amines/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Bacillus subtilis/drug effects , Candida albicans/drug effects , Dose-Response Relationship, Drug , Iron Chelating Agents/chemical synthesis , Iron Chelating Agents/chemistry , Klebsiella pneumoniae/drug effects , Microbial Sensitivity Tests , Molecular Structure , Pseudomonas aeruginosa/drug effects , Pyridones/chemistry , Staphylococcus aureus/drug effects , Structure-Activity Relationship
14.
J Enzyme Inhib Med Chem ; 34(1): 1489-1497, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31416364

ABSTRACT

MAO-B leads to an increase in the levels of hydrogen peroxide and oxidative free radicals, which contribute to the aetiology of the AD. Thus, both iron ion chelators and MAO-B inhibitors can be used to treat AD. Taking the coumarin derivatives and hydroxypyridinones as the lead compounds, a series of dual-target hybrids were designed and synthesised by Click Chemistry. The compounds were biologically evaluated for their iron ion chelating and MAO-B inhibitory activity. Most of the compounds displayed excellent iron ion chelating activity and moderate to good anti-MAO-B activity. Compounds 27b and 27j exhibited the most potent MAO-B inhibitory activity, with IC50 values of 0.68 and 0.86 µM, respectively. In summary, these dual-target compounds have the potential anti-AD activity.


Subject(s)
Alzheimer Disease/drug therapy , Coumarins/pharmacology , Iron Chelating Agents/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Pyridones/pharmacology , Alzheimer Disease/metabolism , Coumarins/chemical synthesis , Coumarins/chemistry , Dose-Response Relationship, Drug , Humans , Iron Chelating Agents/chemical synthesis , Iron Chelating Agents/chemistry , Molecular Structure , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Pyridones/chemical synthesis , Pyridones/chemistry , Structure-Activity Relationship
15.
Eur J Med Chem ; 180: 367-382, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31325784

ABSTRACT

A series of hybrids of hydroxypyridinone and coumarin were rationally designed, synthesized and biologically evaluated for their iron ion chelating and MAO-B inhibitory activities. Most of the compounds displayed excellent iron ion chelating effects and moderate to good anti-MAO-B activities. Compound 27a exhibited the most potent activity against MAO-B, with an IC50 value of 14.7 nM. Importantly, 27a showed good U251 cell protective effect and significantly ameliorated the cognitive dysfunction of scopolamine-induced AD mice. Moreover, molecular docking was performed to elucidate the probable ligand-receptor interaction, and the structure-activity relationships were also summarized.


Subject(s)
Alzheimer Disease/drug therapy , Coumarins/pharmacology , Iron Chelating Agents/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Pyridines/pharmacology , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Coumarins/chemistry , Dose-Response Relationship, Drug , Drug Design , Humans , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/pharmacology , Iron Chelating Agents/chemical synthesis , Iron Chelating Agents/chemistry , Mice , Mice, Inbred ICR , Molecular Docking Simulation , Molecular Structure , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Pyridines/chemistry , Scopolamine , Structure-Activity Relationship
16.
ChemMedChem ; 14(16): 1484-1492, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31162826

ABSTRACT

Metal dyshomeostasis is central to a number of disorders that result from, inter alia, oxidative stress, protein misfolding, and cholesterol dyshomeostasis. In this respect, metal deficiencies are usually readily corrected by treatment with supplements, whereas metal overload can be overcome by the use of metal-selective chelation therapy. Deferasirox, 4-[(3Z,5E)-3,5-bis(6-oxo-1-cyclohexa-2,4-dienylidene)-1,2,4-triazolidin-1-yl]benzoic acid, Exjade, or ICL670, is used clinically to treat hemosiderosis (iron overload), which often results from multiple blood transfusions. Cyclodextrins are cyclic glucose units that are extensively used in the pharmaceutical industry as formulating agents as well as for encapsulating hydrophobic molecules such as in the treatment of Niemann-Pick type C or for hypervitaminosis. We conjugated deferasirox, via an amide coupling reaction, to both 6A -amino-6A -deoxy-ß-cyclodextrin and 3A -amino-3A -deoxy-2A (S),3A (S)-ß-cyclodextrin, at the upper and lower rim, respectively, creating hybrid molecules with dual properties, capable of both metal chelation and cholesterol encapsulation. Our findings emphasize the importance of the conjugation of ß-cyclodextrin with deferasirox to significantly improve the biological properties and to decrease the cytotoxicity of this drug.


Subject(s)
Antioxidants/pharmacology , Cyclodextrins/pharmacology , Deferasirox/analogs & derivatives , Deferasirox/pharmacology , Iron Chelating Agents/pharmacology , Animals , Antioxidants/chemical synthesis , CHO Cells , Cricetulus , Cyclodextrins/chemical synthesis , Deferasirox/chemical synthesis , Hep G2 Cells , Humans , Iron Chelating Agents/chemical synthesis , Protein Multimerization/drug effects , alpha-Synuclein/metabolism
17.
J Org Chem ; 84(10): 6459-6464, 2019 05 17.
Article in English | MEDLINE | ID: mdl-31039303

ABSTRACT

Here, we report the first total synthesis of hinduchelins A-D, a family of nontoxic catechol derivatives from Streptoalloteichus hindustanus, possessing a druglike chemotype and modest iron-chelating ability. A concise synthesis was developed employing methyl 5-methyloxazole-4-carboxylate as a single starting material to provide hinduchelins A-D (and unnatural analogues) in only four steps and 5-15% overall yields; moreover, the stereochemistry of hinduchelin A was reassigned from ( S) to ( R). Biological evaluation confirmed that natural and unnatural hinduchelins are weak iron chelators (siderophores).


Subject(s)
Catechols/chemistry , Catechols/chemical synthesis , Iron Chelating Agents/chemistry , Iron Chelating Agents/chemical synthesis , Actinobacteria/chemistry , Chemistry Techniques, Synthetic , Stereoisomerism
18.
Bioorg Chem ; 88: 102809, 2019 07.
Article in English | MEDLINE | ID: mdl-30999246

ABSTRACT

Ten-eleven translocation protein (TET) 1 plays a key role in control of DNA demethylation and thereby of gene expression. Dysregulation of these processes leads to serious pathological states such as oncological and neurodegenerative ones and thus TET 1 targeting is highly requested. Therefore, in this work, we examined the ability of hydrazones (acyl-, aroyl- and heterocyclic hydrazones) to inhibit the TET 1 protein and its mechanism of action. Inhibitory activity of hydrazones 1-7 towards TET 1 was measured. The results showed a high affinity of the tested chelators for iron(II). The study clearly showed a significant correlation between the chelator's affinity for iron(II) ions (represented by the binding constant) and TET 1 protein inhibitory activity (represented by IC50 values).


Subject(s)
Dioxygenases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Hydrazones/chemistry , Iron Chelating Agents/chemistry , Dioxygenases/chemistry , Enzyme Assays , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/toxicity , Epigenesis, Genetic/drug effects , Hydrazones/chemical synthesis , Hydrazones/toxicity , Iron/chemistry , Iron Chelating Agents/chemical synthesis , Iron Chelating Agents/toxicity
19.
J Inorg Biochem ; 193: 152-165, 2019 04.
Article in English | MEDLINE | ID: mdl-30769225

ABSTRACT

This work presents the simple and low cost synthesis of a new tripodal ligand, in which three units of kojic acid are coupled to a tris(2-aminoethyl)amine (tren) backbone molecule. The protonation equilibria, together with the complex formation equilibria of this ligand with Fe3+, Al3+, Cu2+ and Zn2+ ions were studied. The complementary use of potentiometric, spectrophotometric and NMR techniques, and of Density Functional Theory (DFT) calculations, has allowed a thorough characterization of the different species involved in equilibrium. The stability of the formed complexes with Fe3+ and Al3+ are high enough to consider the new ligand for further studies for its clinical applications as a chelating agent. Biodistribution studies were carried out to assess the capacity the ligand for mobilization of gallium in 67Ga-citrate injected mice. These studies demonstrated that this ligand efficiently chelates the radiometal in our animal model, which suggests that it can be a promising candidate as sequestering agent of iron and other hard trivalent metal ions. Furthermore, the good zinc complexation capacity appears as a stimulating result taking into a potential use of this new ligand in analytical chemistry as well as in agricultural and environmental applications.


Subject(s)
Iron Chelating Agents/pharmacology , Pyrans/pharmacology , Pyrones/pharmacology , Aluminum/chemistry , Animals , Copper/chemistry , Density Functional Theory , Female , Gallium Radioisotopes/chemistry , Iron/chemistry , Iron Chelating Agents/chemical synthesis , Iron Chelating Agents/pharmacokinetics , Mice , Models, Chemical , Pyrans/chemical synthesis , Pyrans/pharmacokinetics , Pyrones/chemical synthesis , Pyrones/pharmacokinetics , Tissue Distribution , Zinc/chemistry
20.
J Inorg Biochem ; 193: 1-8, 2019 04.
Article in English | MEDLINE | ID: mdl-30654208

ABSTRACT

We synthesized five iron chelator derived from 2,6-diacetylpyridine bis(acylhydrazones) and proved their iron complexes structure by X-ray single crystal diffraction. These ligands have a significant anticancer proliferative activity and low cytotoxicity against normal cells. The Fe(III) complexes show reduced cytotoxic activity compared to the metal-free ligands. Anticancer mechanism studies indicate that ligands with a potential anticancer proliferation activity by inhibiting the activity of ribonucleotide reductase. Ligand rather than iron complexes regulate the expression of cell cycle associated proteins and inhibit cell cycle arrest in S phase. Apoptosis mechanism results showed that both ligand and iron complexes did not significantly promote apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Hydrazones/pharmacology , Iron Chelating Agents/pharmacology , Pyridines/pharmacology , Antigens, CD/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/toxicity , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Ferritins/metabolism , Humans , Hydrazones/chemical synthesis , Hydrazones/toxicity , Iron/chemistry , Iron Chelating Agents/chemical synthesis , Iron Chelating Agents/toxicity , Ligands , Pyridines/chemical synthesis , Pyridines/toxicity , Receptors, Transferrin/metabolism , Ribonucleotide Reductases/antagonists & inhibitors , S Phase Cell Cycle Checkpoints/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL