Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.066
Filter
1.
Molecules ; 29(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38893361

ABSTRACT

A versatile family of quaternary propargylamines was synthesized employing the KA2 multicomponent reaction, through the single-step coupling of a number of amines, ketones, and terminal alkynes. Sustainable synthetic procedures using transition metal catalysts were employed in all cases. The inhibitory activity of these molecules was evaluated against human monoaminoxidase (hMAO)-A and hMAO-B enzymes and was found to be significant. The IC50 values for hMAO-B range from 152.1 to 164.7 nM while the IC50 values for hMAO-A range from 765.6 to 861.6 nM. Furthermore, these compounds comply with Lipinski's rule of five and exhibit no predicted toxicity. To understand their binding properties with the two target enzymes, key interactions were studied using molecular docking, all-atom molecular dynamics (MD) simulations, and MM/GBSA binding free energy calculations. Overall, herein, the reported family of propargylamines exhibits promise as potential treatments for neurodegenerative disorders, such as Parkinson's disease. Interestingly, this is the first time a propargylamine scaffold bearing an internal alkyne has been reported to show activity against monoaminoxidases.


Subject(s)
Alkynes , Molecular Docking Simulation , Molecular Dynamics Simulation , Monoamine Oxidase Inhibitors , Monoamine Oxidase , Pargyline , Alkynes/chemistry , Alkynes/pharmacology , Monoamine Oxidase/metabolism , Monoamine Oxidase/chemistry , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/chemical synthesis , Humans , Pargyline/chemistry , Pargyline/analogs & derivatives , Pargyline/pharmacology , Propylamines/chemistry , Structure-Activity Relationship , Molecular Structure
2.
Future Med Chem ; 15(2): 211-224, 2023 01.
Article in English | MEDLINE | ID: mdl-36802855

ABSTRACT

Propargylamine is a chemical moiety whose properties have made it a widely distributed group within the fields of medicinal chemistry and chemical biology. Its particular reactivity has traditionally popularized the preparation of propargylamine derivatives using a large variety of synthetic strategies, which have facilitated the access to these compounds for the study of their biomedical potential. This review comprehensively covers and analyzes the applications that propargylamine-based derivatives have achieved in the drug discovery field, both from a medicinal chemistry perspective and from a chemical biology-oriented approach. The principal therapeutic fields where propargylamine-based compounds have made an impact are identified, and a discussion of their influence and growing potential is included.


Subject(s)
Drug Discovery , Pargyline , Pargyline/pharmacology , Pargyline/chemistry , Propylamines/chemistry
3.
Int J Mol Sci ; 23(23)2022 Nov 27.
Article in English | MEDLINE | ID: mdl-36499156

ABSTRACT

Serotonin (5-HT) plays an essential role in regulating female reproductive function in many animals. 5-HT accumulates in the mammalian ovary with the involvement of membrane serotonin transporter SERT and is functionally active in the oocytes of growing follicles, but shows almost no activity in follicular cells. In this study, we clarified the interplay between 5-HT membrane transport and its degradation by monoamine oxidase (MAO) in the mammalian ovary. Using pharmacologic agents and immunohistochemical staining of the cryosections of ovaries after serotonin administration in vitro, we demonstrated the activity of transport and degradation systems in ovarian follicles. The MAO inhibitor pargyline increased serotonin accumulation in the granulosa cells of growing follicles, indicating the activity of both serotonin uptake and degradation by MAO in these cells. The activity of MAO and the specificity of the membrane transport of serotonin was confirmed in primary granulosa cell culture treated with pargyline and fluoxetine. Moreover, the accumulation of serotonin is more effective in the denuded oocytes and occurs at lower concentrations than in the oocytes within the follicles. This confirms that the activity of SERT and MAO in the granulosa cells surrounding the oocytes impedes the accumulation of serotonin in the oocytes and forms a functional barrier to serotonin.


Subject(s)
Granulosa Cells , Serotonin , Animals , Mice , Female , Serotonin/metabolism , Granulosa Cells/metabolism , Ovarian Follicle/metabolism , Oocytes/metabolism , Monoamine Oxidase/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Pargyline/metabolism , Pargyline/pharmacology , Mammals/metabolism
4.
J Physiol Sci ; 72(1): 27, 2022 Oct 26.
Article in English | MEDLINE | ID: mdl-36289481

ABSTRACT

To investigate the roles of the serotonin (5-HT) transporter (SERT) and plasma membrane monoamine transporter (PMAT) in 5-HT uptake and its metabolism in the heart, we monitored myocardial interstitial levels of 5-HT and 5-HIAA, a metabolite of 5-HT by monoamine oxidase (MAO), in anesthetized rats using a microdialysis technique. Fluoxetine (SERT inhibitor), decynium-22 (PMAT inhibitor), or their mixture was locally administered by reverse-microdialysis for 60 min. Subsequently, pargyline (MAO inhibitor) was co-administered. Fluoxetine rapidly increased dialysate 5-HT concentration, while decynium-22 gradually increased it. The mixture induced a larger increase in dialysate 5-HT concentration compared to fluoxetine or decynium-22 alone. Fluoxetine increased dialysate 5-HIAA concentration, and this increase was abolished by pargyline. Decynium-22 and the mixture did not change dialysate 5-HIAA concentration, which were not affected by pargyline. Both SERT and PMAT regulate myocardial interstitial 5-HT levels by its uptake; however, 5-HT uptake via PMAT leads to 5-HT metabolism by MAO.


Subject(s)
Monoamine Oxidase Inhibitors , Serotonin , Animals , Rats , Dialysis Solutions , Fluoxetine/pharmacology , Hydroxyindoleacetic Acid/metabolism , Membrane Transport Proteins , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Pargyline/pharmacology , Serotonin/metabolism , Heart
5.
J Med Chem ; 64(24): 17824-17845, 2021 12 23.
Article in English | MEDLINE | ID: mdl-34908406

ABSTRACT

Pragmatic insertion of pargyline, a LSD1 inhibitor, as a surface recognition part in the HDAC inhibitory pharmacophore was planned in pursuit of furnishing potent antiprostate cancer agents. Resultantly, compound 14 elicited magnificent cell growth inhibitory effects against the PC-3 and DU-145 cell lines and led to remarkable suppression of tumor growth in human prostate PC-3 and DU-145 xenograft nude mouse models. The outcome of the enzymatic assays ascertained that the substantial antiproliferative effects of compound 14 were mediated through HDAC6 isoform inhibition as well as selective MAO-A and LSD1 inhibition. Moreover, the signatory feature of LSD1 inhibition by 14 in the context of H3K4ME2 accumulation was clearly evident from the results of western blot analysis. Gratifyingly, hydroxamic acid 14 demonstrates good human hepatocytic stability and good oral bioavailability in rats and exhibits enough promise to emerge as a therapeutic for the treatment of prostate cancer in the near future.


Subject(s)
Antineoplastic Agents/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Histone Demethylases/antagonists & inhibitors , Pargyline/pharmacology , Prostatic Neoplasms/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Humans , Male , Pargyline/therapeutic use
6.
Int J Mol Sci ; 22(23)2021 Nov 27.
Article in English | MEDLINE | ID: mdl-34884655

ABSTRACT

The mechanisms of resistance to antidepressant drugs is a key and still unresolved problem of psychopharmacology. Serotonin (5-HT) and brain-derived neurotrophic factor (BDNF) play a key role in the therapeutic effect of many antidepressants. Tryptophan hydroxylase 2 (TPH2) is the rate-limiting enzyme in 5-HT synthesis in the brain. We used zebrafish (Danio rerio) as a promising model organism in order to elucidate the effect of TPH2 deficiency caused by p-chlorophenylalanine (pCPA) on the alterations in behavior and expression of 5-HT-related (Tph2, Slc6a4b, Mao, Htr1aa, Htr2aa) and BDNF-related (Creb, Bdnf, Ntrk2a, Ngfra) genes in the brain after prolonged treatment with two antidepressants, inhibitors of 5-HT reuptake (fluoxetine) and oxidation (pargyline). In one experiment, zebrafish were treated for 72 h with 0.2 mg/L fluoxetine, 2 mg/L pCPA, or the drugs combination. In another experiment, zebrafish were treated for 72 h with 0.5 mg/L pargyline, 2 mg/L pCPA, or the drugs combination. Behavior was studied in the novel tank diving test, mRNA levels were assayed by qPCR, 5-HT and its metabolite concentrations were measured by HPLC. The effects of interaction between pCPA and the drugs on zebrafish behavior were observed: pCPA attenuated "surface dwelling" induced by the drugs. Fluoxetine decreased mRNA levels of Tph2 and Htr2aa genes, while pargyline decreased mRNA levels of Slc6a4b and Htr1aa genes. Pargyline reduced Creb, Bdnf and Ntrk2a genes mRNA concentration only in the zebrafish treated with pCPA. The results show that the disruption of the TPH2 function can cause a refractory to antidepressant treatment.


Subject(s)
Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Brain/drug effects , Fluoxetine/pharmacology , Pargyline/pharmacology , Serotonin/metabolism , Tryptophan Hydroxylase/deficiency , Zebrafish Proteins/deficiency , Animals , Antidepressive Agents/pharmacology , Brain/enzymology , Brain/metabolism , Disease Models, Animal , Monoamine Oxidase Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Tryptophan Hydroxylase/metabolism , Zebrafish , Zebrafish Proteins/metabolism
7.
Bioorg Chem ; 116: 105301, 2021 11.
Article in English | MEDLINE | ID: mdl-34492558

ABSTRACT

A combination of several pharmacophores in one molecule has been successfully used for multi-target-directed ligands (MTDL) design. New propargylamine substituted derivatives combined with salicylic and cinnamic scaffolds were designed and synthesized as potential cholinesterases and monoamine oxidases (MAOs) inhibitors. They were evaluated invitro for inhibition of acetyl- (AChE) and butyrylcholinesterase (BuChE) using Ellman's method. All the compounds act as dual inhibitors. Most of the derivatives are stronger inhibitors of AChE, the best activity showed 5-bromo-N-(prop-2-yn-1-yl)salicylamide 1e (IC50 = 8.05 µM). Carbamates (4-bromo-2-[(prop-2-yn-1-yl)carbamoyl]phenyl ethyl(methyl)carbamate 2d and 2,4-dibromo-6-[(prop-2-yn-1-yl)carbamoyl]phenyl ethyl(methyl)carbamate 2e were selective and the most active for BuChE (25.10 and 26.09 µM). 4-Bromo-2-[(prop-2-yn-1-ylimino)methyl]phenol 4a was the most potent inhibitor of MAOs (IC50 of 3.95 and ≈10 µM for MAO-B and MAO-A, respectively) along with a balanced inhibition of both cholinesterases being a real MTDL. The mechanism of action was proposed, and binding modes of the hits were studied by molecular docking on human enzymes. Some of the derivatives also exhibited antioxidant properties. Insilico prediction of physicochemical parameters affirm that the molecules would be active after oral administration and able to reach brain tissue.


Subject(s)
Antioxidants/pharmacology , Cholinesterase Inhibitors/pharmacology , Molecular Docking Simulation , Monoamine Oxidase Inhibitors/pharmacology , Pargyline/analogs & derivatives , Propylamines/pharmacology , Animals , Antioxidants/chemical synthesis , Antioxidants/chemistry , Butyrylcholinesterase/metabolism , Cells, Cultured , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Cholinesterases/metabolism , Dose-Response Relationship, Drug , Electrophorus , Hepatocytes/drug effects , Hepatocytes/metabolism , Horses , Humans , Male , Molecular Structure , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Pargyline/chemical synthesis , Pargyline/chemistry , Pargyline/pharmacology , Propylamines/chemical synthesis , Propylamines/chemistry , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 45: 128135, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34044119

ABSTRACT

Twenty six propargylamine mycophenolate analogues were designed and synthesized from mycophenolic acid 1 employing a key step A3-coupling reaction. Their cytotoxic activity was examined against six cancer cell lines. Compounds 6a, 6j, 6t, 6u, and 6z exhibited selective cytotoxicity towards neuroblastoma (SH-SY5Y) cancer cells and were less toxic to normal cells in comparison to the lead compound, MPA 1 and a standard drug, ellipticine. Molecular docking results suggested that compound 6a is fit well in the key amino acid of three proteins (CDK9, EGFR, and VEGFR-2) as targets in cancer therapy. The propargylamine mycophenolate scaffold might be a valuable starting point for development of new neuroblastoma anticancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Mycophenolic Acid/pharmacology , Neuroblastoma/drug therapy , Pargyline/analogs & derivatives , Propylamines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Docking Simulation , Molecular Structure , Mycophenolic Acid/chemical synthesis , Mycophenolic Acid/chemistry , Neuroblastoma/pathology , Pargyline/chemical synthesis , Pargyline/chemistry , Pargyline/pharmacology , Propylamines/chemical synthesis , Propylamines/chemistry , Structure-Activity Relationship
9.
Int J Biol Macromol ; 165(Pt A): 767-775, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32991899

ABSTRACT

The magnetically isolable nanobiocomposites have significant impact as the modified new generation catalysts in recent days. This has persuaded us to design and synthesis of a novel Ag NPs decorated biguanidine-chitosan (Bigua-CS) dual biomolecular functionalized core-shell type magnetic nanocomposite (Ag/Bigua-CS@Fe3O4). Bigua-CS could be introducing polysaccharide materials as potential coating agent to immobilizing and stabilizing metal nanoparticles. The material was characterized using several advanced techniques like fourier transformed infrared spectroscopy (FT-IR), inductively coupled plasma (ICP), field emission scanning electron microscopy (FE-SEM), energy dispersive X-ray spectroscopy (EDX), atomic mapping, high resolution transmission electron microscopy (HR-TEM), vibrating sample magnetometer (VSM) and X-ray diffraction (XRD). Towards the chemical applications of the material, we headed the multicomponent synthesis of diverse propargylamines by A3 coupling in water, which ended up with excellent yields. Due to strong paramagnetism, the catalyst was easily isolable and reused in 9cycles without any leaching and considerable change in reactivity. In addition, the catalyst was engaged in biological assays like study of anti-oxidant properties by DPPH mediated free radical scavenging test using BHT as a reference molecule. Thereafter, on having a significant IC50 value in radical scavenging assay, we extended the bio-application of the catalyst in anticancer study of adenocarcinoma cells of human lungs. The three different cancer cell lines, PC-14, LC-2/ad and HLC-1 were used in this regard. The best result was achieved in the case of PC-14 cell line with strong IC50 values.


Subject(s)
Antineoplastic Agents , Chitosan , Coated Materials, Biocompatible , Guanidines , Lung Neoplasms/drug therapy , Magnetite Nanoparticles , Metal Nanoparticles , Silver , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chitosan/chemistry , Chitosan/pharmacology , Coated Materials, Biocompatible/chemical synthesis , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Guanidines/chemistry , Guanidines/pharmacology , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Pargyline/analogs & derivatives , Pargyline/chemical synthesis , Pargyline/chemistry , Pargyline/pharmacology , Propylamines/chemical synthesis , Propylamines/chemistry , Propylamines/pharmacology , Silver/chemistry , Silver/pharmacology
10.
Nat Metab ; 2(9): 934-945, 2020 09.
Article in English | MEDLINE | ID: mdl-32719542

ABSTRACT

Type 1 diabetes (T1D) is caused by the autoimmune destruction of pancreatic beta cells. Pluripotent stem cells can now be differentiated into beta cells, thus raising the prospect of a cell replacement therapy for T1D. However, autoimmunity would rapidly destroy newly transplanted beta cells. Using a genome-scale CRISPR screen in a mouse model for T1D, we show that deleting RNLS, a genome-wide association study candidate gene for T1D, made beta cells resistant to autoimmune killing. Structure-based modelling identified the U.S. Food and Drug Administration-approved drug pargyline as a potential RNLS inhibitor. Oral pargyline treatment protected transplanted beta cells in diabetic mice, thus leading to disease reversal. Furthermore, pargyline prevented or delayed diabetes onset in several mouse models for T1D. Our results identify RNLS as a modifier of beta cell vulnerability and as a potential therapeutic target to avert beta cell loss in T1D.


Subject(s)
CRISPR-Cas Systems , Diabetes Mellitus, Type 1/drug therapy , Genome-Wide Association Study , Insulin-Secreting Cells/drug effects , Monoamine Oxidase/drug effects , Animals , Autoimmunity/drug effects , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Endoplasmic Reticulum Stress , Enzyme Inhibitors/pharmacology , Female , Induced Pluripotent Stem Cells/immunology , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Islets of Langerhans Transplantation , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mutation , Pargyline/pharmacology
11.
Mol Biol (Mosk) ; 54(2): 313-320, 2020.
Article in Russian | MEDLINE | ID: mdl-32392202

ABSTRACT

Striatal-enriched protein tyrosine phosphatase (STEP), which was initially identified in the striatum, is encoded by the Ptpn5 gene and is expressed in neurons of various structures of the brain. STEP is involved in regulating neuroplasticity, and its expression abnormalities are associated with human neurodegenerative disorders. The STEP inhibitor 8-trifluoromethyl-1,2,3,4,5-benzopentathiepin-6-amine hydrochloride (TC-2153) has been shown to affect the serotoninergic system of the brain. However, the influence of the serotoninergic system on the STEP regulation has not been studied yet. The aim of the study was to investigate how pharmacologically induced changes in the brain serotonin (5-HT) level affect Ptpn5 expression and STEP activity in adult male C57BL/6J mice. To modulate the 5-HT level in the brain, the 5-HT synthesis inhibitor p-chlorophenylalanine or 5-HT degradation inhibitor pargyline was administered intraperitoneally for three successive days. Changes in 5-HT concentration in the brain were assayed using high-performance liquid chromatography. The STEP activity was determined spectrophotometrically in the supernatant by the rate of p-nitrophenyl phosphate dephosphorylation in the absence and presence of the selective STEP inhibitor TC-2153. The Ptpn5 mRNA level was determined using quantitative RT-PCR. The Ptpn5 expression level in the striatum was three times higher than in the cortex and hippocampus. Both increases and decreases in brain 5-HT were for the first time associated with a decrease in Ptpn5 mRNA in the striatum. STEP activity in the striatum and cortex was significantly higher than in the hippocampus. However, p-chlorophenylalanine and pargyline did not affect the STEP activity in the brain structures tested. Thus, a new method was proposed to study the STEP activity in the brain and p-chlorophenylalanine and pargyline were shown to decrease Ptpn5 expression in the striatum in mice.


Subject(s)
Alanine/analogs & derivatives , Chloramines/pharmacology , Corpus Striatum/metabolism , Pargyline/pharmacology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Alanine/pharmacology , Animals , Male , Mice , Mice, Inbred C57BL , Neurons , Serotonin
12.
Bioorg Med Chem Lett ; 30(3): 126880, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31864798

ABSTRACT

Current options for the treatment of Alzheimers disease have been restricted to prescription of acetylcholinesterase inhibitors or N-methyl-d-aspartate receptor antagonist, memantine. Propargylamine-derived multi-target directed ligands, such as ladostigil, M30, ASS234 and contilisant, involve different pathways. Apart from acting as inhibitors of both cholinesterases and monoamine oxidases, they show improvement of cognitive impairment, antioxidant activities, enhancement of iron-chelating activities, protect against tau hyperphosphorylation, block metal-associated oxidative stress, regulate APP and Aß expression processing by the non-amyloidogenic α-secretase pathway, suppress mitochondrial permeability transition pore opening, and coordinate protein kinase C signaling and Bcl-2 family proteins. Other hybrid propargylamine derivatives are also reported.


Subject(s)
Neuroprotective Agents/chemistry , Pargyline/analogs & derivatives , Propylamines/chemistry , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Cholinesterases/chemistry , Cholinesterases/metabolism , Humans , Ligands , Monoamine Oxidase/chemistry , Monoamine Oxidase/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Pargyline/chemistry , Pargyline/pharmacology , Pargyline/therapeutic use , Propylamines/pharmacology , Propylamines/therapeutic use
13.
Comput Biol Chem ; 83: 107124, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31563021

ABSTRACT

We have recently explored novel class of potentially anti-breast cancer active enamidines in which four molecules 4a-c and 4h showed higher anticancer activity compared to standard drug doxorubicin. As a part of extension of this work, we have further evaluated in silico cheminformatic studies on bioactivity prediction of synthesized series of enamidines using mole information. The normal cell line study of four lead compounds 4a-c and 4h against African green monkey kidney vero strain further revealed that the compounds complemented good selectivity in inhibition of cancer cells. The in silico bioactivity and molecular docking studies also revealed that the compounds have significant interactions with the drug targets. The results reveal that enamidine moieties are vital for anti-breast cancer activity as they possess excellent drug-like characteristics, being potentially good inhibitors of cyclin dependent kinases7 (CDK7).


Subject(s)
Amines/pharmacology , Antineoplastic Agents/pharmacology , Azides/pharmacology , Breast Neoplasms/drug therapy , Computer Simulation , Cyclin-Dependent Kinases/antagonists & inhibitors , Pargyline/analogs & derivatives , Protein Kinase Inhibitors/pharmacology , Amines/chemical synthesis , Amines/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Azides/chemical synthesis , Azides/chemistry , Binding Sites/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Chlorocebus aethiops , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , Drug Screening Assays, Antitumor , Female , Humans , Hydrogen Bonding , MCF-7 Cells , Molecular Docking Simulation , Molecular Structure , Pargyline/chemical synthesis , Pargyline/chemistry , Pargyline/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Vero Cells , Cyclin-Dependent Kinase-Activating Kinase
14.
Bioorg Med Chem Lett ; 29(18): 2626-2631, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31362921

ABSTRACT

Pyrroline-5-carboxylate reductase 1 (PYCR1) is the final enzyme involved in the biosynthesis of proline and has been found to be upregulated in various forms of cancer. Due to the role of proline in maintaining the redox balance of cells and preventing apoptosis, PYCR1 is emerging as an attractive oncology target. Previous PYCR1 knockout studies led to a reduction in tumor growth. Accordingly, a small molecule inhibitor of PYCR1 could lead to new treatments for cancer, and a focused screening effort identified pargyline as a fragment-like hit. We report the design and synthesis of the first tool compounds as PYCR1 inhibitors, derived from pargyline, which were assayed to assess their ability to attenuate the production of proline. Structural activity studies have revealed the key determinants of activity, with the most potent compound (4) showing improved activity in vitro in enzyme (IC50 = 8.8 µM) and pathway relevant effects in cell-based assays.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Pargyline/pharmacology , Pyrroline Carboxylate Reductases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Pargyline/chemical synthesis , Pargyline/chemistry , Pyrroline Carboxylate Reductases/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , delta-1-Pyrroline-5-Carboxylate Reductase
15.
Eur J Med Chem ; 163: 83-94, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30503945

ABSTRACT

The multifactorial pathophysiology of neurodegenerative disorders remains one of the main challenges in the design of a single molecule that may ultimately prevent the progression of these disorders in affected patients. In this article, we report on twelve novel polycyclic amine cage derivatives, synthesized with or without a propargylamine function, designed to possess inherent multifunctional neuroprotective activity. The MTT cytotoxicity assay results showed the SH-SY5Y human neuroblastoma cells to be viable with the twelve compounds, particularly at concentrations less than 10 µM. The compounds also showed significant neuroprotective activity, ranging from 31% to 61% at 1 µM, when assayed on SH-SY5Y human neuroblastoma cells in which neurodegeneration was induced by MPP+. Calcium regulation assays conducted on the same cell line showed the compounds to be significant VGCC blockers with activity ranging from 26.6% to 51.3% at 10 µM; as well as significant NMDAr antagonists with compound 5 showing the best activity of 88.3% at 10 µM. When assayed on human MAO isoenzymes, most of the compounds showed significant inhibitory activity, with compound 5 showing the best activity (MAO-B: IC50 = 1.70 µM). Generally, the compounds were about 3-52 times more selective to the MAO-B isoenzyme than the MAO-A isoenzyme. Based on the time-dependency studies conducted, the compounds can be defined as reversible MAO inhibitors. Several structure activity relationships were derived from the various assays conducted, and the compounds' possible putative binding modes within the MAO-B enzyme cavity were assessed in silico.


Subject(s)
Neuroprotective Agents/chemical synthesis , Pargyline/analogs & derivatives , Propylamines/pharmacology , Cell Line, Tumor , Drug Design , Humans , Monoamine Oxidase/drug effects , Neuroblastoma/enzymology , Neuroblastoma/pathology , Neuroprotective Agents/pharmacology , Pargyline/chemical synthesis , Pargyline/pharmacology , Propylamines/chemical synthesis , Protein Binding , Structure-Activity Relationship
16.
Biochim Biophys Acta Mol Basis Dis ; 1864(9 Pt B): 3050-3059, 2018 09.
Article in English | MEDLINE | ID: mdl-29953926

ABSTRACT

Monoamine oxidase (MAO), a mitochondrial enzyme that oxidizes biogenic amines generating hydrogen peroxide, is a major source of oxidative stress in cardiac injury. However, the molecular mechanisms underlying its overactivation in pathological conditions are still poorly characterized. Here, we investigated whether the enhanced MAO-dependent hydrogen peroxide production can be due to increased substrate availability using a metabolomic profiling method. We identified N1-methylhistamine -the main catabolite of histamine- as an important substrate fueling MAO in Langendorff mouse hearts, directly perfused with a buffer containing hydrogen peroxide or subjected to ischemia/reperfusion protocol. Indeed, when these hearts were pretreated with the MAO inhibitor pargyline we observed N1-methylhistamine accumulation along with reduced oxidative stress. Next, we showed that synaptic terminals are the major source of N1-methylhistamine. Indeed, in vivo sympathectomy caused a decrease of N1-methylhistamine levels, which was associated with a marked protection in post-ischemic reperfused hearts. As far as the mechanism is concerned, we demonstrate that exogenous histamine is transported into isolated cardiomyocytes and triggers a rise in the levels of reactive oxygen species (ROS). Once again, pargyline pretreatment induced intracellular accumulation of N1-methylhistamine along with decrease in ROS levels. These findings uncover a receptor-independent mechanism for histamine in cardiomyocytes. In summary, our study reveals a novel and important pathophysiological causative link between MAO activation and histamine availability during pathophysiological conditions such as oxidative stress/cardiac injury.


Subject(s)
Heart Ventricles/pathology , Histamine/metabolism , Monoamine Oxidase/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Animals , Disease Models, Animal , Heart Ventricles/cytology , Humans , Isolated Heart Preparation , Male , Metabolomics , Methylhistamines/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Myocardial Reperfusion Injury/etiology , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidation-Reduction , Oxidative Stress , Pargyline/pharmacology , Reactive Oxygen Species/metabolism
17.
Biopharm Drug Dispos ; 39(3): 164-174, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29451686

ABSTRACT

We previously reported that KW-2449, (E)-1-{4-[2-(1H-Indazol-3-yl)vinyl]benzoyl}piperazine, a novel multikinase inhibitor developed for the treatment of leukemia patients, was oxidized to an iminium ion intermediate by monoamine oxidase B (MAO-B) and then converted to its oxo-piperazine form (M1) by aldehyde oxidase (AO). However, it was found that the significant decrease in the pharmacologically active metabolite M1 following repeated administration of KW-2449 in primates might hamper the effectiveness of the drug. The mechanism underlying this phenomenon was investigated and it was found that the AO activity was inhibited in a time-dependent manner in vitro under the co-incubation of KW-2449 and MAO-B, while neither KW-2449 nor M1 strongly inhibited MAO-B or AO activity. These results clearly suggest that MAO-B catalysed iminium ion metabolite inhibited AO, prompting us to investigate whether or not the iminium ion metabolite covalently binds to endogenous proteins, as has been reported with other reactive metabolites as a cause for idiosyncratic toxicity. The association of the radioactivity derived from 14 C-KW-2449 with endogenous proteins both in vivo and in vitro was confirmed and it was verified that this covalent binding was inhibited by the addition of sodium cyanide, an iminium ion-trapping reagent, and pargyline, a MAO-B inhibitor. These findings strongly suggest that the iminium ion metabolite of KW-2449 is highly reactive in inhibiting AO irreversibly and binding to endogenous macromolecules covalently.


Subject(s)
Aldehyde Oxidase/antagonists & inhibitors , Indazoles/metabolism , Indazoles/pharmacology , Piperazines/metabolism , Piperazines/pharmacology , Proteins/metabolism , Aldehyde Oxidase/metabolism , Animals , Carbon Isotopes , Humans , Macaca fascicularis , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Oxidation-Reduction , Pargyline/pharmacology , Protein Binding , Radioligand Assay , Sodium Cyanide/pharmacology
18.
Oncogene ; 37(4): 534-543, 2018 01 25.
Article in English | MEDLINE | ID: mdl-28991226

ABSTRACT

Histone H3 lysine-9 (H3K9) methylation is essential for retinoblastoma protein (RB)-mediated heterochromatin formation, epigenetic silencing of S-phase genes and permanent cell cycle arrest or cellular senescence. Besides as an H3K4 demethylase, lysine-specific demethylase-1 (LSD1) has been shown to promote H3K9 demethylation. However, it is unexplored whether LSD1 has a causal role in regulating cell cycle entry and senescence. Here we demonstrate that genetic depletion or pharmacological inhibition of LSD1 triggers G1 arrest and cellular senescence. Genome-wide chromatin immunoprecipitation-sequencing analysis reveals that LSD1 binding sites overlap significantly with those bound by the S-phase gene transcription factor E2F1. Gene ontology analysis demonstrates that a large portion of E2F1 and LSD1 cotargeted genes are involved in cell cycle and proliferation. Further analyses show that depletion of LSD1 increases the level of H3K9me2 and thereby represses expression of the LSD1-E2F1 cotarget genes, but has no effects on H3K4me2 level in those loci. In contrast, knockdown of the H3K4me2 reader PHF8 decreases the H3K4me2 level at the LSD1-E2F1 cotargeted loci, but this effect is rescued by codepletion of LSD1. Furthermore, the enzymatic activity of LSD1 is essential for H3K9me2 demethylation at cell cycle gene loci. Notably, cotreatment of chemotherapeutic agent camptothecin enhanced LSD1 inhibitor-induced senescence and growth inhibition of cancer cells in vitro and in mice. Our data reveal LSD1 as a molecular rheostat selectively regulating H3K9 demethylation at cell cycle gene loci, thereby representing a key player in oncogenesis and a viable target for cancer therapy.


Subject(s)
Carcinogenesis/genetics , Cellular Senescence/genetics , Chromatin/metabolism , E2F1 Transcription Factor/metabolism , Histone Demethylases/metabolism , Neoplasms/pathology , S Phase/genetics , Animals , Camptothecin/pharmacology , Cell Line, Tumor , Cellular Senescence/drug effects , Demethylation , E2F1 Transcription Factor/genetics , G1 Phase Cell Cycle Checkpoints/drug effects , G1 Phase Cell Cycle Checkpoints/genetics , Gene Knockdown Techniques , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/genetics , Histones/genetics , Histones/metabolism , Humans , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Pargyline/pharmacology , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
19.
Eur J Med Chem ; 143: 33-47, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29172081

ABSTRACT

A series of novel propargylamine-modified pyrimidinylthiourea derivatives (1-3) were designed and synthesized as multifunctional agents for Alzheimer's disease (AD) therapy, and their potential was evaluated through various biological experiments. Among these derivatives, compound 1b displayed good selective inhibitory activity against AChE (vs BuChE, IC50 = 0.324 µM, SI > 123) and MAO-B (vs MAO-A, IC50 = 1.427 µM, SI > 35). Molecular docking study showed that the pyrimidinylthiourea moiety of 1b could bind to the catalytic active site (CAS) of AChE, and the propargylamine moiety interacted directly with the flavin adenine dinucleotide (FAD) of MAO-B. Moreover, 1b demonstrated mild antioxidant ability, good copper chelating property, effective inhibitory activity against Cu2+-induced Aß1-42 aggregation, moderate neuroprotection, low cytotoxicity, and appropriate blood-brain barrier (BBB) permeability in vitro and was capable of ameliorating scopolamine-induced cognitive impairment in mice. These results indicated that 1b has the potential to be a multifunctional candidate for the treatment of Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Drug Discovery , Imidazoles/pharmacology , Pargyline/analogs & derivatives , Propylamines/pharmacology , Pyrimidines/pharmacology , Thiourea/pharmacology , Acetylcholinesterase/metabolism , Animals , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Dose-Response Relationship, Drug , Humans , Imidazoles/chemistry , Mice , Molecular Structure , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/pharmacology , Pargyline/chemistry , Pargyline/pharmacology , Propylamines/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Rats , Scopolamine , Structure-Activity Relationship , Thiourea/chemical synthesis , Thiourea/chemistry
20.
ChemMedChem ; 12(24): 2044-2053, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29120081

ABSTRACT

As histone deacetylases (HDACs) play an important role in the treatment of cancer, their selective inhibition has been the subject of various studies. These continuous investigations have given rise to a large collection of pan- and selective HDAC inhibitors, containing diverse US Food and Drug Administration (FDA)-approved representatives. In previous studies, a class of alkyne-based HDAC inhibitors was presented. We modified this scaffold in two previously neglected regions and compared their cytotoxicity and affinity toward HDAC1, HDAC6, and HDAC8. We were able to show that R-configured propargylamines contribute to increased selectivity for HDAC6. Docking studies on available HDAC crystal structures were carried out to rationalize the observed selectivity of the compounds. Substitution of the aromatic portion by a thiophene derivative results in high affinity and low cytotoxicity, indicating an improved drug tolerance.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/pharmacology , Pargyline/analogs & derivatives , Propylamines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Humans , Molecular Structure , Pargyline/chemical synthesis , Pargyline/chemistry , Pargyline/pharmacology , Propylamines/chemical synthesis , Propylamines/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL