Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Pharmacol Rev ; 76(1): 142-193, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37940347

ABSTRACT

The neutral amino acid transporter subfamily that consists of six members, consecutively SLC6A15-SLC620, also called orphan transporters, represents membrane, sodium-dependent symporter proteins that belong to the family of solute carrier 6 (SLC6). Primarily, they mediate the transport of neutral amino acids from the extracellular milieu toward cell or storage vesicles utilizing an electric membrane potential as the driving force. Orphan transporters are widely distributed throughout the body, covering many systems; for instance, the central nervous, renal, or intestinal system, supplying cells into molecules used in biochemical, signaling, and building pathways afterward. They are responsible for intestinal absorption and renal reabsorption of amino acids. In the central nervous system, orphan transporters constitute a significant medium for the provision of neurotransmitter precursors. Diseases related with aforementioned transporters highlight their significance; SLC6A19 mutations are associated with metabolic Hartnup disorder, whereas altered expression of SLC6A15 has been associated with a depression/stress-related disorders. Mutations of SLC6A18-SLCA20 cause iminoglycinuria and/or hyperglycinuria. SLC6A18-SLC6A20 to reach the cellular membrane require an ancillary unit ACE2 that is a molecular target for the spike protein of the SARS-CoV-2 virus. SLC6A19 has been proposed as a molecular target for the treatment of metabolic disorders resembling gastric surgery bypass. Inhibition of SLC6A15 appears to have a promising outcome in the treatment of psychiatric disorders. SLC6A19 and SLC6A20 have been suggested as potential targets in the treatment of COVID-19. In this review, we gathered recent advances on orphan transporters, their structure, functions, related disorders, and diseases, and in particular their relevance as therapeutic targets. SIGNIFICANCE STATEMENT: The following review systematizes current knowledge about the SLC6A15-SLCA20 neutral amino acid transporter subfamily and their therapeutic relevance in the treatment of different diseases.


Subject(s)
Amino Acid Transport Systems, Neutral , Amino Acids, Neutral , COVID-19 , Humans , Amino Acid Transport Systems, Neutral/chemistry , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Kidney/metabolism , Amino Acids/metabolism , Amino Acids, Neutral/metabolism , COVID-19/metabolism , Membrane Transport Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
2.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39000444

ABSTRACT

The taurine transporter (TauT, SLC6A6) is a member of the solute carrier 6 (SLC6) family, which plays multiple physiological roles. The SLC6 family is divided into four subfamilies: GABA (γ-aminobutyric acid), monoamine, glycine and neutral amino acid transporters. Proteins from the GABA group, including the taurine transporter, are primarily considered therapeutic targets for treating central nervous system disorders. However, recent studies have suggested that inhibitors of SLC6A6 could also serve as anticancer agents. Overexpression of TauT has been associated with the progression of colon and gastric cancer. The pool of known ligands of this transporter is limited and the exact spatial structure of taurine transporter remains unsolved. Understanding its structure could aid in the development of novel inhibitors. Therefore, we utilized homology modelling techniques to create models of TauT. Docking studies and molecular dynamics simulations were conducted to describe protein-ligand interactions. We compared the obtained information for TauT with literature data on other members of the GABA transporter group. Our in silico analysis allowed us to characterize the transporter structure and point out amino acids crucial for ligand binding: Glu406, Gly62 and Tyr138. The significance of selected residues was confirmed through structural studies of mutants. These results will aid in the development of novel taurine transporter inhibitors, which can be explored as anticancer agents.


Subject(s)
GABA Plasma Membrane Transport Proteins , Membrane Transport Proteins , Molecular Docking Simulation , Molecular Dynamics Simulation , Humans , GABA Plasma Membrane Transport Proteins/metabolism , GABA Plasma Membrane Transport Proteins/chemistry , GABA Plasma Membrane Transport Proteins/genetics , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Ligands , Amino Acid Sequence , Protein Binding
3.
J Enzyme Inhib Med Chem ; 38(1): 2158822, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36629422

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative brain disease. Thus, drugs including donepezil, rivastigmine, and galantamine are not entirely effective in the treatment of this multifactorial disease. The present study evaluates eight derivatives (3a-3h) as candidates with stronger anti-AD potential but with less side effects. Reactive oxygen species (ROS) assays were used to assess oxidative stress which involve in the neurodegeneration. The neuroprotective properties of 3e against oxidative stress were done in three experiments using MTT test. The anti-AD potential was determined based on their anticholinesterase inhibition ability, determined using Ellman's method, Aß aggregation potential according to thioflavin (Th) fluorescence assay, and their antioxidative and anti-inflammatory activities. Compound 3e exhibited moderate cholinesterase inhibition activity (AChE, IC50 = 0.131 µM; BuChE, IC50 = 0.116 µM; SI = 1.13), significant inhibition of Aß(1-42) aggregation (55.7%, at 5 µM) and acceptable neuroprotective activity. Extensive analysis of in vitro and in vivo assays indicates that new cyclopentaquinoline derivatives offer promise as candidates for new anti-AD drugs.


Subject(s)
Alzheimer Disease , Neuroprotective Agents , Humans , Alzheimer Disease/drug therapy , Neuroprotection , Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Oxidative Stress , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
4.
Int J Mol Sci ; 24(4)2023 Feb 14.
Article in English | MEDLINE | ID: mdl-36835201

ABSTRACT

Cancer cells are characterized by uncontrolled growth, proliferation, and impaired apoptosis. Tumour progression could be related to poor prognosis and due to this fact, researchers have been working on novel therapeutic strategies and antineoplastic agents. It is known that altered expression and function of solute carrier proteins from the SLC6 family could be associated with severe diseases, including cancers. These proteins were noticed to play important physiological roles through transferring nutrient amino acids, osmolytes, neurotransmitters, and ions, and many of them are necessary for survival of the cells. Herein, we present the potential role of taurine (SLC6A6) and creatine (SLC6A8) transporters in cancer development as well as therapeutic potential of their inhibitors. Experimental data indicate that overexpression of analyzed proteins could be connected with colon or breast cancers, which are the most common types of cancers. The pool of known inhibitors of these transporters is limited; however, one ligand of SLC6A8 protein is currently tested in the first phase of clinical trials. Therefore, we also highlight structural aspects useful for ligand development. In this review, we discuss SLC6A6 and SLC6A8 transporters as potential biological targets for anticancer agents.


Subject(s)
Membrane Transport Proteins , Neoplasms , Taurine , Creatine/metabolism , Ligands , Membrane Transport Proteins/metabolism , Neoplasms/drug therapy , Taurine/metabolism
5.
Int J Mol Sci ; 23(14)2022 Jul 21.
Article in English | MEDLINE | ID: mdl-35887394

ABSTRACT

Glycine transporters are interesting therapeutic targets as they play significant roles in glycinergic and glutamatergic systems. The search for new selective inhibitors of particular types of glycine transporters (GlyT-1 and GlyT-2) with beneficial kinetics is hampered by limited knowledge about the spatial structure of these proteins. In this study, a pool of homology models of GlyT-1 and GlyT-2 in different conformational states was constructed using the crystal structures of related transporters from the SLC6 family and the recently revealed structure of GlyT-1 in the inward-open state, in order to investigate their binding sites. The binding mode of the known GlyT-1 and GlyT-2 inhibitors was determined using molecular docking studies, molecular dynamics simulations, and MM-GBSA free energy calculations. The results of this study indicate that two amino acids, Gly373 and Leu476 in GlyT-1 and the corresponding Ser479 and Thr582 in GlyT-2, are mainly responsible for the selective binding of ligands within the S1 site. Apart from these, one pocket of the S2 site, which lies between TM3 and TM10, may also be important. Moreover, selective binding of noncompetitive GlyT-1 inhibitors in the intracellular release pathway is affected by hydrophobic interactions with Ile399, Met382, and Leu158. These results can be useful in the rational design of new glycine transporter inhibitors with desired selectivity and properties in the future.


Subject(s)
Glycine Plasma Membrane Transport Proteins , Glycine , Binding Sites , Glycine/pharmacology , Glycine Plasma Membrane Transport Proteins/metabolism , Ligands , Molecular Docking Simulation
6.
Int J Mol Sci ; 23(15)2022 Aug 08.
Article in English | MEDLINE | ID: mdl-35955932

ABSTRACT

Kainate receptors belong to the family of glutamate receptors ion channels, which are responsible for the majority of rapid excitatory synaptic transmission in the central nervous system. The therapeutic potential of kainate receptors is still poorly understood, which is also due to the lack of potent and subunit-selective pharmacological tools. In search of selective ligands for the GluK3 kainate receptor subtype, a series of quinoxaline-2,3-dione analogues was synthesized and pharmacologically characterized at selected recombinant ionotropic glutamate receptors. Among them, compound 28 was found to be a competitive GluK3 antagonist with submicromolar affinity and unprecedented high binding selectivity, showing a 400-fold preference for GluK3 over other homomeric receptors GluK1, GluK2, GluK5 and GluA2. Furthermore, in functional assays performed for selected metabotropic glutamate receptor subtypes, 28 did not show agonist or antagonist activity. The molecular determinants underlying the observed affinity profile of 28 were analyzed using molecular docking and molecular dynamics simulations performed for individual GluK1 and GluK3 ligand-binding domains.


Subject(s)
Receptors, Kainic Acid , Ligands , Molecular Docking Simulation , Protein Domains , Receptors, Kainic Acid/metabolism , GluK3 Kainate Receptor
7.
Molecules ; 28(1)2022 Dec 28.
Article in English | MEDLINE | ID: mdl-36615435

ABSTRACT

The multitarget-directed ligands demonstrating affinity to histamine H3 receptor and additional cholinesterase inhibitory potency represent a promising strategy for research into the effective treatment of Alzheimer's disease. In this study, a novel series of benzophenone derivatives was designed and synthesized. Among these derivatives, we identified compound 6 with a high affinity for H3R (Ki = 8 nM) and significant inhibitory activity toward BuChE (IC50 = 172 nM and 1.16 µM for eqBuChE and hBuChE, respectively). Further in vitro studies revealed that compound 6 (4-fluorophenyl) (4-((5-(piperidin-1-yl)pentyl)oxy)phenyl)methanone) displays moderate metabolic stability in mouse liver microsomes, good permeability with a permeability coefficient value (Pe) of 6.3 × 10-6 cm/s, and its safety was confirmed in terms of hepatotoxicity in the HepG2 cell line. Therefore, we investigated the in vivo activity of compound 6 in the Passive Avoidance Test and the Formalin Test. While compound 6 did not show a statistically significant influence on memory and learning, it showed analgesic properties in both acute (ED50 = 20.9 mg/kg) and inflammatory (ED50 = 17.5 mg/kg) pain.


Subject(s)
Alzheimer Disease , Receptors, Histamine H3 , Mice , Animals , Cholinesterases/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Histamine , Receptors, Histamine H3/metabolism , Cholinesterase Inhibitors/pharmacology , Receptors, Histamine , Ligands , Structure-Activity Relationship
8.
Bioorg Chem ; 107: 104617, 2021 02.
Article in English | MEDLINE | ID: mdl-33444983

ABSTRACT

Two series of novel 1,3,4-thiadiazole-resorcinol conjugates were efficiently synthesized and evaluated as cholinesterases inhibitors. N-Butyl- and N-chlorophenyl-5-amino-1,3,4-thiadiazol-2-yl)benzene-1,3-diols were identified as the most promising compounds of low nanomolar activity against AChE (IC50 = 29-76 nM) and moderate activity against BuChE. The inhibition mechanism studies proved that the compounds are mixed type inhibitors. The docking simulations showed great affinity of the compounds for both enzymes. The modelled amine derivatives exhibited a similar arrangement in the catalytic anionic site of AChE similar to that of tacrine. The thiadiazole ring interacted with Trp84 and the phenyl groups created π-π stacking interactions with the residue - Phe330. The compounds showed better inhibition of the in vitro self-induced Aß (1-42) aggregation than that compared with curcumin as well as antioxidant properties similar to those of quercetin. They exhibited metal ion chelating properties, acceptable cytotoxicity in vitro and favourable ADMET profile determined in silico.


Subject(s)
Cholinesterase Inhibitors/chemistry , Resorcinols/chemistry , Thiadiazoles/chemistry , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Antioxidants/chemistry , Binding Sites , Butyrylcholinesterase/chemistry , Butyrylcholinesterase/metabolism , Catalytic Domain , Cell Line , Cell Survival/drug effects , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/pharmacology , Humans , Kinetics , Molecular Docking Simulation , Protein Aggregates/drug effects
9.
Bioorg Chem ; 114: 105129, 2021 09.
Article in English | MEDLINE | ID: mdl-34217977

ABSTRACT

Alzheimer's disease (AD) is a complex and incurable illness that requires the urgent approval of new effective drugs. However, since 2003, no new molecules have shown successful results in clinical trials, thereby making the common "one compound - one target" paradigm questionable. Recently, the multitarget-directed ligand (MTDL) approach has gained popularity, as compounds targeting at least two biological targets may be potentially more effective in treating AD. On the basis of these findings, we designed, synthesized, and evaluated through biological assays a series of derivatives of alicyclic amines linked by an alkoxy bridge to an aromatic lipophilic moiety of [1,1'-biphenyl]-4-carbonitrile. The research results revealed promising biological activity of the obtained compounds toward the chosen targets involved in AD pathophysiology; the compounds showed high affinity (mostly low nanomolar range of Ki values) for human histamine H3 receptors (hH3R) and good nonselective inhibitory potency (micromolar range of IC50 values) against acetylcholinesterase from electric eel (eeAChE) and equine serum butyrylcholinesterase (eqBuChE). Moreover, micromolar/submicromolar potency against human monoamine oxidase B (hMAO B) was detected for some compounds. The study identified compound 5 as a multiple hH3R/eeAChE/eqBuChE/hMAO B ligand (5: hH3R Ki = 9.2 nM; eeAChE IC50 = 2.63 µM; eqBuChE IC50 = 1.30 µM; hMAO B IC50 = 0.60 µM). Further in vitro studies revealed that compound 5 exhibits a mixed type of eeAChE and eqBuChE inhibition, good metabolic stability, and moderate hepatotoxicity effect on HepG2 cells. Finally, compound 5 showed a beneficial effect on scopolamine-induced memory impairments, as assessed by the passive avoidance test, thus revealing the potential of this compound as a promising agent for further optimization for AD treatment.


Subject(s)
Alzheimer Disease/drug therapy , Biphenyl Compounds/pharmacology , Cholinesterase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Receptors, Histamine H3/metabolism , Acetylcholinesterase/metabolism , Alzheimer Disease/metabolism , Animals , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/chemistry , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Dose-Response Relationship, Drug , Electrophorus , Horses , Humans , Ligands , Molecular Structure , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Structure-Activity Relationship
10.
J Enzyme Inhib Med Chem ; 36(1): 437-449, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33467931

ABSTRACT

The pyridinium-2-carbaldoximes with quinolinium carboxamide moiety were designed and synthesised as cholinesterase reactivators. The prepared compounds showed intermediate-to-high inhibition of both cholinesterases when compared to standard oximes. Their reactivation ability was evaluated in vitro on human recombinant acetylcholinesterase (hrAChE) and human recombinant butyrylcholinesterase (hrBChE) inhibited by nerve agent surrogates (NIMP, NEMP, and NEDPA) or paraoxon. In the reactivation screening, one compound was able to reactivate hrAChE inhibited by all used organophosphates and two novel compounds were able to reactivate NIMP/NEMP-hrBChE. The reactivation kinetics revealed compound 11 that proved to be excellent reactivator of paraoxon-hrAChE better to obidoxime and showed increased reactivation of NIMP/NEMP-hrBChE, although worse to obidoxime. The molecular interactions of studied reactivators were further identified by in silico calculations. Molecular modelling results revealed the importance of creation of the pre-reactivation complex that could lead to better reactivation of both cholinesterases together with reducing particular interactions for lower intrinsic inhibition by the oxime.


Subject(s)
Cholinesterase Inhibitors/pharmacology , Pyridinium Compounds/pharmacology , Quinolinium Compounds/pharmacology , Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Molecular Structure , Pyridinium Compounds/chemical synthesis , Pyridinium Compounds/chemistry , Quinolinium Compounds/chemical synthesis , Quinolinium Compounds/chemistry , Recombinant Proteins/metabolism , Structure-Activity Relationship
11.
Molecules ; 26(12)2021 Jun 11.
Article in English | MEDLINE | ID: mdl-34208297

ABSTRACT

Neurodegenerative diseases, e.g., Alzheimer's disease (AD), are a key health problem in the aging population. The lack of effective therapy and diagnostics does not help to improve this situation. It is thought that ligands influencing multiple but interconnected targets can contribute to a desired pharmacological effect in these complex illnesses. Histamine H3 receptors (H3Rs) play an important role in the brain, influencing the release of important neurotransmitters, such as acetylcholine. Compounds blocking their activity can increase the level of these neurotransmitters. Cholinesterases (acetyl- and butyrylcholinesterase) are responsible for the hydrolysis of acetylcholine and inactivation of the neurotransmitter. Increased activity of these enzymes, especially butyrylcholinesterase (BuChE), is observed in neurodegenerative diseases. Currently, cholinesterase inhibitors: donepezil, rivastigmine and galantamine are used in the symptomatic treatment of AD. Thus, compounds simultaneously blocking H3R and inhibiting cholinesterases could be a promising treatment for AD. Herein, we describe the BuChE inhibitory activity of H3R ligands. Most of these compounds show high affinity for human H3R (Ki < 150 nM) and submicromolar inhibition of BuChE (IC50 < 1 µM). Among all the tested compounds, 19 (E153, 1-(5-([1,1'-biphenyl]-4-yloxy)pentyl)azepane) exhibited the most promising in vitro affinity for human H3R, with a Ki value of 33.9 nM, and for equine serum BuChE, with an IC50 of 590 nM. Moreover, 19 (E153) showed inhibitory activity towards human MAO B with an IC50 of 243 nM. Furthermore, in vivo studies using the Passive Avoidance Task showed that compound 19 (E153) effectively alleviated memory deficits caused by scopolamine. Taken together, these findings suggest that compound 19 can be a lead structure for developing new anti-AD agents.


Subject(s)
Acetylcholinesterase/chemistry , Alzheimer Disease/drug therapy , Amines/chemistry , Butyrylcholinesterase/chemistry , Cholinesterase Inhibitors/pharmacology , Monoamine Oxidase/chemistry , Receptors, Histamine H3/metabolism , Acetylcholinesterase/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Butyrylcholinesterase/metabolism , Cell Line , Cholinesterase Inhibitors/chemical synthesis , Humans , Ligands , Male , Mice , Models, Animal , Molecular Docking Simulation , Molecular Structure , Monoamine Oxidase/metabolism , Receptors, Histamine H3/chemistry , Structure-Activity Relationship
12.
J Enzyme Inhib Med Chem ; 35(1): 1944-1952, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33092411

ABSTRACT

Effective therapy of Alzheimer's disease (AD) requires treatment with a combination of drugs that modulate various pathomechanisms contributing to the disease. In our research, we have focused on the development of multi-target-directed ligands - 5-HT6 receptor antagonists and cholinesterase inhibitors - with disease-modifying properties. We have performed extended in vitro (FRET assay) and in cellulo (Escherichia coli model of protein aggregation) studies on their ß-secretase, tau, and amyloid ß aggregation inhibitory activity. Within these multifunctional ligands, we have identified compound 17 with inhibitory potency against tau and amyloid ß aggregation in in cellulo assay of 59% and 56% at 10 µM, respectively, hBACE IC50=4 µM, h5TH6 K i=94 nM, hAChE IC50=26 nM, and eqBuChE IC50=5 nM. This study led to the development of multifunctional ligands with a broad range of biological activities crucial not only for the symptomatic but also for the disease-modifying treatment of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cholinesterase Inhibitors/chemistry , Cholinesterases/metabolism , Receptors, Serotonin/metabolism , tau Proteins/metabolism , Alzheimer Disease/prevention & control , Cholinesterase Inhibitors/metabolism , Drug Design , Escherichia coli , Fluorescence Resonance Energy Transfer , Humans , Ligands , Models, Molecular , Protein Aggregates , Structure-Activity Relationship
13.
Int J Mol Sci ; 21(11)2020 May 26.
Article in English | MEDLINE | ID: mdl-32466601

ABSTRACT

A series of new tetrahydroacridine and 3,5-dichlorobenzoic acid hybrids with different spacers were designed, synthesized, and evaluated for their ability to inhibit both cholinesterase enzymes. Compounds 3a, 3b, 3f, and 3g exhibited selective butyrylcholinesterase (EqBuChE) inhibition with IC50 values ranging from 24 to 607 nM. Among them, compound 3b was the most active (IC50 = 24 nM). Additionally, 3c (IC50 for EeAChE = 25 nM and IC50 for EqBuChE = 123 nM) displayed dual cholinesterase inhibitory activity and was the most active compound against acetylcholinesterase (AChE). Active compound 3c was also tested for the ability to inhibit Aß aggregation. Theoretical physicochemical properties of the compounds were calculated using ACD Labs Percepta and Chemaxon. A Lineweaver-Burk plot and docking study showed that 3c targeted both the catalytic active site (CAS) and the peripheral anionic site (PAS) of AChE. Moreover, 3c appears to possess neuroprotective activity and could be considered a free-radical scavenger. In addition, 3c did not cause DNA damage and was found to be less toxic than tacrine after oral administration; it also demonstrated little inhibitory activity towards hyaluronidase (HYAL), which may indicate that it possesses anti-inflammatory properties. The screening for new in vivo interactions between 3c and known receptors was realized by yeast three-hybrid technology (Y3H).


Subject(s)
Alzheimer Disease/drug therapy , Chlorobenzoates/chemistry , Cholinesterase Inhibitors/chemical synthesis , Neuroprotective Agents/chemical synthesis , Tacrine/analogs & derivatives , Amyloid beta-Protein Precursor/metabolism , Animals , Catalytic Domain , Cell Line, Tumor , Cells, Cultured , Cholinesterase Inhibitors/adverse effects , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Cholinesterases/chemistry , Cholinesterases/metabolism , Free Radical Scavengers/adverse effects , Free Radical Scavengers/chemical synthesis , Free Radical Scavengers/pharmacology , Free Radical Scavengers/therapeutic use , Humans , Hyaluronoglucosaminidase/antagonists & inhibitors , Mice , Molecular Docking Simulation , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Protein Binding , Protein Multimerization/drug effects
14.
Bioorg Chem ; 91: 103136, 2019 10.
Article in English | MEDLINE | ID: mdl-31374521

ABSTRACT

Design, physicochemical and biological studies of novel radioconjugates for the early diagnosis of Alzheimer's disease, based on the newly synthesized tacrine derivatives were performed. Novel tacrine analogues were labeled with technetium-99m and gallium-68. For all obtained radioconjugates ([99mTc]Tc-Hynic-(tricine)2NH(CH2)ntacrine and [68Ga]Ga-DOTA-NH(CH2)9tacrine, where n = 2-9 denotes the number of methylene groups CH2) the studies of physicochemical properties (lipophilicity, stability in the presence of an excess of standard amino acids cysteine or histidine, human serum and in cerebrospinal fluid) were performed. For two selected radioconjugates [99mTc]Tc-Hynic-(tricine)2NH(CH2)9Tac and [68Ga]Ga-DOTA-NH(CH2)9tacrine (characterized with the highest lipophilicity values) the biological tests (inhibition of cholinesterases action, molecular docking and biodistribution studies) have been performed. All novel radioconjugates showed high stability in biological solutions used. Both selected radioconjugates proved to be good inhibitors of cholinesterases and be able to cross the blood-brain barrier. Radioconjugates [99mTc]Tc-Hynic-(tricine)2NH(CH2)9tacrine and [68Ga]Ga-DOTA-NH(CH2)9tacrine fulfil the conditions for application in nuclear medicine. Radiopharmaceutical [68Ga]Ga-DOTA-NH(CH2)9tacrine, due to increased accuracy and improved sensitivity in PET imaging, may be better potential diagnostic tool for early diagnosis of Alzheimer's disease.


Subject(s)
Cholinesterase Inhibitors/pharmacology , Organotechnetium Compounds/pharmacology , Radiopharmaceuticals/pharmacology , Tacrine/analogs & derivatives , Tacrine/pharmacology , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Alzheimer Disease/diagnosis , Animals , Brain/metabolism , Butyrylcholinesterase/chemistry , Butyrylcholinesterase/metabolism , Catalytic Domain , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/metabolism , Drug Stability , Gallium Radioisotopes , Humans , Male , Molecular Docking Simulation , Organotechnetium Compounds/chemical synthesis , Organotechnetium Compounds/metabolism , Protein Binding , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/metabolism , Rats, Wistar , Tacrine/chemical synthesis , Tacrine/metabolism
15.
Bioorg Chem ; 85: 209-220, 2019 04.
Article in English | MEDLINE | ID: mdl-30634096

ABSTRACT

Thymidine phosphorylase (TP) is over expressed in several solid tumors and its inhibition can offer unique target suitable for drug discovery in cancer. A series of 1,2,4-triazoles 3a-3l has been synthesized in good yields and subsequently inhibitory potential of synthesized triazoles 3a-3l against thymidine phosphorylase enzyme was evaluated. Out of these twelve analogs five analogues 3b, 3c, 3f, 3l and 3l exhibited a good inhibitory potential against thymidine phosphorylase. Inhibitory potential in term of IC50 values were found in the range of 61.98 ±â€¯0.43 to 273.43 ±â€¯0.96 µM and 7-Deazaxanthine was taken as a standard inhibitor with IC50 = 38.68 ±â€¯4.42 µM. Encouraged by these results, more analogues 1,2,4-triazole-3-mercaptocarboxylic acids 4a-4g were synthesized and their inhibitory potential against thymidine phosphorylase was evaluated. In this series, six analogues 4b-4g exhibited a good inhibitory potential in the range of 43.86 ±â€¯1.11-163.43 ±â€¯2.03 µM. Angiogenic response of 1,2,4-triazole acid 4d was estimated using the chick chorionic allantoic membrane (CAM) assay. In the light of these findings, structure activity relationship and molecular docking studies of selected triazoles to determine the key binding interactions was discussed. Docking studies demonstrate that synthesized analogues interacted with active site residues of thymidine phosphorylase enzyme through π-π stacking, thiolate and hydrogen bonding interactions.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Thymidine Phosphorylase/antagonists & inhibitors , Triazoles/pharmacology , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Catalytic Domain , Chickens , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Escherichia coli/enzymology , Hydrogels/chemistry , Molecular Docking Simulation , Molecular Structure , Protein Binding , Structure-Activity Relationship , Thymidine Phosphorylase/chemistry , Thymidine Phosphorylase/metabolism , Tissue Engineering/methods , Triazoles/chemical synthesis , Triazoles/metabolism
16.
Bioorg Chem ; 90: 103084, 2019 09.
Article in English | MEDLINE | ID: mdl-31271942

ABSTRACT

In the search for new treatments for complex disorders such as Alzheimer's disease the Multi-Target-Directed Ligands represent a very promising approach. The aim of the present study was to identify multifunctional compounds among several series of non-imidazole histamine H3 receptor ligands, derivatives of 1-[2-thiazol-5-yl-(2-aminoethyl)]-4-n-propylpiperazine, 1-[2-thiazol-4-yl-(2-aminoethyl)]-4-n-propylpiperazine and 1-phenoxyalkyl-4-(amino)alkylopiperazine using in vitro and in vivo pharmacological evaluation and computational studies. Performed in vitro assays showed moderate potency of tested compounds against acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE). Molecular modeling studies have revealed possible interactions between the active compounds and both AChE and BuChE as well as the human H3 histamine receptor. Computational studies showed the high drug-likeness of selected compounds with very good physicochemical profiles. The parallel artificial membrane permeation assay proved outstanding blood-brain barrier penetration in test conditions. The most promising compound, A12, chemically methyl(4-phenylbutyl){2-[2-(4-propylpiperazin-1-yl)-1,3-thiazol-5-yl]ethyl}amine, possesses good balanced multifunctional profile with potency toward studied targets - H3 antagonist activity (pA2 = 8.27), inhibitory activity against both AChE (IC50 = 13.96 µM), and BuChE (IC50 = 14.62 µM). The in vivo pharmacological studies revealed the anti-amnestic properties of compound A12 in the passive avoidance test on mice.


Subject(s)
Alzheimer Disease/drug therapy , Amnesia/drug therapy , Cholinesterase Inhibitors/pharmacology , Disease Models, Animal , Piperazines/chemistry , Receptors, Histamine H3/metabolism , Acetylcholinesterase/chemistry , Adjuvants, Anesthesia/toxicity , Amnesia/chemically induced , Animals , Butyrylcholinesterase/chemistry , Cholinesterase Inhibitors/chemistry , Computational Biology , In Vitro Techniques , Ligands , Male , Mice , Models, Molecular , Molecular Structure , Receptors, Histamine H3/chemistry , Scopolamine/toxicity , Structure-Activity Relationship
17.
Int J Mol Sci ; 20(3)2019 Jan 24.
Article in English | MEDLINE | ID: mdl-30678364

ABSTRACT

Here we report the two-step synthesis of 8 new cyclopentaquinoline derivatives as modifications of the tetrahydroacridine structure. Next, the biological assessment of each of them was performed. Based on the obtained results we identified 6-chloro-N-[2-(2,3-dihydro-1H-cyclopenta[b]quinolin-9-ylamino)-hexyl]]-nicotinamide hydrochloride (3e) as the most promising compound with inhibitory potencies against EeAChE and EqBuChE in the low nanomolar level 67 and 153 nM, respectively. Moreover, 3e compound is non-hepatotoxic, able to inhibit amyloid beta aggregation, and shows a mix-type of cholinesterase's inhibition. The mixed type of inhibition of the compound was confirmed by molecular modeling. Then, yeast three-hybrid (Y3H) technology was used to confirm the known ligand-receptor interactions. New derivatives do not show antioxidant activity (confirmed by the use of two different tests). A pKa assay method was developed to identify the basic physicochemical properties of 3e compound. A LogP assay confirmed that 3e compound fulfills Lipinsky's rule of five.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/chemical synthesis , Neuroprotective Agents/chemical synthesis , Quinolines/chemistry , Amyloid beta-Peptides/metabolism , Cell Line , Cell Line, Tumor , Cholinesterase Inhibitors/pharmacology , Humans , Molecular Docking Simulation , Neuroprotective Agents/pharmacology , Protein Binding
18.
Electrophoresis ; 39(19): 2446-2453, 2018 10.
Article in English | MEDLINE | ID: mdl-30051931

ABSTRACT

Discovering hit compounds and optimization processes in medicinal chemistry nowadays could be improved by predictive tools, based on the relationship between structure of molecules and lipophilic properties. Lipophilicity of drug candidate can affect both the pharmacokinetic and pharmacodynamics properties, in particular, the ability of a molecule to cross the cell membrane. Among the new methods for determination of the lipophilicity of compounds, micellar electrokinetic chromatography (MEKC) is considered to be an appropriate one for bioactive molecules, as it closely mimics the physiological conditions. In this paper MEKC was used for the estimation of the lipophilicity of 24 derivatives of 8-alkoxy-7H-purine-2,6-dione, designed and synthesized as potential antidepressant/anxiolytic and antipsychotic agents. The results of experimental method were compared with calculated in silico parameters (AlogPs and milogP by Virtual Computational Laboratory website, log PPallas by Pallas 3.1, Mlog P by Marvin, log PChemS by ChemSketch, log PChemDraw by ChemBioUltra) using Principal Component Analysis (PCA) method. Finally, using estimated log P values for selected compounds ligand - lipophilicity efficiency (LLE), per cent efficiency index (PEI), and binding efficiency index (BEI) parameters were calculated. Applied MEKC procedure could be used for selection of potential lead structure in a group of 7H-purine-2,6-dione derivatives.


Subject(s)
Chromatography, Micellar Electrokinetic Capillary/methods , Psychotropic Drugs/chemistry , Xanthines/chemistry , Drug Discovery , Hydrophobic and Hydrophilic Interactions , Ligands , Linear Models , Psychotropic Drugs/analysis , Psychotropic Drugs/pharmacokinetics , Xanthines/analysis , Xanthines/pharmacokinetics
19.
Bioorg Med Chem Lett ; 28(23-24): 3596-3600, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30404719

ABSTRACT

The synthesis and biological activity of 4-tert-pentylphenoxypropyl derivatives are described in this manuscript. All compounds (except one) showed human histamine H3 receptor affinity with Ki values below 760 nM. The inhibitory activity toward human monoamine oxidase B (hMAO B) was evaluated using a fluorometric Amplex-Red assay, and most of the compounds were effective in the submicromolar range. Among them, 1-(3-(4-tert-pPentylphenoxy)propyl)pyrrolidine (5) exhibited hMAO B inhibitory activity with an IC50 value of 4.5 nM. In addition, hMAO B inhibition by 5 was shown to be non-competitive and reversible. Further, recently described potent histamine H3 receptor ligands - 4-tert-pentylphenoxyalkyl derivatives (with a 4-8 carbon spacer) - were evaluated for hMAO B inhibitory activity, and some of them displayed activity in the submicromolar range. Selected compounds were also tested for human MAO A (hMAO A) inhibitory potencies and exhibited no activity. Moreover, molecular modeling studies were carried out for tested compounds to explain their molecular mechanism of hMAO B inhibition and the selectivity of compounds for hMAO B over hMAO A.


Subject(s)
Ligands , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase/metabolism , Phenols/chemistry , Receptors, Histamine H3/metabolism , Binding Sites , Catalytic Domain , Humans , Inhibitory Concentration 50 , Kinetics , Molecular Docking Simulation , Monoamine Oxidase/chemistry , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/metabolism , Phenols/chemical synthesis , Phenols/metabolism , Receptors, Histamine H3/chemistry , Structure-Activity Relationship
20.
Bioorg Chem ; 78: 29-38, 2018 08.
Article in English | MEDLINE | ID: mdl-29529519

ABSTRACT

Selective butyrylcholinesterase inhibitors could be the promising drug candidates, used in treatment of Alzheimer's disease. The study describes the synthesis and biological activity of novel carbamate derivatives with N-phenylpiperazine, N-benzylpiperazine and 4-benzylpiperidine moieties. Biological studies revealed that most of these compounds displayed significant activity against BuChE. Compound 16 (3-(4-phenyl-piperazin-1-ylmethyl)-phenyl phenylcarbamate) turned out to be the most active (IC50 = 2.00 µM for BuChE). For all synthesized compounds lipophilicity and other physicochemical properties were calculated using computer programs. Relationship between these properties and activity was also checked. Binding mode with enzyme and the ensuing differences in activity were explained by the molecular modeling studies.


Subject(s)
Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Carbamates/pharmacology , Cholinesterase Inhibitors/pharmacology , Animals , Carbamates/chemical synthesis , Carbamates/chemistry , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Dose-Response Relationship, Drug , Electrophorus , Horses , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL