1
|
Rajić M, Stare J. Investigation of Electrostatic Effects on Enyzme Catalysis: Insights from Computational Simulations of Monoamine Oxidase A Pathological Variants Leading to the Brunner Syndrome. J Chem Inf Model 2025; 65:3439-3450. [PMID: 40135540 PMCID: PMC12004519 DOI: 10.1021/acs.jcim.4c01698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Brunner syndrome is a rare genetic disorder characterized by impulsive aggressiveness and intellectual disability, which is linked to impaired function of the monoamine oxidase A (MAO-A) enzyme. Patients with specific point mutations in the MAOA gene have been reported to exhibit these symptoms, along with notably elevated serotonin levels, which suggest a decreased catalytic performance of the mutated MAO-A enzymes. In this study, we present multiscale molecular simulations focusing on the rate-limiting step of MAO-A-catalyzed serotonin degradation for the C266F and V244I variants that are reportedly associated with pathologies characteristic of the Brunner syndrome. We found that the C266F mutation causes an approximately 18,000-fold slowdown of enzymatic function, which is equivalent to a MAOA gene knockout. For the V244I mutant, a somewhat smaller, yet still significant 300-fold slowdown has been estimated. Furthermore, we conducted a comprehensive comparison of the impact of enzyme electrostatics on the catalytic function of the wild-type (WT) MAO-A and both aforementioned mutants (C266F and V244I), as well as on the E446K mutant investigated in one of our earlier studies. The results have shown that the mutation induces a noteworthy change in electrostatic interactions between the reacting moiety and its enzymatic surroundings, leading to a decreased catalytic performance in all of the considered MAO-A variants. An analysis of mutation effects supported by geometry comparison of mutants and the wild-type enzyme at a residue level suggests that a principal driving force behind the altered catalytic performance of the mutants is subtle structural changes scattered along the entire enzyme. These shifts in geometry also affect domains most relevant to catalysis, where structural offsets of few tenths of an Å can significantly change contribution to the barrier of the involved residues. These results are in full agreement with the reasoning derived from clinical observations and biochemical data. Our research represents a step forward in the attempts of using fundamental principles of chemical physics in order to explain genetically driven pathologies. In addition, our results support the view that the catalytic function of enzymes is crucially driven by electrostatic interactions.
Collapse
Affiliation(s)
- Martina Rajić
- Theory Department, Laboratory
for Computational Biochemistry and Drug Design, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| | - Jernej Stare
- Theory Department, Laboratory
for Computational Biochemistry and Drug Design, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
2
|
Rajić M, Prah A, Stare J. Deciphering the Two-Step Hydride Mechanism of Monoamine Oxidase Flavoenzymes. ACS OMEGA 2024; 9:43046-43057. [PMID: 39464429 PMCID: PMC11500147 DOI: 10.1021/acsomega.4c06575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024]
Abstract
The complete two-step hydride transfer mechanism of amine oxidation involved in the metabolism of monoamine neurotransmitters was scrutinized by DFT calculations. In living organisms, this process is catalyzed by monoamine oxidase enzymes. Herein, we focus on some intriguing aspects of the reaction that may have been previously noticed but have not been clarified to date. The first step of the reaction includes the C-H bond cleavage on the methylene group vicinal to the amino group of the monoamine substrate and the subsequent transfer of hydrogen to the N5 atom of the flavin prosthetic group of the enzyme. We confirmed the nature of this step to be hydride transfer by evaluation of the pertinent HOMO-LUMO gap together with analysis of orbital contours alongside the intrinsic reaction coordinate profile. Next, we investigated the rather peculiar intermediate adduct that may form between the amine substrate and the flavin molecule, featuring an unusually long C-N bond of ∼1.62 Å. Although this bond is quite stable in the gas phase, the presence of just a few explicit water molecules facilitates its dissociation almost without energy input so that the amine-flavin intermediate can form an ionic pair instead. We attribute the existence of the unusual C-N bond to a fragile balance between opposing electronic structure effects, as evaluated by the natural bond orbital analysis. In line with this, the intermediate in the solution or in the enzyme active site can exist in two energetically almost equivalent forms, namely, as a covalently bound complex or as an ion pair, as suggested by previous studies. Finally, we characterized the transformation of the intermediate to the fully reduced flavin and imine products via proton transfer from the amino group to the flavin N1 atom, completing the reductive part of the catalytic cycle. Although we found that explicit solvation substantially boosts the kinetics of this step, the corresponding barrier is significantly lower than that in the hydride transfer step, confirming hydrogen abstraction as the rate-limiting step of amine oxidation and validating the two-step hydride transfer mechanism of monoamine oxidases.
Collapse
Affiliation(s)
- Martina Rajić
- Theory Department, Laboratory for Computational
Biochemistry and Drug Design, National Institute
of Chemistry, Hajdrihova
19, Ljubljana SI-1000, Slovenia
| | - Alja Prah
- Theory Department, Laboratory for Computational
Biochemistry and Drug Design, National Institute
of Chemistry, Hajdrihova
19, Ljubljana SI-1000, Slovenia
| | - Jernej Stare
- Theory Department, Laboratory for Computational
Biochemistry and Drug Design, National Institute
of Chemistry, Hajdrihova
19, Ljubljana SI-1000, Slovenia
| |
Collapse
|
3
|
Vrban L, Vianello R. Prominent Neuroprotective Potential of Indole-2- N-methylpropargylamine: High Affinity and Irreversible Inhibition Efficiency towards Monoamine Oxidase B Revealed by Computational Scaffold Analysis. Pharmaceuticals (Basel) 2024; 17:1292. [PMID: 39458932 PMCID: PMC11510145 DOI: 10.3390/ph17101292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Monoamine oxidases (MAO) are flavoenzymes that metabolize a range of brain neurotransmitters, whose dysregulation is closely associated with the development of various neurological disorders. This is why MAOs have been the central target in pharmacological interventions for neurodegeneration for more than 60 years. Still, existing drugs only address symptoms and not the cause of the disease, which underlines the need to develop more efficient inhibitors without adverse effects. Methods: Our drug design strategy relied on docking 25 organic scaffolds to MAO-B, which were extracted from the ChEMBL20 database with the highest cumulative counts of unique member compounds and bioactivity assays. The most promising candidates were substituted with the inactivating propargylamine group, while further affinity adjustment was made by its N-methylation. A total of 46 propargylamines were submitted to the docking and molecular dynamics simulations, while the best binders underwent mechanistic DFT analysis that confirmed the hydride abstraction mechanism of the covalent inhibition reaction. Results: We identified indole-2-propargylamine 4fH and indole-2-N-methylpropargylamine 4fMe as superior MAO-B binders over the clinical drugs rasagiline and selegiline. DFT calculations highlighted 4fMe as more potent over selegiline, evident in a reduced kinetic requirement (ΔΔG‡ = -2.5 kcal mol-1) and an improved reaction exergonicity (ΔΔGR = -4.3 kcal mol-1), together with its higher binding affinity, consistently determined by docking (ΔΔGBIND = -0.1 kcal mol-1) and MM-PBSA analysis (ΔΔGBIND = -1.5 kcal mol-1). Conclusions: Our findings strongly advocate 4fMe as an excellent drug candidate, whose synthesis and biological evaluation are highly recommended. Also, our results reveal the structural determinants that influenced the affinity and inhibition rates that should cooperate when designing further MAO inhibitors, which are of utmost significance and urgency with the increasing prevalence of brain diseases.
Collapse
Affiliation(s)
| | - Robert Vianello
- Laboratory for the Computational Design and Synthesis of Functional Materials, Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| |
Collapse
|
4
|
Stare J. Oxidation of Flavin by Molecular Oxygen: Computational Insights into a Possible Radical Mechanism. ACS OMEGA 2024; 9:23431-23441. [PMID: 38854520 PMCID: PMC11154890 DOI: 10.1021/acsomega.4c00307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/19/2024] [Accepted: 05/13/2024] [Indexed: 06/11/2024]
Abstract
As a highly electrophilic moiety capable of oxidizing a variety of small organic molecules and biomolecules, flavin is an important prosthetic group in many enzymes. Upon oxidation of the substrate, flavin is converted into its reduced (dihydrogenated) form. The catalytic cycle is completed through oxidation back to the oxidized form, thus restoring the enzyme's oxidizing capability. While it has been firmly established that oxidation of the reduced form of flavin is cast by molecular oxygen, yielding oxidized flavin and hydrogen peroxide, the mechanism of this process is still poorly understood. Herein, we investigate the radical mechanism, which is one of the possible reaction mechanisms, by quantum chemical calculations. Because molecular oxygen exists as a triplet in its electronic ground state, whereas the products are singlets, the reaction is accompanied by hopping between electronic surfaces. We find that the rate-limiting factor of flavin oxidation is likely associated with the change in the spin state of the system. By considering several possible reactions involving flavin and its derivatives in the radical form and by examining the corresponding parts of the potential energy surface in various spin states, we estimate the effective barrier of the kinetically and thermodynamically preferred variant of flavin oxidation to be about 15 kcal/mol in the gas phase and about 7 kcal/mol in a polar (aqueous) environment. This is in agreement with kinetic studies of the corresponding monoamine oxidase enzymes, confirming the radical mechanism as a viable option for flavin regeneration in enzymes.
Collapse
Affiliation(s)
- Jernej Stare
- National Institute of Chemistry,Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
5
|
Zaib S, Khan I, Ali HS, Younas MT, Ibrar A, Al-Odayni AB, Al-Kahtani AA. Design and discovery of anthranilamide derivatives as a potential treatment for neurodegenerative disorders via targeting cholinesterases and monoamine oxidases. Int J Biol Macromol 2024; 272:132748. [PMID: 38821306 DOI: 10.1016/j.ijbiomac.2024.132748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Neurodegenerative diseases with progressive cellular loss of the central nervous system and elusive disease etiology provide a continuous impetus to explore drug discovery programmes aiming at identifying robust and effective inhibitors of cholinesterase and monoamine oxidase enzymes. We herein present a concise library of anthranilamide derivatives involving a palladium-catalyzed Suzuki-Miyaura cross-coupling reaction to install the diverse structural diversity required for the desired biological action. Using Ellman's method, cholinesterase inhibitory activity was performed against AChE and BuChE enzymes. In vitro assay results demonstrated that anthranilamides are potent inhibitors with remarkable potency. Compound 6k emerged as the lead candidate and dual inhibitor of both enzymes with IC50 values of 0.12 ± 0.01 and 0.49 ± 0.02 μM against AChE and BuChE, respectively. Several other compounds were found as highly potent and selective inhibitors. Anthranilamide derivatives were also tested against monoamine oxidase (A and B) enzymes using fluorometric method. In vitro data revealed compound 6h as the most potent inhibitor against MAO-A, showing an IC50 value of 0.44 ± 0.02 μM, whereas, compound 6k emerged as the top inhibitor of MAO-B with an IC50 value of 0.06 ± 0.01 μM. All the lead inhibitors were analyzed for the identification of their mechanism of action using Michaelis-Menten kinetics experiments. Compound 6k and 6h depicted a competitive mode of action against AChE and MAO-A, whereas, a non-competitive and mixed-type of inhibition was observed against BuChE and MAO-B by compounds 6k. Molecular docking analysis revealed remarkable binding affinities of the potent inhibitors with specific residues inside the active site of receptors. Furthermore, molecular dynamics simulations were performed to explore the ability of potent compounds to form energetically stable complexes with the target protein. Finally, in silico ADME calculations also demonstrated that the potent compounds exhibit promising pharmacokinetic profile, satisfying the essential criteria for drug-likeness. Altogether, the findings reported in the current work clearly suggest that the identified anthranilamide derivatives have the potential to serve as effective drug candidates for future investigations.
Collapse
Affiliation(s)
- Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore 54590, Pakistan.
| | - Imtiaz Khan
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester MI 7DN, UK.
| | - Hafiz Saqib Ali
- Chemistry Research Laboratory, Department of Chemistry, the INEOS Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK
| | - Muhammad Tayyab Younas
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore 54590, Pakistan
| | - Aliya Ibrar
- Department of Chemistry, Faculty of Physical and Applied Sciences, The University of Haripur, Haripur, KPK 22620, Pakistan.
| | - Abdel-Basit Al-Odayni
- Department of Restorative Dental Sciences, College of Dentistry, King Saud University, P.O. Box 60169, Riyadh 11545, Saudi Arabia
| | - Abdullah A Al-Kahtani
- Chemistry Department, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
6
|
Price NJ, Nakamura A, Castagnoli N, Tanko JM. Why Does Monoamine Oxidase (MAO) Catalyze the Oxidation of Some Tetrahydropyridines? Chembiochem 2024; 25:e202400126. [PMID: 38602445 DOI: 10.1002/cbic.202400126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/18/2024] [Indexed: 04/12/2024]
Abstract
Results pertaining to the mechanism of the oxidation of the tertiary amine 1-methyl-4-(1-methyl-1-H-pyrrol-2-yl)-1,2,3,6-tetrahydropyridine (MMTP, a close analog of the Parkinsonism inducing compound MPTP) by 3-methyllumiflavin (3MLF), a chemical model for the FAD cofactor of monoamine oxidase, are reported. MMTP and related compounds are among the few tertiary amines that are monoamine oxidase B (MAO-B) substrates. The MMTP/3MLF reaction is catalytic in the presence of O2 and the results under anaerobic conditions strongly suggest the involvement of radical intermediates, consistent with a single electron transfer mechanism. These observations support a new hypothesis to explain the MAO-catalyzed oxidations of amines. In general, electron transfer is thermodynamically unfavorable, and as a result, most 1° and 2° amines react via one of the currently accepted polar pathways. Steric constraints prevent 3° amines from reacting via a polar pathway. Those select 3° amines that are MAO substrates possess certain structural features (e. g., a C-H bond that is α- both to nitrogen and a C=C) that dramatically lower the pKa of the corresponding radical cation. Consequently, the thermodynamically unfavorable electron transfer equilibrium is driven towards products by an extremely favorable deprotonation step in the context of Le Chatelier's principle.
Collapse
Affiliation(s)
- Nathan J Price
- Department of Chemistry, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Akiko Nakamura
- Department of Chemistry, Colorado State University, Fort Collins, CO, 80523, USA
| | - Neal Castagnoli
- Department of Chemistry, Virginia Tech, Blacksburg, VA, 24061, USA
| | - James M Tanko
- Department of Chemistry, Virginia Tech, Blacksburg, VA, 24061, USA
| |
Collapse
|
7
|
Bongaerts D, Bouchez A, De Roos J, Cnockaert M, Wieme AD, Vandamme P, Weckx S, De Vuyst L. Refermentation and maturation of lambic beer in bottles: a necessary step for gueuze production. Appl Environ Microbiol 2024; 90:e0186923. [PMID: 38446583 PMCID: PMC11022581 DOI: 10.1128/aem.01869-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/04/2024] [Indexed: 03/08/2024] Open
Abstract
The production of gueuze beers through refermentation and maturation of blends of lambic beer in bottles is a way for lambic brewers to cope with the variability among different lambic beer batches. The resulting gueuze beers are more carbonated than lambic beers and are supposed to possess a unique flavor profile that varies over time. To map this refermentation and maturation process for gueuze production, a blend of lambic beers was made and bottled, whereby one of them was produced with the old wheat landrace Zeeuwse Witte. Through the use of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and high-throughput sequencing of bacterial and fungal amplicons, in combination with metabolite target analysis, new insights into gueuze production were obtained. During the initial stages of refermentation, the conditions in the bottles were similar to those encountered during the maturation phase of lambic beer productions in wooden barrels, which was also reflected microbiologically (presence of Brettanomyces species, Pediococcus damnosus, and Acetobacter lambici) and biochemically (ethanol, higher alcohols, lactic acid, acetic acid, volatile phenolic compounds, and ethyl esters). However, after a few weeks of maturation, a switch from a favorable environment to one with nutrient and dissolved oxygen depletion resulted in several changes. Concerning the microbiology, a sequential prevalence of three lactic acid bacterial species occurred, namely, P. damnosus, Lentilactobacillus buchneri, and Lactobacillus acetotolerans, while the diversity of the yeasts decreased. Concerning the metabolites produced, mainly those of the Brettanomyces yeasts determined the metabolic profiles encountered during later stages of the gueuze production.IMPORTANCEGueuze beers are the result of a refermentation and maturation process of a blend of lambic beers carried out in bottles. These gueuze beers are known to have a long shelf life, and their quality typically varies over time. However, knowledge about gueuze production in bottles is scarce. The present study provided more insights into the varying microbial and metabolite composition of gueuze beers during the first 2 years of this refermentation and maturation process. This will allow gueuze producers to gain more information about the influence of the refermentation and maturation time on their beers. These insights can also be used by gueuze producers to better inform their customers about the quality of young and old gueuze beers.
Collapse
Affiliation(s)
- Dries Bongaerts
- Department of Bioengineering Sciences, Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Arne Bouchez
- Department of Bioengineering Sciences, Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jonas De Roos
- Department of Bioengineering Sciences, Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Margo Cnockaert
- Department of Biochemistry and Microbiology, Laboratory for Microbiology, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Anneleen D. Wieme
- Department of Biochemistry and Microbiology, Laboratory for Microbiology, Faculty of Sciences, Ghent University, Ghent, Belgium
- Department of Biochemistry and Microbiology, BCCM/LMG Bacteria Collection, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Peter Vandamme
- Department of Biochemistry and Microbiology, Laboratory for Microbiology, Faculty of Sciences, Ghent University, Ghent, Belgium
- Department of Biochemistry and Microbiology, BCCM/LMG Bacteria Collection, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Stefan Weckx
- Department of Bioengineering Sciences, Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Luc De Vuyst
- Department of Bioengineering Sciences, Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
8
|
Świder O, Roszko MŁ, Wójcicki M. The inhibitory effects of plant additives on biogenic amine formation in fermented foods - a review. Crit Rev Food Sci Nutr 2023; 64:12935-12960. [PMID: 37724793 DOI: 10.1080/10408398.2023.2258964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
Fermented food has unique properties and high nutritional value, and thus, should constitute a basic element of a balanced and health-promoting diet. However, it can accumulate considerable amount of biogenic amines (BAs), which ingested in excess can lead to adverse health effects. The application of plant-derived additives represents a promising strategy to ensure safety or enhance the functional and organoleptic properties of fermented food. This review summarizes currently available data on the application of plant-origin additives with the aim to reduce BA content in fermented products. The importance of ensuring fermented food safety has been highlighted considering the growing evidence of beneficial effects resulting from the consumption of this type of food, as well as the increasing number of individuals sensitive to BAs. The examined plant-origin additives reduced the BA concentration to varying degrees, and their efficacy depended on the type of additive, matrix, autochthonous, and inoculated microorganisms, as well as the manufacturing conditions. The main mechanisms of action include antimicrobial effects and the inhibition of microbial decarboxylases. Further research on the optimization of bioactive substances extraction, standardization of their chemical composition, and development of detailed procedures for its use in fermented products manufacturing are needed.
Collapse
Affiliation(s)
- Olga Świder
- Department of Food Safety and Chemical Analysis, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology - State Research Institute, Warsaw, Poland
| | - Marek Łukasz Roszko
- Department of Food Safety and Chemical Analysis, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology - State Research Institute, Warsaw, Poland
| | - Michał Wójcicki
- Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology - State Research Institute, Warsaw, Poland
| |
Collapse
|
9
|
Schober L, Dobiašová H, Jurkaš V, Parmeggiani F, Rudroff F, Winkler M. Enzymatic reactions towards aldehydes: An overview. FLAVOUR FRAG J 2023; 38:221-242. [PMID: 38505272 PMCID: PMC10947199 DOI: 10.1002/ffj.3739] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/01/2023] [Accepted: 03/06/2023] [Indexed: 03/21/2024]
Abstract
Many aldehydes are volatile compounds with distinct and characteristic olfactory properties. The aldehydic functional group is reactive and, as such, an invaluable chemical multi-tool to make all sorts of products. Owing to the reactivity, the selective synthesis of aldehydic is a challenging task. Nature has evolved a number of enzymatic reactions to produce aldehydes, and this review provides an overview of aldehyde-forming reactions in biological systems and beyond. Whereas some of these biotransformations are still in their infancy in terms of synthetic applicability, others are developed to an extent that allows their implementation as industrial biocatalysts.
Collapse
Affiliation(s)
- Lukas Schober
- Institute of Molecular BiotechnologyGraz University of TechnologyGrazAustria
| | - Hana Dobiašová
- Institute of Chemical and Environmental EngineeringSlovak University of TechnologyBratislavaSlovakia
| | - Valentina Jurkaš
- Institute of Molecular BiotechnologyGraz University of TechnologyGrazAustria
| | - Fabio Parmeggiani
- Dipartimento di Chimica, Materiali ed Ingegneria Chimica “Giulio Natta”Politecnico di MilanoMilanItaly
| | - Florian Rudroff
- Institute of Applied Synthetic ChemistryTU WienViennaAustria
| | - Margit Winkler
- Institute of Molecular BiotechnologyGraz University of TechnologyGrazAustria
- Area BiotransformationsAustrian Center of Industrial BiotechnologyGrazAustria
| |
Collapse
|
10
|
Hok L, Vianello R. Selective Deuteration Improves the Affinity of Adenosine A 2A Receptor Ligands: A Computational Case Study with Istradefylline and Caffeine. J Chem Inf Model 2023; 63:3138-3149. [PMID: 37155356 DOI: 10.1021/acs.jcim.3c00424] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
We used a range of computational techniques to assess the effect of selective C-H deuteration on the antagonist istradefylline affinity for the adenosine A2A receptor, which was discussed relative to its structural analogue caffeine, a well-known and likely the most widely used stimulant. The obtained results revealed that smaller caffeine shows high receptor flexibility and exchanges between two distinct poses, which agrees with crystallographic data. In contrast, the additional C8-trans-styryl fragment in istradefylline locks the ligand within a uniform binding pose, while contributing to the affinity through the C-H···π and π···π contacts with surface residues, which, together with its much lower hydration prior to binding, enhances the affinity over caffeine. In addition, the aromatic C8-unit shows a higher deuteration sensitivity over the xanthine part, so when both of its methoxy groups are d6-deuterated, the affinity improvement is -0.4 kcal mol-1, which surpasses the overall affinity gain of -0.3 kcal mol-1 in the perdeuterated d9-caffeine. Yet, the latter predicts around 1.7-fold potency increase, being relevant for its pharmaceutical implementations, and also those within the coffee and energy drink production industries. Still, the full potential of our strategy is achieved in polydeuterated d19-istradefylline, whose A2A affinity improves by -0.6 kcal mol-1, signifying a 2.8-fold potency increase that strongly promotes it as a potential synthetic target. This knowledge supports deuterium application in drug design, and while the literature already reports about over 20 deuterated drugs currently in the clinical development, it is easily foreseen that more examples will hit the market in the years to come. With this in mind, we propose that the devised computational methodology, involving the ONIOM division of the QM region for the ligand and the MM region for its environment, with an implicit quantization of nuclear motions relevant for the H/D exchange, allows fast and efficient estimates of the binding isotope effects in any biological system.
Collapse
Affiliation(s)
- Lucija Hok
- Laboratory for the Computational Design and Synthesis of Functional Materials, Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Robert Vianello
- Laboratory for the Computational Design and Synthesis of Functional Materials, Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
11
|
Sharma H, Raju B, Narendra G, Motiwale M, Sharma B, Verma H, Silakari O. QM/MM Studies on Enzyme Catalysis and Insight into Designing of New Inhibitors by ONIOM Approach: Recent Update. ChemistrySelect 2023. [DOI: 10.1002/slct.202203319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Himani Sharma
- Molecular Modeling Lab (MML) Department of Pharmaceutical Sciences and Drug Research Punjabi University Patiala Punjab 147002 India
| | - Baddipadige Raju
- Molecular Modeling Lab (MML) Department of Pharmaceutical Sciences and Drug Research Punjabi University Patiala Punjab 147002 India
| | - Gera Narendra
- Molecular Modeling Lab (MML) Department of Pharmaceutical Sciences and Drug Research Punjabi University Patiala Punjab 147002 India
| | - Mohit Motiwale
- Molecular Modeling Lab (MML) Department of Pharmaceutical Sciences and Drug Research Punjabi University Patiala Punjab 147002 India
| | - Bhavna Sharma
- Molecular Modeling Lab (MML) Department of Pharmaceutical Sciences and Drug Research Punjabi University Patiala Punjab 147002 India
| | - Himanshu Verma
- Molecular Modeling Lab (MML) Department of Pharmaceutical Sciences and Drug Research Punjabi University Patiala Punjab 147002 India
| | - Om Silakari
- Molecular Modeling Lab (MML) Department of Pharmaceutical Sciences and Drug Research Punjabi University Patiala Punjab 147002 India
| |
Collapse
|
12
|
El-Damasy AK, Park JE, Kim HJ, Lee J, Bang EK, Kim H, Keum G. Identification of New N-methyl-piperazine Chalcones as Dual MAO-B/AChE Inhibitors. Pharmaceuticals (Basel) 2023; 16:ph16010083. [PMID: 36678580 PMCID: PMC9860728 DOI: 10.3390/ph16010083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Monoamine oxidase-B (MAO-B), acetylcholinesterase (AChE), and butyrylcholinesterase (BChE) have been considered target enzymes of depression and neurodegenerative diseases, including Alzheimer's disease (AD). In this study, seventeen N-methyl-piperazine chalcones were synthesized, and their inhibitory activities were evaluated against the target enzymes. Compound 2k (3-trifluoromethyl-4-fluorinated derivative) showed the highest selective inhibition against MAO-B with an IC50 of 0.71 μM and selectivity index (SI) of 56.34, followed by 2n (2-fluoro-5-bromophenyl derivative) (IC50 = 1.11 μM, SI = 16.04). Compounds 2k and 2n were reversible competitive MAO-B inhibitors with Ki values of 0.21 and 0.28 μM, respectively. Moreover, 2k and 2n effectively inhibited AChE with IC50 of 8.10 and 4.32 μM, which underscored their multi-target inhibitory modes. Interestingly, compound 2o elicited remarkable inhibitions over MAO-B, AChE, and BChE with IC50 of 1.19-3.87 μM. A cell-based assay of compounds 2k and 2n against Vero normal cells pointed out their low cytotoxicity. In a docking simulation, 2k showed the lowest energy for MAO-B (-11.6 kcal/mol) with four hydrogen bonds and two π-π interactions. Furthermore, in silico studies were conducted, and disclosed that 2k and 2n are expected to possess favorable pharmacokinetic properties, such as the ability to penetrate the blood-brain barrier (BBB). In view of these findings, compounds 2k and 2n could serve as promising potential candidates for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ashraf K. El-Damasy
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
- Correspondence: (A.K.E.-D.); (H.K.); (G.K.)
| | - Jong Eun Park
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Hyun Ji Kim
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Jinhyuk Lee
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
- Department of Bioinformatics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Eun-Kyoung Bang
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hoon Kim
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
- Correspondence: (A.K.E.-D.); (H.K.); (G.K.)
| | - Gyochang Keum
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Correspondence: (A.K.E.-D.); (H.K.); (G.K.)
| |
Collapse
|
13
|
Brunner syndrome caused by point mutation explained by multiscale simulation of enzyme reaction. Sci Rep 2022; 12:21889. [PMID: 36536002 PMCID: PMC9763434 DOI: 10.1038/s41598-022-26296-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Brunner syndrome is a disorder characterized by intellectual disability and impulsive, aggressive behavior associated with deficient function of the monoamine oxidase A (MAO-A) enzyme. These symptoms (along with particularly high serotonin levels) have been reported in patients with two missense variants in MAO-A (p.R45W and p.E446K). Herein, we report molecular simulations of the rate-limiting step of MAO-A-catalyzed serotonin degradation for these variants. We found that the R45W mutation causes a 6000-fold slowdown of enzymatic function, whereas the E446K mutation causes a 450-fold reduction of serotonin degradation rate, both of which are practically equivalent to a gene knockout. In addition, we thoroughly compared the influence of enzyme electrostatics on the catalytic function of both the wild type MAO-A and the p.R45W variant relative to the wild type enzyme, revealing that the mutation represents a significant electrostatic perturbation that contributes to the barrier increase. Understanding genetic disorders is closely linked to understanding the associated chemical mechanisms, and our research represents a novel attempt to bridge the gap between clinical genetics and the underlying chemical physics.
Collapse
|
14
|
Prah A, Gavranić T, Perdih A, Sollner Dolenc M, Mavri J. Computational Insights into β-Carboline Inhibition of Monoamine Oxidase A. Molecules 2022; 27:molecules27196711. [PMID: 36235246 PMCID: PMC9571839 DOI: 10.3390/molecules27196711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/03/2022] [Accepted: 10/06/2022] [Indexed: 11/07/2022] Open
Abstract
Monoamine oxidases (MAOs) are an important group of enzymes involved in the degradation of neurotransmitters and their imbalanced mode of action may lead to the development of various neuropsychiatric or neurodegenerative disorders. In this work, we report the results of an in-depth computational study in which we performed a static and a dynamic analysis of a series of substituted β-carboline natural products, found mainly in roasted coffee and tobacco smoke, that bind to the active site of the MAO-A isoform. By applying molecular docking in conjunction with structure-based pharmacophores and molecular dynamics simulations coupled with dynamic pharmacophores, we extensively investigated the geometric aspects of MAO-A binding. To gain insight into the energetics of binding, we used the linear interaction energy (LIE) method and determined the key anchors that allow productive β-carboline binding to MAO-A. The results presented herein could be applied in the rational structure-based design and optimization of β-carbolines towards preclinical candidates that would target the MAO-A enzyme and would be applicable especially in the treatment of mental disorders such as depression.
Collapse
Affiliation(s)
- Alja Prah
- National Institute of Chemistry, SI-1000 Ljubljana, Slovenia
- Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia
| | - Tanja Gavranić
- National Institute of Chemistry, SI-1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Andrej Perdih
- National Institute of Chemistry, SI-1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | | | - Janez Mavri
- National Institute of Chemistry, SI-1000 Ljubljana, Slovenia
- Correspondence:
| |
Collapse
|
15
|
Tsafack PB, Tsopmo A. Effects of bioactive molecules on the concentration of biogenic amines in foods and biological systems. Heliyon 2022; 8:e10456. [PMID: 36105466 PMCID: PMC9465362 DOI: 10.1016/j.heliyon.2022.e10456] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/14/2022] [Accepted: 08/22/2022] [Indexed: 11/20/2022] Open
Abstract
Biogenic amines (BAs) are a group of molecules naturally present in foods that contain amino acids, peptides, and proteins as well as in biological systems. In foods, their concentrations typically increase during processing and storage because of exposure to microorganisms that catalyze their formation by releasing amino acid decarboxylases. The concentrations of BAs above certain values are indicative of unsafe foods due to associate neuronal toxicity, allergenic reactions, and increase risks of cardiovascular diseases. There are therefore various strategies that focus on the control of BAs in foods mostly through elimination, inactivation, or inhibition of the growth of microorganisms. Increasingly, there are works on bioactive compounds that can decrease the concentration of BAs through their antimicrobial activity as well as the inhibition of decarboxylating enzymes that control their formation in foods or amine oxidases and N-acetyltransferase that control the degradation in vivo. This review focusses on the role of food-derived bioactive compounds and the mechanism by which they regulate the concentration of BAs. The findings are that most active molecules belong to polyphenols, one of the largest groups of plant secondary metabolites, additionally other useful +compounds are present in extracts of different herbs and spices. Different mechanisms have been proposed for the effects of polyphenols depending on the model system. Studies on the effects in vivo are limited and there is a lack of bioavailability and transport data which are important to assess the importance of the bioactive molecules.
Collapse
Affiliation(s)
- Patrick Blondin Tsafack
- Nutrition and Functional Food, School of Biosciences and Veterinary Medicine, University of Camerino, Via A. D'Accorso, 16, Camerino, Italy
| | - Apollinaire Tsopmo
- Food Science and Nutrition Program, Department of Chemistry and Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, K1S 5B6, Ottawa, ON, Canada
| |
Collapse
|
16
|
Scoditti S, Dabbish E, Pieslinger GE, Rezabal E, Lopez X, Sicilia E, Salassa L. Flavin-mediated photoactivation of Pt(IV) anticancer complexes: computational insights on the catalytic mechanism. Phys Chem Chem Phys 2022; 24:5323-5329. [PMID: 35188500 DOI: 10.1039/d1cp05507k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The mechanism for the photocatalytic activation of Pt(IV) anticancer prodrugs by riboflavin in the presence of NADH has been investigated by DFT. In the first step of the reaction, the oxidation kinetics of NADH to afford the catalytically active riboflavin hydroquinone is dramatically favoured by generation of the flavin triplet excited state. In the triplet, formation of a π-π stacked adduct promotes the hydride transfer from NADH to riboflavin with an almost barrierless pathway (2.7 kcal mol-1). In the singlet channel, conversely, the process is endergonic and requires overcoming a higher activation energy (19.2 kcal mol-1). In the second half of the reaction, the reduction of the studied Pt(IV) complexes by riboflavin hydroquinone occurs via an inner sphere mechanism, displaying free energy barriers smaller than 10 kcal mol-1. Pt reduction by bioreductants such as NADH and ascorbate involve instead less stabilized transition states (22.2-38.3 kcal mol-1), suggesting that riboflavin hydroquinone is an efficient reducing agent for Pt(IV) derivatives in biological settings.
Collapse
Affiliation(s)
- Stefano Scoditti
- Department of Chemistry and Chemical Technologies, Università della Calabria, Arcavacata di Rende (CS), 87036, Italy.
| | - Eslam Dabbish
- Department of Chemistry and Chemical Technologies, Università della Calabria, Arcavacata di Rende (CS), 87036, Italy.
| | - German E Pieslinger
- CONICET - Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Buenos Aires, Argentina
| | - Elixabete Rezabal
- Donostia International Physics Center, Paseo Manuel de Lardizabal 4, Donostia, 20018, Spain. .,Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, Euskal Herriko Unibertsitatea UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia, 20018, Spain
| | - Xabier Lopez
- Donostia International Physics Center, Paseo Manuel de Lardizabal 4, Donostia, 20018, Spain. .,Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, Euskal Herriko Unibertsitatea UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia, 20018, Spain
| | - Emilia Sicilia
- Department of Chemistry and Chemical Technologies, Università della Calabria, Arcavacata di Rende (CS), 87036, Italy.
| | - Luca Salassa
- Donostia International Physics Center, Paseo Manuel de Lardizabal 4, Donostia, 20018, Spain. .,Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, Euskal Herriko Unibertsitatea UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia, 20018, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, 48011, Spain
| |
Collapse
|
17
|
Why Monoamine Oxidase B Preferably Metabolizes N-Methylhistamine over Histamine: Evidence from the Multiscale Simulation of the Rate-Limiting Step. Int J Mol Sci 2022; 23:ijms23031910. [PMID: 35163835 PMCID: PMC8836602 DOI: 10.3390/ijms23031910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 11/18/2022] Open
Abstract
Histamine levels in the human brain are controlled by rather peculiar metabolic pathways. In the first step, histamine is enzymatically methylated at its imidazole Nτ atom, and the produced N-methylhistamine undergoes an oxidative deamination catalyzed by monoamine oxidase B (MAO-B), as is common with other monoaminergic neurotransmitters and neuromodulators of the central nervous system. The fact that histamine requires such a conversion prior to oxidative deamination is intriguing since MAO-B is known to be relatively promiscuous towards monoaminergic substrates; its in-vitro oxidation of N-methylhistamine is about 10 times faster than that for histamine, yet this rather subtle difference appears to be governing the decomposition pathway. This work clarifies the MAO-B selectivity toward histamine and N-methylhistamine by multiscale simulations of the rate-limiting hydride abstraction step for both compounds in the gas phase, in aqueous solution, and in the enzyme, using the established empirical valence bond methodology, assisted by gas-phase density functional theory (DFT) calculations. The computed barriers are in very good agreement with experimental kinetic data, especially for relative trends among systems, thereby reproducing the observed MAO-B selectivity. Simulations clearly demonstrate that solvation effects govern the reactivity, both in aqueous solution as well as in the enzyme although with an opposing effect on the free energy barrier. In the aqueous solution, the transition-state structure involving histamine is better solvated than its methylated analog, leading to a lower barrier for histamine oxidation. In the enzyme, the higher hydrophobicity of N-methylhistamine results in a decreased number of water molecules at the active side, leading to decreased dielectric shielding of the preorganized catalytic electrostatic environment provided by the enzyme. This renders the catalytic environment more efficient for N-methylhistamine, giving rise to a lower barrier relative to histamine. In addition, the transition state involving N-methylhistamine appears to be stabilized by the surrounding nonpolar residues to a larger extent than with unsubstituted histamine, contributing to a lower barrier with the former.
Collapse
|
18
|
Administration of N,N-dimethyltryptamine (DMT) in psychedelic therapeutics and research and the study of endogenous DMT. Psychopharmacology (Berl) 2022; 239:1749-1763. [PMID: 35064294 PMCID: PMC8782705 DOI: 10.1007/s00213-022-06065-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 01/11/2022] [Indexed: 12/20/2022]
Abstract
As with all drugs, the route, form, and/or dose of a substance administered or applied can play a defining role in its overall pharmacology and use as a therapeutic. This review will focus on these factors as they relate to the psychedelic N,N-dimethyltryptamine (DMT). It will examine the positive and negative aspects of different formulations and routes of administration of DMT and the observed effects from such administrations in the form of ayahuasca teas; oral "pharmahuasca"; injections by intravenous (IV) and intramuscular (IM) routes; inhalation, insufflation; and other routes; and high-dose, low-dose, and "micro-dose" effects. The review will consider possible oral route of administration alternatives that would not require concomitant use of a monoamine oxidase inhibitor. The review will then address the current research findings for DMT from in vivo and in vitro studies as well as the possibility that these findings may be revealing the role of endogenous DMT in normal brain function.
Collapse
|
19
|
Prah A, Mavri J, Stare J. An electrostatic duel: subtle differences in the catalytic performance of monoamine oxidase A and B isoenzymes elucidated at the residue level using quantum computations. Phys Chem Chem Phys 2021; 23:26459-26467. [PMID: 34806105 DOI: 10.1039/d1cp03993h] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The origin of the immense catalytic power of enzymes remains one of the biggest unresolved questions in biochemistry, with electrostatics being one of the main contenders. Herein, we report results that not only confirm that electrostatics is the driving force behind enzyme catalysis, but also that it is capable of tuning subtle differences in the catalytic performance between structurally similar enzymes, as demonstrated using the example of isoenzymes, monoamine oxidases A and B. Using our own computationally efficient multiscale model [A. Prah, et al., ACS Catal., 2019, 9, 1231] we analyzed the rate-limiting step of the reaction between phenylethylamine and both isoenzymes and deduced that the electrostatic environment provided by isoenzyme B has a perceivably higher catalytic influence on all the considered parameters of the reaction (energy barrier, charge transfer, dipole moment, and HOMO-LUMO gap). This is in full agreement with the available experimental kinetic data and with our own simulations of the reaction in question. In-depth analysis of individual amino acid contributions of both isoenzymes to the barrier (based on the interaction between the electric field provided by the enzyme and the dipole moment of the reacting moiety) shows that the majority of the difference between the isoenzymes can be attributed to a small number of sizable differences between the aligned amino acid pairs, whereas in most of the pairs the difference in contribution to the barrier is vanishingly small. These results suggest that electrostatics largely controls the substrate selectivity of enzymes and validates our approach as being capable of discerning fine nuances in the selectivity of structurally related isoenzymes.
Collapse
Affiliation(s)
- Alja Prah
- Theory Department, National Institute of Chemistry, Slovenia. .,University of Ljubljana, Faculty of Pharmacy, Slovenia
| | - Janez Mavri
- Theory Department, National Institute of Chemistry, Slovenia.
| | - Jernej Stare
- Theory Department, National Institute of Chemistry, Slovenia.
| |
Collapse
|
20
|
Abstract
We have structure, a wealth of kinetic data, thousands of chemical ligands and clinical information for the effects of a range of drugs on monoamine oxidase activity in vivo. We have comparative information from various species and mutations on kinetics and effects of inhibition. Nevertheless, there are what seem like simple questions still to be answered. This article presents a brief summary of existing experimental evidence the background and poses questions that remain intriguing for chemists and biochemists researching the chemical enzymology of and drug design for monoamine oxidases (FAD-containing EC 4.1.3.4).
Collapse
|
21
|
Bai A, Shanmugasundaram V, Selkirk JV, Surapaneni S, Dalvie D. Investigation into MAO B-Mediated Formation of CC112273, a Major Circulating Metabolite of Ozanimod, in Humans and Preclinical Species: Stereospecific Oxidative Deamination of ( S)-Enantiomer of Indaneamine (RP101075) by MAO B. Drug Metab Dispos 2021; 49:601-609. [PMID: 34011531 DOI: 10.1124/dmd.121.000447] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/03/2021] [Indexed: 11/22/2022] Open
Abstract
Ozanimod, recently approved for treating relapsing multiple sclerosis, produced a disproportionate, active, MAO B-catalyzed metabolite (CC112273) that showed remarkable interspecies differences and led to challenges in safety testing. This study explored the kinetics of CC112273 formation from its precursor RP101075. Incubations with human liver mitochondrial fractions revealed K Mapp, V max, and intrinsic clearance (Clint) for CC112273 formation to be 4.8 μM, 50.3 pmol/min/mg protein, and 12 μl/min/mg, respectively, whereas Michaelis-Menten constant (K M) with human recombinant MAO B was 1.1 μM. Studies with liver mitochondrial fractions from preclinical species led to K Mapp, V max, and Clint estimates of 3.0, 35, and 33 μM, 80.6, 114, 37.3 pmol/min/mg, and 27.2, 3.25, and 1.14 μl/min/mg in monkey, rat, and mouse, respectively, and revealed marked differences between rodents and primates, primarily attributable to differences in the K M Comparison of Clint estimates revealed monkey to be ∼2-fold more efficient and the mouse and rat to be 11- and 4-fold less efficient than humans in CC112273 formation. The influence of stereochemistry on MAO B-mediated oxidation was also investigated using the R-isomer of RP101075 (RP101074). This showed marked selectivity toward catalysis of the S-isomer (RP101075) only. Docking into MAO B crystal structure suggested that although both the isomers occupied its active site, only the orientation of RP101075 presented the C-H on the α-carbon that was ideal for the C-H bond cleavage, which is a requisite for oxidative deamination. These studies explain the basis for the observed interspecies differences in the metabolism of ozanimod as well as the substrate stereospecificity for formation of CC112273. SIGNIFICANCE STATEMENT: This study evaluates the enzymology and the species differences of the major circulating metabolite of ozanimod, CC112273. Additionally, the study also explores the influence of stereochemistry on MAO B-catalyzed reactions. The study is of significance to the DMD readers given that this oxidation is catalyzed by a non-cytochrome P450 enzyme, and that marked species difference and notable stereospecificity was observed in MAO B-catalyzed biotransformation when the indaneamine enantiomers were used as substrates.
Collapse
Affiliation(s)
- April Bai
- Non-clinical Research and Development (A.B., D.D.) and Neuroscience TRC (J.V.S.), Bristol-Myers Squibb, San Diego, California; Molecular Structure and Design, Bristol-Myers Squibb, Cambridge, Massachusetts (V.S.); and Non-clinical Research and Development, Bristol-Myers Squibb, Summit, New Jersey (S.S.)
| | - Veerabahu Shanmugasundaram
- Non-clinical Research and Development (A.B., D.D.) and Neuroscience TRC (J.V.S.), Bristol-Myers Squibb, San Diego, California; Molecular Structure and Design, Bristol-Myers Squibb, Cambridge, Massachusetts (V.S.); and Non-clinical Research and Development, Bristol-Myers Squibb, Summit, New Jersey (S.S.)
| | - Julie V Selkirk
- Non-clinical Research and Development (A.B., D.D.) and Neuroscience TRC (J.V.S.), Bristol-Myers Squibb, San Diego, California; Molecular Structure and Design, Bristol-Myers Squibb, Cambridge, Massachusetts (V.S.); and Non-clinical Research and Development, Bristol-Myers Squibb, Summit, New Jersey (S.S.)
| | - Sekhar Surapaneni
- Non-clinical Research and Development (A.B., D.D.) and Neuroscience TRC (J.V.S.), Bristol-Myers Squibb, San Diego, California; Molecular Structure and Design, Bristol-Myers Squibb, Cambridge, Massachusetts (V.S.); and Non-clinical Research and Development, Bristol-Myers Squibb, Summit, New Jersey (S.S.)
| | - Deepak Dalvie
- Non-clinical Research and Development (A.B., D.D.) and Neuroscience TRC (J.V.S.), Bristol-Myers Squibb, San Diego, California; Molecular Structure and Design, Bristol-Myers Squibb, Cambridge, Massachusetts (V.S.); and Non-clinical Research and Development, Bristol-Myers Squibb, Summit, New Jersey (S.S.)
| |
Collapse
|
22
|
Sanchez-Andrada P, Vidal-Vidal A, Prieto T, Elguero J, Alkorta I, Marin-Luna M. Alkylammonium Cation Affinities of Nitrogenated Organobases: The Roles of Hydrogen Bonding and Proton Transfer. Chempluschem 2021; 86:1097-1105. [PMID: 34251758 DOI: 10.1002/cplu.202100235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/29/2021] [Indexed: 11/06/2022]
Abstract
Alkylammonium cation affinities of 64 nitrogen-containing organobases, as well as the respective proton transfer processes from the alkylammonium cations to the base, have been computed in the gas phase by using DFT methods. The guanidine bases show the highest proton transfer values (191.9-233 kJ mol-1 ) whereas the cis-2,2'-biimidazole presents the largest affinity towards the alkylammonium cations (>200 kJ mol-1 ) values. The resulting data have been compared with the experimentally reported proton affinities of the studied nitrogen-containing organobases revealing that the propensity of an organobase for the proton transfer process increases linearly with its proton affinity. This work can provide a tool for designing senors for bioactive compounds containing amino groups that are protonated at physiological pH.
Collapse
Affiliation(s)
- Pilar Sanchez-Andrada
- Departamento de Química Orgánica Regional Campus of International Excellence "Campus Mare Nostrum", Universidad de Murcia Facultad de Química, Campus de Espinardo, E-30100, Murcia, Spain
| | - Angel Vidal-Vidal
- Departamento de Química Orgánica, Universidade de Vigo Campus Lagoas-Marcosende, Vigo, Spain
| | - Tania Prieto
- Departamento de Química Orgánica, Universidade de Vigo Campus Lagoas-Marcosende, Vigo, Spain
| | - José Elguero
- Instituto de Química Médica, Centro Superior de Investigaciones Científicas (CSIC), Juan de la Cierva, 3, E-28006, Madrid, Spain
| | - Ibon Alkorta
- Instituto de Química Médica, Centro Superior de Investigaciones Científicas (CSIC), Juan de la Cierva, 3, E-28006, Madrid, Spain
| | - Marta Marin-Luna
- Departamento de Química Orgánica Regional Campus of International Excellence "Campus Mare Nostrum", Universidad de Murcia Facultad de Química, Campus de Espinardo, E-30100, Murcia, Spain
| |
Collapse
|
23
|
Govindasamy H, Magudeeswaran S, Kandasamy S, Poomani K. Binding mechanism of naringenin with monoamine oxidase - B enzyme: QM/MM and molecular dynamics perspective. Heliyon 2021; 7:e06684. [PMID: 33898820 PMCID: PMC8055563 DOI: 10.1016/j.heliyon.2021.e06684] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/31/2020] [Accepted: 03/30/2021] [Indexed: 01/20/2023] Open
Abstract
The reduced level of dopamine at midbrain (substantia nigra) leads to Parkinson disease by the influence of monoamine oxidation process of monoamine oxidase B (MAO-B) enzyme. This disease mostly affects the aged people. Reports outline that the naringenin molecule acts as an inhibitor of MAO-B enzyme and it potentially prevents the development of PD. To elucidate the binding mechanism of naringenin with MAO-B, we performed the molecular docking, QM/MM and molecular dynamics (MD) simulations. The molecular docking results confirm that the naringenin strongly binds with the substrate binding site of MAO-B enzyme (-12.0 kcal/mol). The low values of RMSD, RMSF and Rg indicate that the naringenin - MAO-B complex is stable over the entire period of MD simulation. Naringenin forms strong interaction with the orient keeper residue Tyr326 and other binding site residues Leu171, Glu206 and these interactions were maintained throughout the MD simulation. It is also important to block the function of MAO-B enzyme. The QM/MM study coupled with the charge density analysis reveals the charge density distribution and the strength of intermolecular interactions of naringenin-MAO-B complex. The above results suggest that this molecule is a potential inhibitor of MAO-B enzyme.
Collapse
Affiliation(s)
- Hunday Govindasamy
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, 636 011, India
| | - Sivanandam Magudeeswaran
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, 636 011, India
| | - Saravanan Kandasamy
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, 636 011, India
| | - Kumaradhas Poomani
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, 636 011, India
| |
Collapse
|
24
|
Kubicskó K, Farkas Ö. Quantum chemical (QM:MM) investigation of the mechanism of enzymatic reaction of tryptamine and N,N-dimethyltryptamine with monoamine oxidase A. Org Biomol Chem 2020; 18:9660-9674. [PMID: 33215182 DOI: 10.1039/d0ob01118e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The endogenous psychedelic (mind-altering) N,N-dimethyltryptamine (DMT) molecule has an important role in tissue protection, regeneration, and immunity via sigma-1 receptor activation as its natural ligand. The immunologic properties of DMT suggest this biogenic compound should be investigated thoroughly in other aspects as well. In our in silico project, we examined the metabolism of DMT and its primary analogue, the tryptamine (T), by the monoamine oxidase (MAO) flavoenzyme. MAO has two isoforms, MAO-A and MAO-B. MAOs perform the oxidation of various monoamines by their flavin adenine dinucleotide (FAD) cofactor. Two-layer QM:MM calculations at the ONIOM(M06-2X/6-31++G(d,p):UFF=QEq) level were performed including the whole enzyme to explore the potential energy surface (PES) of the reactions. Our findings reinforced that a hybrid mechanism, a mixture of pure H+ and H- transfer pathways, describes precisely the rate-determining step of amine oxidation as suggested by earlier works. Additionally, our results show that the oxidation of tertiary amine DMT requires a lower activation barrier than the primary amine T. This may reflect a general rule, thus we recommend further investigations. Furthermore, we demonstrated that at pH 7.4 the protonated form of these substrates enter the enzyme. As the deprotonation of substrates is crucial, we presumed protonated cofactor, FADH+, may form. Surprisingly, the activation barriers are much lower compared to FAD with both substrates. Therefore, we suggest further investigations in this direction.
Collapse
Affiliation(s)
- Károly Kubicskó
- Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary.
| | | |
Collapse
|
25
|
Hok L, Mavri J, Vianello R. The Effect of Deuteration on the H 2 Receptor Histamine Binding Profile: A Computational Insight into Modified Hydrogen Bonding Interactions. Molecules 2020; 25:molecules25246017. [PMID: 33353215 PMCID: PMC7766521 DOI: 10.3390/molecules25246017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 01/04/2023] Open
Abstract
We used a range of computational techniques to reveal an increased histamine affinity for its H2 receptor upon deuteration, which was interpreted through altered hydrogen bonding interactions within the receptor and the aqueous environment preceding the binding. Molecular docking identified the area between third and fifth transmembrane α-helices as the likely binding pocket for several histamine poses, with the most favorable binding energy of −7.4 kcal mol−1 closely matching the experimental value of −5.9 kcal mol−1. The subsequent molecular dynamics simulation and MM-GBSA analysis recognized Asp98 as the most dominant residue, accounting for 40% of the total binding energy, established through a persistent hydrogen bonding with the histamine −NH3+ group, the latter further held in place through the N–H∙∙∙O hydrogen bonding with Tyr250. Unlike earlier literature proposals, the important role of Thr190 is not evident in hydrogen bonds through its −OH group, but rather in the C–H∙∙∙π contacts with the imidazole ring, while its former moiety is constantly engaged in the hydrogen bonding with Asp186. Lastly, quantum-chemical calculations within the receptor cluster model and utilizing the empirical quantization of the ionizable X–H bonds (X = N, O, S), supported the deuteration-induced affinity increase, with the calculated difference in the binding free energy of −0.85 kcal mol−1, being in excellent agreement with an experimental value of −0.75 kcal mol−1, thus confirming the relevance of hydrogen bonding for the H2 receptor activation.
Collapse
Affiliation(s)
- Lucija Hok
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia;
| | - Janez Mavri
- Laboratory for Computational Biochemistry and Drug Design, National Institute of Chemistry, SI-1001 Ljubljana, Slovenia;
| | - Robert Vianello
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia;
- Correspondence:
| |
Collapse
|
26
|
Prah A, Purg M, Stare J, Vianello R, Mavri J. How Monoamine Oxidase A Decomposes Serotonin: An Empirical Valence Bond Simulation of the Reactive Step. J Phys Chem B 2020; 124:8259-8265. [PMID: 32845149 PMCID: PMC7520887 DOI: 10.1021/acs.jpcb.0c06502] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/26/2020] [Indexed: 12/15/2022]
Abstract
The enzyme-catalyzed degradation of the biogenic amine serotonin is an essential regulatory mechanism of its level in the human organism. In particular, monoamine oxidase A (MAO A) is an important flavoenzyme involved in the metabolism of monoamine neurotransmitters. Despite extensive research efforts, neither the catalytic nor the inhibition mechanisms of MAO enzymes are currently fully understood. In this article, we present the quantum mechanics/molecular mechanics simulation of the rate-limiting step for the serotonin decomposition, which consists of hydride transfer from the serotonin methylene group to the N5 atom of the flavin moiety. Free-energy profiles of the reaction were computed by the empirical valence bond method. Apart from the enzymatic environment, the reference reaction in the gas phase was also simulated, facilitating the estimation of the catalytic effect of the enzyme. The calculated barrier for the enzyme-catalyzed reaction of 14.82 ± 0.81 kcal mol-1 is in good agreement with the experimental value of 16.0 kcal mol-1, which provides strong evidence for the validity of the proposed hydride-transfer mechanism. Together with additional experimental and computational work, the results presented herein contribute to a deeper understanding of the catalytic mechanism of MAO A and flavoenzymes in general, and in the long run, they should pave the way toward applications in neuropsychiatry.
Collapse
Affiliation(s)
- Alja Prah
- Laboratory
for Computational Biochemistry and Drug Design, National Institute of Chemistry, Ljubljana 1001, Slovenia
- Faculty
of Pharmacy, University of Ljubljana, Ljubljana 1001, Slovenia
| | - Miha Purg
- Department
of Cell and Molecular Biology, Uppsala University, Uppsala SE-751 24, Sweden
| | - Jernej Stare
- Laboratory
for Computational Biochemistry and Drug Design, National Institute of Chemistry, Ljubljana 1001, Slovenia
| | - Robert Vianello
- Division
of Organic Chemistry and Biochemistry, Rud̵er
Bošković Institute, Zagreb 10002, Croatia
| | - Janez Mavri
- Laboratory
for Computational Biochemistry and Drug Design, National Institute of Chemistry, Ljubljana 1001, Slovenia
| |
Collapse
|
27
|
Manzoor S, Hoda N. A comprehensive review of monoamine oxidase inhibitors as Anti-Alzheimer's disease agents: A review. Eur J Med Chem 2020; 206:112787. [PMID: 32942081 DOI: 10.1016/j.ejmech.2020.112787] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/22/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023]
Abstract
Monoamine oxidases (MAO-A and MAO-B) are mammalian flavoenzyme, which catalyze the oxidative deamination of several neurotransmitters like norepinephrine, dopamine, tyramine, serotonin, and some other amines. The oxidative deamination produces several harmful side products like ammonia, peroxides, and aldehydes during the biochemical reaction. The concentration of biochemical neurotransmitter alteration in the brain by MAO is directly related with several neurological disorders like Alzheimer's disease and Parkinson's disease (PD). Activated MAO also contributes to the amyloid beta (Aβ) aggregation by two successive cleft β-secretase and γ-secretase of amyloid precursor protein (APP). Additionally, activated MAO is also involved in aggregation of neurofibrillary tangles and cognitive destruction through the cholinergic neuronal damage and disorder of the cholinergic system. MAO inhibition has general anti-Alzheimer's disease effect as a consequence of oxidative stress reduction prompted by MAO enzymes. In this review, we outlined and addressed recent understanding on MAO enzymes such as their structure, physiological function, catalytic mechanism, and possible therapeutic goals in AD. In addition, it also highlights the current development and discovery of potential MAO inhibitors (MAOIs) from various chemical scaffolds.
Collapse
Affiliation(s)
- Shoaib Manzoor
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi, 110025, India
| | - Nasimul Hoda
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi, 110025, India.
| |
Collapse
|
28
|
Hydride Abstraction as the Rate-Limiting Step of the Irreversible Inhibition of Monoamine Oxidase B by Rasagiline and Selegiline: A Computational Empirical Valence Bond Study. Int J Mol Sci 2020; 21:ijms21176151. [PMID: 32858935 PMCID: PMC7503497 DOI: 10.3390/ijms21176151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/11/2020] [Accepted: 08/21/2020] [Indexed: 12/23/2022] Open
Abstract
Monoamine oxidases (MAOs) catalyze the degradation of a very broad range of biogenic and dietary amines including many neurotransmitters in the brain, whose imbalance is extensively linked with the biochemical pathology of various neurological disorders, and are, accordingly, used as primary pharmacological targets to treat these debilitating cognitive diseases. Still, despite this practical significance, the precise molecular mechanism underlying the irreversible MAO inhibition with clinically used propargylamine inhibitors rasagiline and selegiline is still not unambiguously determined, which hinders the rational design of improved inhibitors devoid of side effects current drugs are experiencing. To address this challenge, we present empirical valence bond QM/MM simulations of the rate-limiting step of the MAO inhibition involving the hydride anion transfer from the inhibitor α-carbon onto the N5 atom of the flavin adenin dinucleotide (FAD) cofactor. The proposed mechanism is strongly supported by the obtained free energy profiles, which confirm a higher reactivity of selegiline over rasagiline, while the calculated difference in the activation Gibbs energies of ΔΔG‡ = 3.1 kcal mol-1 is found to be in very good agreement with that from the measured literature kinact values that predict a 1.7 kcal mol-1 higher selegiline reactivity. Given the similarity with the hydride transfer mechanism during the MAO catalytic activity, these results verify that both rasagiline and selegiline are mechanism-based irreversible inhibitors and offer guidelines in designing new and improved inhibitors, which are all clinically employed in treating a variety of neuropsychiatric and neurodegenerative conditions.
Collapse
|
29
|
Singh L, Kaur A, Garg S, Singh AP, Bhatti R. Protective Effect of Esculetin, Natural Coumarin in Mice Model of Fibromyalgia: Targeting Pro-Inflammatory Cytokines and MAO-A. Neurochem Res 2020; 45:2364-2374. [DOI: 10.1007/s11064-020-03095-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/04/2020] [Accepted: 07/09/2020] [Indexed: 10/23/2022]
|
30
|
Zhao F, Masci D, Ferla S, Varricchio C, Brancale A, Colonna S, Black GW, Turner NJ, Castagnolo D. Monoamine Oxidase (MAO-N) Biocatalyzed Synthesis of Indoles from Indolines Prepared via Photocatalytic Cyclization/Arylative Dearomatization. ACS Catal 2020. [DOI: 10.1021/acscatal.0c01351] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Fei Zhao
- School of Cancer and Pharmaceutical Sciences, King’s College London, London SE1 9NH, United Kingdom
| | - Domiziana Masci
- School of Cancer and Pharmaceutical Sciences, King’s College London, London SE1 9NH, United Kingdom
| | - Salvatore Ferla
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, United Kingdom
| | - Carmine Varricchio
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, United Kingdom
| | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, United Kingdom
| | - Serena Colonna
- School of Cancer and Pharmaceutical Sciences, King’s College London, London SE1 9NH, United Kingdom
| | - Gary W. Black
- Department of Applied Sciences, Northumbria University, Ellison Place, Newcastle upon Tyne NE1 8ST, United Kingdom
| | - Nicholas J. Turner
- School of Chemistry, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Daniele Castagnolo
- School of Cancer and Pharmaceutical Sciences, King’s College London, London SE1 9NH, United Kingdom
| |
Collapse
|
31
|
Herrera-Arozamena C, Estrada-Valencia M, Pérez C, Lagartera L, Morales-García JA, Pérez-Castillo A, Franco-Gonzalez JF, Michalska P, Duarte P, León R, López MG, Mills A, Gago F, García-Yagüe ÁJ, Fernández-Ginés R, Cuadrado A, Rodríguez-Franco MI. Tuning melatonin receptor subtype selectivity in oxadiazolone-based analogues: Discovery of QR2 ligands and NRF2 activators with neurogenic properties. Eur J Med Chem 2020; 190:112090. [DOI: 10.1016/j.ejmech.2020.112090] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/18/2022]
|
32
|
Relevance of Hydrogen Bonds for the Histamine H2 Receptor-Ligand Interactions: A Lesson from Deuteration. Biomolecules 2020; 10:biom10020196. [PMID: 32013143 PMCID: PMC7072573 DOI: 10.3390/biom10020196] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/27/2019] [Accepted: 01/24/2020] [Indexed: 12/13/2022] Open
Abstract
We used a combination of density functional theory (DFT) calculations and the implicit quantization of the acidic N–H and O–H bonds to assess the effect of deuteration on the binding of agonists (2-methylhistamine and 4-methylhistamine) and antagonists (cimetidine and famotidine) to the histamine H2 receptor. The results show that deuteration significantly increases the affinity for 4-methylhistamine and reduces it for 2-methylhistamine, while leaving it unchanged for both antagonists, which is found in excellent agreement with experiments. The revealed trends are interpreted in the light of the altered strength of the hydrogen bonding upon deuteration, known as the Ubbelohde effect, which affects ligand interactions with both active sites residues and solvent molecules preceding the binding, thus providing strong evidence for the relevance of hydrogen bonding for this process. In addition, computations further underline an important role of the Tyr250 residue for the binding. The obtained insight is relevant for the therapy in the context of (per)deuterated drugs that are expected to enter therapeutic practice in the near future, while this approach may contribute towards understanding receptor activation and its discrimination between agonists and antagonists.
Collapse
|
33
|
Prah A, Ogrin P, Mavri J, Stare J. Nuclear quantum effects in enzymatic reactions: simulation of the kinetic isotope effect of phenylethylamine oxidation catalyzed by monoamine oxidase A. Phys Chem Chem Phys 2020; 22:6838-6847. [DOI: 10.1039/d0cp00131g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
By using computational techniques for quantizing nuclear motion one can accurately reproduce kinetic isotope effect of enzymatic reactions, as demonstrated for phenylethylamine oxidation catalyzed by the monoamine oxidase A enzyme.
Collapse
Affiliation(s)
- Alja Prah
- Theory Department
- National Institute of Chemistry
- Ljubljana
- Slovenia
- University of Ljubljana
| | - Peter Ogrin
- Theory Department
- National Institute of Chemistry
- Ljubljana
- Slovenia
- University of Ljubljana
| | - Janez Mavri
- Theory Department
- National Institute of Chemistry
- Ljubljana
- Slovenia
| | - Jernej Stare
- Theory Department
- National Institute of Chemistry
- Ljubljana
- Slovenia
| |
Collapse
|
34
|
How Important Is the Use of Cocaine and Amphetamines in the Development of Parkinson Disease? A Computational Study. Neurotox Res 2019; 37:724-731. [DOI: 10.1007/s12640-019-00149-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/18/2019] [Accepted: 11/29/2019] [Indexed: 11/25/2022]
|
35
|
Path Integral Calculation of the Hydrogen/Deuterium Kinetic Isotope Effect in Monoamine Oxidase A-Catalyzed Decomposition of Benzylamine. Molecules 2019; 24:molecules24234359. [PMID: 31795294 PMCID: PMC6930584 DOI: 10.3390/molecules24234359] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/21/2019] [Accepted: 11/24/2019] [Indexed: 12/03/2022] Open
Abstract
Monoamine oxidase A (MAO A) is a well-known enzyme responsible for the oxidative deamination of several important monoaminergic neurotransmitters. The rate-limiting step of amine decomposition is hydride anion transfer from the substrate α–CH2 group to the N5 atom of the flavin cofactor moiety. In this work, we focus on MAO A-catalyzed benzylamine decomposition in order to elucidate nuclear quantum effects through the calculation of the hydrogen/deuterium (H/D) kinetic isotope effect. The rate-limiting step of the reaction was simulated using a multiscale approach at the empirical valence bond (EVB) level. We applied path integral quantization using the quantum classical path method (QCP) for the substrate benzylamine as well as the MAO cofactor flavin adenine dinucleotide. The calculated H/D kinetic isotope effect of 6.5 ± 1.4 is in reasonable agreement with the available experimental values.
Collapse
|
36
|
Tandarić T, Vianello R. Computational Insight into the Mechanism of the Irreversible Inhibition of Monoamine Oxidase Enzymes by the Antiparkinsonian Propargylamine Inhibitors Rasagiline and Selegiline. ACS Chem Neurosci 2019; 10:3532-3542. [PMID: 31264403 DOI: 10.1021/acschemneuro.9b00147] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Monoamine oxidases (MAOs) are flavin adenine dinucleotide containing flavoenzymes that catalyze the degradation of a range of brain neurotransmitters, whose imbalance is extensively linked with the pathology of various neurological disorders. This is why MAOs have been the central pharmacological targets in treating neurodegeneration for more than 60 years. Still, despite this practical importance, the precise chemical mechanisms underlying the irreversible inhibition of the MAO B isoform with clinical drugs rasagiline (RAS) and selegiline (SEL) remained unknown. Here we employed a combination of MD simulations, MM-GBSA binding free energy evaluations, and QM cluster calculations to show the MAO inactivation proceeds in three steps, where, in the rate-limiting first step, FAD utilizes its N5 atom to abstracts a hydride anion from the inhibitor α-CH2 group to ultimately give the final inhibitor-FAD adduct matching crystallographic data. The obtained free energy profiles reveal a lower activation energy for SEL by 1.2 kcal mol-1 and a higher reaction exergonicity by 0.8 kcal mol-1, with the former being in excellent agreement with experimental ΔΔG‡EXP = 1.7 kcal mol-1, thus rationalizing its higher in vivo reactivity over RAS. The calculated ΔGBIND energies confirm SEL binds better due to its bigger size and flexibility allowing it to optimize hydrophobic C-H···π and π···π interactions with residues throughout both of enzyme's cavities, particularly with FAD, Gln206 and four active site tyrosines, thus overcoming a larger ability of RAS to form hydrogen bonds that only position it in less reactive orientations for the hydride abstraction. Offered results elucidate structural determinants affecting the affinity and rates of the inhibition reaction that should be considered to cooperate when designing more effective compounds devoid of untoward effects, which are of utmost significance and urgency with the growing prevalence of brain diseases.
Collapse
Affiliation(s)
- Tana Tandarić
- Computational Organic Chemistry and Biochemistry Group, Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia
| | - Robert Vianello
- Computational Organic Chemistry and Biochemistry Group, Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia
| |
Collapse
|
37
|
Role of introduced surface cysteine of NADH oxidase from Lactobacillus rhamnosus. Int J Biol Macromol 2019; 132:150-156. [DOI: 10.1016/j.ijbiomac.2019.03.168] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/10/2019] [Accepted: 03/25/2019] [Indexed: 12/15/2022]
|
38
|
Jones HBL, Crean RM, Mullen A, Kendrick EG, Bull SD, Wells SA, Carbery DR, MacMillan F, van der Kamp MW, Pudney CR. Exposing the Interplay Between Enzyme Turnover, Protein Dynamics, and the Membrane Environment in Monoamine Oxidase B. Biochemistry 2019; 58:2362-2372. [PMID: 30964996 DOI: 10.1021/acs.biochem.9b00213] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
There is an increasing realization that structure-based drug design may show improved success by understanding the ensemble of conformations accessible to an enzyme and how the environment affects this ensemble. Human monoamine oxidase B (MAO-B) catalyzes the oxidation of amines and is inhibited for the treatment of both Parkinson's disease and depression. Despite its clinical importance, its catalytic mechanism remains unclear, and routes to drugging this target would be valuable. Evidence of a radical in either the transition state or the resting state of MAO-B is present throughout the literature and is suggested to be a flavin semiquinone, a tyrosyl radical, or both. Here we see evidence of a resting-state flavin semiquinone, via absorption redox studies and electron paramagnetic resonance, suggesting that the anionic semiquinone is biologically relevant. On the basis of enzyme kinetic studies, enzyme variants, and molecular dynamics simulations, we find evidence for the importance of the membrane environment in mediating the activity of MAO-B and that this mediation is related to the protein dynamics of MAO-B. Further, our MD simulations identify a hitherto undescribed entrance for substrate binding, membrane modulated substrate access, and indications for half-site reactivity: only one active site is accessible to binding at a time. Our study combines both experimental and computational evidence to illustrate the subtle interplay between enzyme activity and protein dynamics and the immediate membrane environment. Understanding key biomedical enzymes to this level of detail will be crucial to inform strategies (and binding sites) for rational drug design for these targets.
Collapse
Affiliation(s)
| | | | - Anna Mullen
- School of Chemistry , University of East Anglia , Norwich Research Park , Norwich NR4 7TJ , United Kingdom
| | | | | | | | | | - Fraser MacMillan
- School of Chemistry , University of East Anglia , Norwich Research Park , Norwich NR4 7TJ , United Kingdom
| | - Marc W van der Kamp
- School of Biochemistry , University of Bristol , Biomedical Sciences Building, University Walk , Bristol BS8 1TD , United Kingdom
| | | |
Collapse
|
39
|
Tripathi RKP, Ayyannan SR. Monoamine oxidase-B inhibitors as potential neurotherapeutic agents: An overview and update. Med Res Rev 2019; 39:1603-1706. [PMID: 30604512 DOI: 10.1002/med.21561] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 12/23/2022]
Abstract
Monoamine oxidase (MAO) inhibitors have made significant contributions and remain an indispensable approach of molecular and mechanistic diversity for the discovery of antineurodegenerative drugs. However, their usage has been hampered by nonselective and/or irreversible action which resulted in drawbacks like liver toxicity, cheese effect, and so forth. Hence, the search for selective MAO inhibitors (MAOIs) has become a substantial focus in current drug discovery. This review summarizes our current understanding on MAO-A/MAO-B including their structure, catalytic mechanism, and biological functions with emphases on the role of MAO-B as a potential therapeutic target for the development of medications treating neurodegenerative disorders. It also highlights the recent developments in the discovery of potential MAO-B inhibitors (MAO-BIs) belonging to diverse chemical scaffolds, arising from intensive chemical-mechanistic and computational studies documented during past 3 years (2015-2018), with emphases on their potency and selectivity. Importantly, readers will gain knowledge of various newly established MAO-BI scaffolds and their development potentials. The comprehensive information provided herein will hopefully accelerate ideas for designing novel selective MAO-BIs with superior activity profiles and critical discussions will inflict more caution in the decision-making process in the MAOIs discovery.
Collapse
Affiliation(s)
- Rati Kailash Prasad Tripathi
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India.,Department of Pharmaceutical Chemistry, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Senthil Raja Ayyannan
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| |
Collapse
|
40
|
Prah A, Frančišković E, Mavri J, Stare J. Electrostatics as the Driving Force Behind the Catalytic Function of the Monoamine Oxidase A Enzyme Confirmed by Quantum Computations. ACS Catal 2019. [DOI: 10.1021/acscatal.8b04045] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Alja Prah
- Theory Department, National Institute of Chemistry, 1001 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Eric Frančišković
- Theory Department, National Institute of Chemistry, 1001 Ljubljana, Slovenia
| | - Janez Mavri
- Theory Department, National Institute of Chemistry, 1001 Ljubljana, Slovenia
| | - Jernej Stare
- Theory Department, National Institute of Chemistry, 1001 Ljubljana, Slovenia
| |
Collapse
|
41
|
Umek N, Geršak B, Vintar N, Šoštarič M, Mavri J. Dopamine Autoxidation Is Controlled by Acidic pH. Front Mol Neurosci 2018; 11:467. [PMID: 30618616 PMCID: PMC6305604 DOI: 10.3389/fnmol.2018.00467] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/03/2018] [Indexed: 11/16/2022] Open
Abstract
We studied the reaction mechanism of dopamine autoxidation using quantum chemical methods. Unlike other biogenic amines important in the central nervous system, dopamine and noradrenaline are capable of undergoing a non-enzymatic autoxidative reaction giving rise to a superoxide anion that further decomposes to reactive oxygen species. The reaction in question, which takes place in an aqueous solution, is as such not limited to the mitochondrial membrane where scavenging enzymes such as catalase and superoxide dismutase are located. With the experimental rate constant of 0.147 s−1, the dopamine autoxidation reaction is comparably as fast as the monoamine oxidase B catalyzed dopamine decomposition with a rate constant of 1 s−1. By using quantum chemical calculations, we demonstrated that the rate-limiting step is the formation of a hydroxide ion from a water molecule, which attacks the amino group that enters intramolecular Michael addition, giving rise to a pharmacologically inert aminochrome. We have shown that for dopamine stability on a time scale of days, it is essential that the pH value of the synaptic vesicle interior is acidic. The pathophysiologic correlates of the results are discussed in the context of Parkinson's disease as well as the pathology caused by long-term amphetamine and cocaine administration.
Collapse
Affiliation(s)
- Nejc Umek
- Department of Anesthesiology and Surgical Intensive Therapy, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Blaž Geršak
- Department of Anesthesiology and Surgical Intensive Therapy, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Neli Vintar
- Department of Anesthesiology and Surgical Intensive Therapy, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Department of Anesthesiology and Reanimatology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Maja Šoštarič
- Department of Anesthesiology and Surgical Intensive Therapy, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Department of Anesthesiology and Reanimatology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Janez Mavri
- National Institute of Chemistry, Ljubljana, Slovenia
| |
Collapse
|
42
|
Batista VF, Galman JL, G. A. Pinto DC, Silva AMS, Turner NJ. Monoamine Oxidase: Tunable Activity for Amine Resolution and Functionalization. ACS Catal 2018. [DOI: 10.1021/acscatal.8b03525] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Vasco F. Batista
- Department of Chemistry & QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - James L. Galman
- School of Chemistry, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, U.K
| | - Diana C. G. A. Pinto
- Department of Chemistry & QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Artur M. S. Silva
- Department of Chemistry & QOPNA, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Nicholas J. Turner
- School of Chemistry, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, U.K
| |
Collapse
|
43
|
Montero-Campillo MM, Alkorta I, Elguero J. Enhancement of Thermodynamic Gas-Phase Acidity and Basicity of Water by Means of Secondary Interactions. Chemphyschem 2018; 19:2486-2491. [PMID: 29944196 DOI: 10.1002/cphc.201800518] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Indexed: 11/07/2022]
Abstract
A series of A⋅water, B⋅water complexes (A=acid, B=base) are studied at the G4 level of theory to show that water acidity or basicity can be modulated by non-covalent interactions. Protic and non-protic acids interacting with water form hydrogen bonds or other kinds of non-covalent interactions, respectively, that may dramatically change the acidity of water up to almost 360 kJ ⋅ mol-1 in terms of enthalpy. Similarly, hydrogen bonds responsible for the interaction between typical small nitrogen-containing Lewis bases and water can enhance the proton affinity of water by almost 300 kJ ⋅ mol-1 . Our results reveal that these large enhancements are linearly related with the binding energy of the charged complexes, and are determined by the Lewis acid-base properties of the molecule involved in the interaction, allowing a quite precise modulation of the corresponding acid-base properties of water.
Collapse
Affiliation(s)
- M Merced Montero-Campillo
- Departamento de Química, Módulo 13, Facultad de Ciencias and Institute of Advanced Chemical Sciences (IAdChem), Universidad Autónoma de Madrid, Campus de Excelencia UAM-CSIC, Cantoblanco, E-28049, Madrid, Spain
| | - Ibon Alkorta
- Instituto de Química Médica, CSIC, Juan de la Cierva, 3, E-28006, Madrid, Spain
| | - José Elguero
- Instituto de Química Médica, CSIC, Juan de la Cierva, 3, E-28006, Madrid, Spain
| |
Collapse
|
44
|
Jana K, Ganguly B. DFT Study To Explore the Importance of Ring Size and Effect of Solvents on the Keto-Enol Tautomerization Process of α- and β-Cyclodiones. ACS OMEGA 2018; 3:8429-8439. [PMID: 31458971 PMCID: PMC6644555 DOI: 10.1021/acsomega.8b01008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/18/2018] [Indexed: 06/01/2023]
Abstract
We have explored the effect of ring size on keto-enol tautomerization of α- and β-cyclodiones using the M062X-SMDaq/6-31+G(d,p)//M062X/6-31+G(d,p) level of theory. The calculated results show that the activation free energy barrier for the keto-enol tautomerization process of α-cyclopropanedione (1) is 54.9 kcal/mol, which is lower compared to that of the other cyclic diketo systems studied here. The four-membered α- and β-cyclobutanedione (2 and 6) do not favor keto-enol tautomerization unlike other studied cyclic systems because of the ring strain developed in the transition-state geometries and their corresponding products. Water-assisted keto-enol tautomerization with one molecule reveals that the free energy activation barriers reduce almost half compared to those for the uncatalyzed systems. The two-water-assisted process is favorable as the activation free energy barriers lowered by ∼10 kcal/mol compared to those of the one-water-assisted process. The ion-pair formation seems to govern the lowering of activation barriers of α- and β-cyclodiones with two water molecules during the keto-enol tautomerization process, which however also overcomes the favorable aromatization in the three-membered ring system. The free energy activation barriers calculated with the M062X-SMDaq/6-31+G(d,p) level predicted that the keto-enol tautomerization process for the α-cyclodiones follows the following trend: 2 > 3 > 4 > 5 > 1. Water-assisted tautomerization of α-cyclodiones also predicted 1-W and 1-2W as the most favored processes; however, 5-W and 5-2W were found to be disfavored in this case. The β-cyclodione systems also showed similar trends as obtained with α-diketone systems. The influence of bulk solvent on the keto-enol tautomerization process favors the formation of the enol form in a more polar solvent medium even under mixed solvent conditions in acetonitrile and hexane at M062X-SMDacetonitrile/6-31+G(d,p) and M062X-SMDhexane/6-31+G(d,p) levels of theory.
Collapse
Affiliation(s)
- Kalyanashis Jana
- Computation
and Simulation Unit (Analytical Discipline and Centralized
Instrument Facility) and Academy of Scientific and Innovative Research, CSIR—Central Salt and Marine Chemicals Research
Institute, Bhavnagar 364002, India
| | - Bishwajit Ganguly
- Computation
and Simulation Unit (Analytical Discipline and Centralized
Instrument Facility) and Academy of Scientific and Innovative Research, CSIR—Central Salt and Marine Chemicals Research
Institute, Bhavnagar 364002, India
| |
Collapse
|
45
|
Albreht A, Vovk I, Mavri J, Marco-Contelles J, Ramsay RR. Evidence for a Cyanine Link Between Propargylamine Drugs and Monoamine Oxidase Clarifies the Inactivation Mechanism. Front Chem 2018; 6:169. [PMID: 29892597 PMCID: PMC5985292 DOI: 10.3389/fchem.2018.00169] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/27/2018] [Indexed: 01/06/2023] Open
Abstract
Successful propargylamine drugs such as deprenyl inactivate monoamine oxidase (MAO), a target in multi-faceted approaches to prevent neurodegeneration in the aging population, but the chemical structure and mechanism of the irreversible inhibition are still debated. We characterized the covalent cyanine structure linking the multi-target propargylamine inhibitor ASS234 and the flavin adenine dinucleotide in MAO-A using a combination of ultra-high performance liquid chromatography, spectroscopy, mass spectrometry, and computational methods. The partial double bond character of the cyanine chain gives rise to 4 interconverting geometric isomers of the adduct which were chromatographically separated at low temperatures. The configuration of the cyanine linker governs adduct stability with segments of much higher flexibility and rigidity than previously hypothesized. The findings indicate the importance of intramolecular electrostatic interactions in the MAO binding site and provide key information relevant to incorporation of the propargyl moiety into novel multi-target drugs. Based on the structure, we propose a mechanism of MAO inactivation applicable to all propargylamine inhibitors.
Collapse
Affiliation(s)
- Alen Albreht
- Department of Food Chemistry, National Institute of Chemistry, Ljubljana, Slovenia
| | - Irena Vovk
- Department of Food Chemistry, National Institute of Chemistry, Ljubljana, Slovenia
| | - Janez Mavri
- Laboratory of Computational Biochemistry and Drug Design, Theory Department, National Institute of Chemistry, Ljubljana, Slovenia
| | - Jose Marco-Contelles
- Laboratorio de Química Médica, Instituto de Química Orgánica General (CSIC), Madrid, Spain
| | - Rona R Ramsay
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
46
|
Maršavelski A, Petrović D, Bauer P, Vianello R, Kamerlin SCL. Empirical Valence Bond Simulations Suggest a Direct Hydride Transfer Mechanism for Human Diamine Oxidase. ACS OMEGA 2018; 3:3665-3674. [PMID: 30023875 PMCID: PMC6044848 DOI: 10.1021/acsomega.8b00346] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 03/19/2018] [Indexed: 06/08/2023]
Abstract
Diamine oxidase (DAO) is an enzyme involved in the regulation of cell proliferation and the immune response. This enzyme performs oxidative deamination in the catabolism of biogenic amines, including, among others, histamine, putrescine, spermidine, and spermine. The mechanistic details underlying the reductive half-reaction of the DAO-catalyzed oxidative deamination which leads to the reduced enzyme cofactor and the aldehyde product are, however, still under debate. The catalytic mechanism was proposed to involve a prototropic shift from the substrate-Schiff base to the product-Schiff base, which includes the rate-limiting cleavage of the Cα-H bond by the conserved catalytic aspartate. Our detailed mechanistic study, performed using a combined quantum chemical cluster approach with empirical valence bond simulations, suggests that the rate-limiting cleavage of the Cα-H bond involves direct hydride transfer to the topaquinone cofactor-a mechanism that does not involve the formation of a Schiff base. Additional investigation of the D373E and D373N variants supported the hypothesis that the conserved catalytic aspartate is indeed essential for the reaction; however, it does not appear to serve as the catalytic base, as previously suggested. Rather, the electrostatic contributions of the most significant residues (including D373), together with the proximity of the Cu2+ cation to the reaction site, lower the activation barrier to drive the chemical reaction.
Collapse
Affiliation(s)
- Aleksandra Maršavelski
- Computational
Organic Chemistry and Biochemistry Group, Division of Organic Chemistry
and Biochemistry, Ruđer Bošković
Institute, Bijenička
cesta 54, 10000 Zagreb, Croatia
- Department
of Chemistry, Faculty of Science, University
of Zagreb, Horvatovac
102a, 10000 Zagreb, Croatia
- Department
of Cell and Molecular Biology, Uppsala University, BMC Box 596, S-751 24 Uppsala, Sweden
| | - Dušan Petrović
- Department
of Cell and Molecular Biology, Uppsala University, BMC Box 596, S-751 24 Uppsala, Sweden
| | - Paul Bauer
- Department
of Cell and Molecular Biology, Uppsala University, BMC Box 596, S-751 24 Uppsala, Sweden
- Department
of Biophysics, SciLifeLab, KTH Royal Institute
of Technology, S-10691 Stockholm, Sweden
| | - Robert Vianello
- Computational
Organic Chemistry and Biochemistry Group, Division of Organic Chemistry
and Biochemistry, Ruđer Bošković
Institute, Bijenička
cesta 54, 10000 Zagreb, Croatia
| | | |
Collapse
|
47
|
|
48
|
Dasgupta S, Mukherjee S, Mukhopadhyay BP. Recognition of trans and gauche phenylethylamine conformers in the active site of human monoamine oxidase B: A MD-simulation and DFT studies. COMPUT THEOR CHEM 2018. [DOI: 10.1016/j.comptc.2018.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
49
|
Pregeljc D, Jug U, Mavri J, Stare J. Why does the Y326I mutant of monoamine oxidase B decompose an endogenous amphetamine at a slower rate than the wild type enzyme? Reaction step elucidated by multiscale molecular simulations. Phys Chem Chem Phys 2018; 20:4181-4188. [PMID: 29360121 DOI: 10.1039/c7cp07069a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This work investigates the Y326I point mutation effect on the kinetics of oxidative deamination of phenylethylamine (PEA) catalyzed by the monoamine oxidase B (MAO B) enzyme. PEA is a neuromodulator capable of affecting the plasticity of the brain and is responsible for the mood enhancing effect caused by physical exercise. Due to a similar functionality, PEA is often regarded as an endogenous amphetamine. The rate limiting step of the deamination was simulated at the multiscale level, employing the Empirical Valence Bond approach for the quantum treatment of the involved valence states, whereas the environment (solvated protein) was represented with a classical force field. A comparison of the reaction free energy profiles delivered by simulation of the reaction in the wild type MAO B and its Y326I mutant yields an increase in the barrier by 1.06 kcal mol-1 upon mutation, corresponding to a roughly 6-fold decrease in the reaction rate. This is in excellent agreement with the experimental kinetic studies. Inspection of simulation trajectories reveals possible sources of the point mutation effect, namely vanishing favorable electrostatic interactions between PEA and a Tyr326 side chain and an increased amount of water molecules at the active site due to the replacement of tyrosine by a less spacious isoleucine residue, thereby increasing the dielectric shielding of the catalytic environment provided by the enzyme.
Collapse
Affiliation(s)
- Domen Pregeljc
- Theory Department, National Institute of Chemistry, Ljubljana, Slovenia.
| | | | | | | |
Collapse
|
50
|
Oanca G, Stare J, Mavri J. How fast monoamine oxidases decompose adrenaline? Kinetics of isoenzymes A and B evaluated by empirical valence bond simulation. Proteins 2017; 85:2170-2178. [PMID: 28836294 DOI: 10.1002/prot.25374] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/18/2017] [Accepted: 08/21/2017] [Indexed: 12/22/2022]
Abstract
This work scrutinizes kinetics of decomposition of adrenaline catalyzed by monoamine oxidase (MAO) A and B enzymes, a process controlling the levels of adrenaline in the central nervous system and other tissues. Experimental kinetic data for MAO A and B catalyzed decomposition of adrenaline are reported only in the form of the maximum reaction rate. Therefore, we estimated the experimental free energy barriers form the kinetic data of closely related systems using regression method, as was done in our previous study. By using multiscale simulation on the Empirical Valence Bond (EVB) level, we studied the chemical reactivity of the MAO A catalyzed decomposition of adrenaline and we obtained a value of activation free energy of 17.3 ± 0.4 kcal/mol. The corresponding value for MAO B is 15.7 ± 0.7 kcal/mol. Both values are in good agreement with the estimated experimental barriers of 16.6 and 16.0 kcal/mol for MAO A and MAO B, respectively. The fact that we reproduced the kinetic data and preferential catalytic effect of MAO B over MAO A gives additional support to the validity of the proposed hydride transfer mechanism. Furthermore, we demonstrate that adrenaline is preferably involved in the reaction in a neutral rather than in a protonated form due to considerably higher barriers computed for the protonated adrenaline substrate. The results are discussed in the context of chemical mechanism of MAO enzymes and possible applications of multiscale simulation to rationalize the effects of MAO activity on adrenaline level.
Collapse
Affiliation(s)
- Gabriel Oanca
- Department of Computational Biochemistry and Drug Design, National Institute of Chemistry, Ljubljana, Slovenia.,Faculty of Physics, Alexandru Ioan Cuza University of Iasi, Iasi, Romania
| | - Jernej Stare
- Department of Computational Biochemistry and Drug Design, National Institute of Chemistry, Ljubljana, Slovenia
| | - Janez Mavri
- Department of Computational Biochemistry and Drug Design, National Institute of Chemistry, Ljubljana, Slovenia
| |
Collapse
|