Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochemistry ; 62(15): 2257-2268, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37467463

RESUMO

Phenylethanolamine N-methyltransferase (PNMT) catalyzes the S-adenosyl-l-methionine (SAM)-dependent methylation of norepinephrine to form epinephrine. Epinephrine is implicated in the regulation of blood pressure, respiration, Alzheimer's disease, and post-traumatic stress disorder (PTSD). Transition-state (TS) analogues bind their target enzymes orders of magnitude more tightly than their substrates. A synthetic strategy for first-generation TS analogues of human PNMT (hPNMT) permitted structural analysis of hPNMT and revealed potential for second-generation inhibitors [Mahmoodi, N.; J. Am. Chem. Soc. 2020, 142, 14222-14233]. A second-generation TS analogue inhibitor of PNMT was designed, synthesized, and characterized to yield a Ki value of 1.2 nM. PNMT isothermal titration calorimetry (ITC) measurements of inhibitor 4 indicated a negative cooperative binding mechanism driven by large favorable entropic contributions and smaller enthalpic contributions. Cell-based assays with HEK293T cells expressing PNMT revealed a cell permeable, intracellular PNMT inhibitor with an IC50 value of 81 nM. Structural analysis demonstrated inhibitor 4 filling catalytic site regions to recapitulate both norepinephrine and SAM interactions. Conformation of the second-generation inhibitor in the catalytic site of PNMT improves contacts relative to those from the first-generation inhibitors. Inhibitor 4 demonstrates up to 51,000-fold specificity for PNMT relative to DNA and protein methyltransferases. Inhibitor 4 also exhibits a 12,000-fold specificity for PNMT over the α2-adrenoceptor.


Assuntos
Norepinefrina , Feniletanolamina N-Metiltransferase , Humanos , Feniletanolamina N-Metiltransferase/química , Feniletanolamina N-Metiltransferase/metabolismo , Células HEK293 , Epinefrina , Domínio Catalítico
2.
J Med Chem ; 64(8): 4553-4566, 2021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33843213

RESUMO

Finasteride, a 5-alpha reductase (5α-R) inhibitor, is a widely used drug for treating androgen-dependent conditions. However, its use is associated with sexual, psychological, and physical complaints, suggesting that other mechanisms, in addition to 5α-R inhibition, may be involved. Here, a multidisciplinary approach has been used to identify potential finasteride off-target proteins. SPILLO-PBSS software suggests an additional inhibitory activity of finasteride on phenylethanolamine N-methyltransferase (PNMT), the limiting enzyme in formation of the stress hormone epinephrine. The interaction of finasteride with PNMT was supported by docking and molecular dynamics analysis and by in vitro assay, confirming the inhibitory nature of the binding. Finally, this inhibition was also confirmed in an in vivo rat model. Literature data indicate that PNMT activity perturbation may be correlated with sexual and psychological side effects. Therefore, results here obtained suggest that the binding of finasteride to PNMT might have a role in producing the side effects exerted by finasteride treatment.


Assuntos
Inibidores de 5-alfa Redutase/química , Finasterida/química , Feniletanolamina N-Metiltransferase/metabolismo , Inibidores de 5-alfa Redutase/metabolismo , Inibidores de 5-alfa Redutase/farmacologia , Animais , Sítios de Ligação , Ligação Competitiva , Catecolaminas/análise , Catecolaminas/metabolismo , Cromatografia Líquida de Alta Pressão , Bases de Dados de Proteínas , Epinefrina/metabolismo , Finasterida/metabolismo , Finasterida/farmacologia , Humanos , Masculino , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Feniletanolamina N-Metiltransferase/química , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas em Tandem , Termodinâmica
3.
J Med Chem ; 63(22): 13878-13898, 2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33147410

RESUMO

The enzyme phenylethanolamine N-methyltransferase (PNMT, EC 2.1.1.28) catalyzes the final step in the biosynthesis of epinephrine and is a potential drug target, primarily for the control of hypertension. Unfortunately, many potent PNMT inhibitors also possess significant affinity for the a2-adrenoceptor, which complicates the interpretation of their pharmacology. A bisubstrate analogue approach offers the potential for development of highly selective inhibitors of PNMT. This paper documents the design, synthesis, and evaluation of such analogues, several of which were found to possess human PNMT (hPNMT) inhibitory potency <5 nM versus AdoMet. Site-directed mutagenesis studies were consistent with bisubstrate binding. Two of these compounds (19 and 29) were co-crystallized with hPNMT and the resulting structures revealed both compounds bound as predicted, simultaneously occupying both substrate binding domains. This bisubstrate inhibitor approach has resulted in one of the most potent (20) and selective (vs the a2-adrenoceptor) inhibitors of hPNMT yet reported.


Assuntos
Adenosina/farmacologia , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Isoquinolinas/metabolismo , Feniletanolamina N-Metiltransferase/química , Feniletanolamina N-Metiltransferase/metabolismo , S-Adenosilmetionina/metabolismo , Adenosina/química , Sítios de Ligação , Cristalografia por Raios X , Inibidores Enzimáticos/química , Humanos , Modelos Moleculares , Domínios Proteicos , Relação Estrutura-Atividade
4.
J Am Chem Soc ; 142(33): 14222-14233, 2020 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-32702980

RESUMO

Phenylethanolamine N-methyltransferase (PNMT) is a critical enzyme in catecholamine synthesis. It transfers the methyl group of S-adenosylmethionine (SAM) to catalyze the synthesis of epinephrine from norepinephrine. Epinephrine has been associated with diverse human processes, including the regulation of blood pressure and respiration, as well as neurodegeneration found in Alzheimer's disease. Human PNMT (hPNMT) proceeds through an SN2 transition state (TS) in which the transfer of the methyl group is rate limiting. TS analogue enzyme inhibitors are specific for their target and bind orders of magnitude more tightly than their substrates. Molecules resembling the TS of hPNMT were designed, synthesized, and kinetically characterized. This new inhibitory scaffold was designed to mimic the geometry and electronic properties of the hPNMT TS. Synthetic efforts resulted in a tight-binding inhibitor with a Ki value of 12.0 nM. This is among the first of the TS analogue inhibitors of methyltransferase enzymes to show an affinity in the nanomolar range. Isothermal titration calorimetry (ITC) measurements indicated negative cooperative binding of inhibitor to the dimeric protein, driven by favorable entropic contributions. Structural analysis revealed that inhibitor 3 binds to hPNMT by filling the catalytic binding pockets for the cofactor (SAM) and the substrate (norepinephrine) binding sites.


Assuntos
Inibidores Enzimáticos/farmacologia , Feniletanolamina N-Metiltransferase/antagonistas & inibidores , Calorimetria , Inibidores Enzimáticos/química , Humanos , Estrutura Molecular , Feniletanolamina N-Metiltransferase/química , Feniletanolamina N-Metiltransferase/metabolismo
5.
J Chem Theory Comput ; 14(6): 3321-3331, 2018 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-29768914

RESUMO

Cryptic (hidden) pockets are sites that are not visible on unliganded target proteins' structures and only become apparent when a ligand binds. They might provide a valid alternative to classical binding sites in otherwise "undruggable" targets, but their hidden nature makes it difficult to use standard structure-based or computer-aided drug discovery approaches. Our group recently developed a Hamiltonian replica-exchange method (sampling water interfaces through scaled Hamiltonians or SWISH) that improves the sampling of hydrophobic cavities by scaling the interactions between water molecules and protein atoms. Here, we discuss further improvements to SWISH and its combination with fragment probe simulations. We tested the robustness and general applicability of the improved approach in a variety of pharmaceutically relevant targets. The chosen proteins: NPC2, p38α, LfrR, and hPNMT, represent a set of diversified and interesting targets harboring nontrivial cryptic binding sites. In all cases, the updated version of our algorithm efficiently explored the cryptic sites.


Assuntos
Algoritmos , Proteínas de Transporte/metabolismo , Glicoproteínas/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Feniletanolamina N-Metiltransferase/metabolismo , Sítios de Ligação , Proteínas de Transporte/química , Glicoproteínas/química , Humanos , Proteína Quinase 14 Ativada por Mitógeno/química , Simulação de Dinâmica Molecular , Feniletanolamina N-Metiltransferase/química , Ligação Proteica , Estrutura Secundária de Proteína , Proteínas de Transporte Vesicular
6.
ACS Chem Biol ; 12(2): 342-346, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-27997103

RESUMO

Phenylethanolamine N-methyltransferase (PNMT) catalyzes the S-adenosyl-l-methionine (SAM)-dependent conversion of norepinephrine to epinephrine. Epinephrine has been associated with critical processes in humans including the control of respiration and blood pressure. Additionally, PNMT activity has been suggested to play a role in hypertension and Alzheimer's disease. In the current study, labeled SAM substrates were used to measure primary methyl-14C and 36S and secondary methyl-3H, 5'-3H, and 5'-14C intrinsic kinetic isotope effects for human PNMT. The transition state of human PNMT was modeled by matching kinetic isotope effects predicted via quantum chemical calculations to intrinsic values. The model provides information on the geometry and electrostatics of the human PNMT transition state structure and indicates that human PNMT catalyzes the formation of epinephrine through an early SN2 transition state in which methyl transfer is rate-limiting.


Assuntos
Feniletanolamina N-Metiltransferase/química , Humanos , Isótopos , Cinética , Conformação Proteica
7.
J Chem Phys ; 145(15): 154114, 2016 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-27782485

RESUMO

In molecular dynamics (MD) simulations, free-energy differences are often calculated using free energy perturbation or thermodynamic integration (TI) methods. However, both techniques are only suited to calculate free-energy differences between two end states. Enveloping distribution sampling (EDS) presents an attractive alternative that allows to calculate multiple free-energy differences in a single simulation. In EDS, a reference state is simulated which "envelopes" the end states. The challenge of this methodology is the determination of optimal reference-state parameters to ensure equal sampling of all end states. Currently, the automatic determination of the reference-state parameters for multiple end states is an unsolved issue that limits the application of the methodology. To resolve this, we have generalised the replica-exchange EDS (RE-EDS) approach, introduced by Lee et al. [J. Chem. Theory Comput. 10, 2738 (2014)] for constant-pH MD simulations. By exchanging configurations between replicas with different reference-state parameters, the complexity of the parameter-choice problem can be substantially reduced. A new robust scheme to estimate the reference-state parameters from a short initial RE-EDS simulation with default parameters was developed, which allowed the calculation of 36 free-energy differences between nine small-molecule inhibitors of phenylethanolamine N-methyltransferase from a single simulation. The resulting free-energy differences were in excellent agreement with values obtained previously by TI and two-state EDS simulations.


Assuntos
Simulação de Dinâmica Molecular , Termodinâmica , Feniletanolamina N-Metiltransferase/química , Bibliotecas de Moléculas Pequenas/química
8.
Arch Biochem Biophys ; 539(1): 1-8, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24018397

RESUMO

Phenylethanolamine N-methyltransferase (PNMT) catalyzes the conversion of norepinephrine (noradrenaline) to epinephrine (adrenaline) while, concomitantly, S-adenosyl-L-methionine (AdoMet) is converted to S-adenosyl-L-homocysteine. This reaction represents the terminal step in catecholamine biosynthesis and inhibitors of PNMT have been investigated, inter alia, as potential antihypertensive agents. At various times the kinetic mechanism of PNMT has been reported to operate by a random mechanism, an ordered mechanism in which norepinephrine binds first, and an ordered mechanism in which AdoMet binds first. Here we report the results of initial velocity studies on human PNMT in the absence and presence of product and dead end inhibitors. These, coupled with isothermal titration calorimetry and fluorescence binding experiments, clearly shown that hPNMT operates by an ordered sequential mechanism in which AdoMet binds first. Although the logV pH-profile was not well defined, plots of logV/K versus pH for AdoMet and phenylethanolamine, as well as the pKi versus pH for the inhibitor, SK&F 29661, were all bell-shaped indicating that a protonated and an unprotonated group are required for catalysis.


Assuntos
Feniletanolamina N-Metiltransferase/metabolismo , Biocatálise , Inibidores Enzimáticos/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Isoquinolinas/farmacologia , Cinética , Modelos Moleculares , Feniletanolamina N-Metiltransferase/antagonistas & inibidores , Feniletanolamina N-Metiltransferase/química , Conformação Proteica
9.
Int J Mol Sci ; 13(10): 12857-79, 2012 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-23202926

RESUMO

Fragment-based drug discovery (FBDD) concerns the screening of low-molecular weight compounds against macromolecular targets of clinical relevance. These compounds act as starting points for the development of drugs. FBDD has evolved and grown in popularity over the past 15 years. In this paper, the rationale and technology behind the use of X-ray crystallography in fragment based screening (FBS) will be described, including fragment library design and use of synchrotron radiation and robotics for high-throughput X-ray data collection. Some recent uses of crystallography in FBS will be described in detail, including interrogation of the drug targets ß-secretase, phenylethanolamine N-methyltransferase, phosphodiesterase 4A and Hsp90. These examples provide illustrations of projects where crystallography is straightforward or difficult, and where other screening methods can help overcome the limitations of crystallography necessitated by diffraction quality.


Assuntos
Proteínas/química , Secretases da Proteína Precursora do Amiloide/química , Sítios de Ligação , Cristalografia por Raios X , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/química , Desenho de Fármacos , Proteínas de Choque Térmico HSP90/química , Preparações Farmacêuticas/química , Preparações Farmacêuticas/metabolismo , Feniletanolamina N-Metiltransferase/química , Estrutura Terciária de Proteína , Proteínas/metabolismo
10.
Biochim Biophys Acta ; 1824(4): 533-41, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22326747

RESUMO

Epinephrine is a naturally occurring adrenomedullary hormone that transduces environmental stressors into cardiovascular actions. As the only route in the catecholamine biosynthetic pathway, Phenylethanolamine N-methyltransferase (PNMT) catalyzes the synthesis of epinephrine. To elucidate the detailed mechanism of enzymatic catalysis of PNMT, combined quantum-mechanical/molecular-mechanical (QM/MM) calculations were performed. The calculation results reveal that this catalysis contains three elementary steps: the deprotonation of protonated norepinphrine, the methyl transferring step and deprotonation of the methylated norepinphrine. The methyl transferring step was proved to be the rate-determining step undergoing a SN2 mechanism with an energy barrier of 16.4kcal/mol. During the whole catalysis, two glutamic acids Glu185 and Glu219 were proved to be loaded with different effects according to the calculations results of the mutants. These calculation results can be used to explain the experimental observations and make a good complementarity for the previous QM study.


Assuntos
Simulação por Computador , Modelos Químicos , Feniletanolamina N-Metiltransferase/química , Biocatálise , Domínio Catalítico , Ácido Glutâmico/química , Humanos , Cinética , Metilação , Modelos Moleculares , Nordefrin/química , Oxirredução , Teoria Quântica , Termodinâmica
11.
Biochem J ; 431(1): 51-61, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20642456

RESUMO

CNS (central nervous system) adrenaline (epinephrine) is implicated in a wide range of physiological and pathological conditions. PNMT (phenylethanolamine N-methyltransferase) catalyses the final step in the biosynthesis of adrenaline, the conversion of noradrenaline (norepinephrine) to adrenaline by methylation. To help elucidate the role of CNS adrenaline, and to develop potential drug leads, potent, selective and CNS-active inhibitors are required. The fragment screening approach has advantages over other lead discovery methods including high hit rates, more efficient hits and the ability to sample chemical diversity more easily. In the present study we applied fragment-based screening approaches to the enzyme PNMT. We used crystallography as the primary screen and identified 12 hits from a small commercial library of 384 drug-like fragments. The hits include nine chemicals with two fused rings and three single-ring chemical systems. Eight of the hits come from three chemical classes: benzimidazoles (a known class of PNMT inhibitor), purines and quinolines. Nine of the hits have measurable binding affinities (~5-700 µM) as determined by isothermal titration calorimetry and all nine have ligand efficiencies of 0.39 kcal/mol per heavy atom or better (1 kcal≈4.184 kJ). We synthesized five elaborated benzimidazole compounds and characterized their binding to PNMT, showing for the first time how this class of inhibitors interact with the noradrenaline-binding site. Finally, we performed a pilot study with PNMT for fragment-based screening by MS showing that this approach could be used as a fast and efficient first-pass screening method prior to characterization of binding mode and affinity of hits.


Assuntos
Inibidores Enzimáticos/química , Feniletanolamina N-Metiltransferase/antagonistas & inibidores , Feniletanolamina N-Metiltransferase/química , Benzimidazóis/química , Sítios de Ligação , Calorimetria , Cristalografia por Raios X , Cinética , Ligantes , Espectrometria de Massas , Modelos Moleculares , Feniletanolamina N-Metiltransferase/metabolismo
12.
Biochim Biophys Acta ; 1794(12): 1831-7, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19733262

RESUMO

Hybrid density functional theory methods were used to investigate the reaction mechanism of human phenylethanolamine N-methyltransferase (hPNMT). This enzyme catalyzes the S-adenosyl-L-methionine-dependent conversion of norepinephrine to epinephrine, which constitutes the terminal step in the catecholamine biosynthesis. Several models of the active site were constructed based on the X-ray structure. Geometries of the stationary points along the reaction path were optimized and the reaction barrier and energy were calculated and compared to the experimental values. The calculations demonstrate that the reaction takes place via an SN2 mechanism with methyl transfer being rate-limiting, a suggestion supported by mutagenesis studies. Optimal agreement with experimental data is reached using a model in which both active site glutamates are protonated. Overall, the mechanism of hPNMT is more similar to those of catechol O-methyltransferase and glycine N-methyltransferase than to that of guanidinoacetate N-methyltransferase in which methyl transfer is coupled to proton transfer.


Assuntos
Feniletanolamina N-Metiltransferase/química , Feniletanolamina N-Metiltransferase/metabolismo , Substituição de Aminoácidos , Domínio Catalítico , Catecol O-Metiltransferase/química , Catecol O-Metiltransferase/metabolismo , Cristalografia por Raios X , Glicina N-Metiltransferase/química , Glicina N-Metiltransferase/metabolismo , Guanidinoacetato N-Metiltransferase/química , Guanidinoacetato N-Metiltransferase/metabolismo , Humanos , Técnicas In Vitro , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Feniletanolamina N-Metiltransferase/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Termodinâmica
13.
Biochem J ; 422(3): 463-71, 2009 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-19570037

RESUMO

Substrate specificity is critically important for enzyme catalysis. In the adrenaline-synthesizing enzyme PNMT (phenylethanolamine N-methyltransferase), minor changes in substituents can convert substrates into inhibitors. Here we report the crystal structures of six human PNMT complexes, including the first structure of the enzyme in complex with its physiological ligand R-noradrenaline. Determining this structure required rapid soak methods because of the tendency for noradrenaline to oxidize. Comparison of the PNMT-noradrenaline complex with the previously determined PNMT-p-octopamine complex demonstrates that these two substrates form almost equivalent interactions with the enzyme and show that p-octopamine is a valid model substrate for PNMT. The crystal structures illustrate the adaptability of the PNMT substrate binding site in accepting multi-fused ring systems, such as substituted norbornene, as well as noradrenochrome, the oxidation product of noradrenaline. These results explain why only a subset of ligands recognized by PNMT are methylated by the enzyme; bulky substituents dictate the binding orientation of the ligand and can thereby place the acceptor amine too far from the donor methyl group for methylation to occur. We also show how the critical Glu(185) catalytic residue can be replaced by aspartic acid with a loss of only 10-fold in catalytic efficiency. This is because protein backbone movements place the Asp(185) carboxylate almost coincident with the carboxylate of Glu(185). Conversely, replacement of Glu(185) by glutamine reduces catalytic efficiency almost 300-fold, not only because of the loss of charge, but also because the variant residue does not adopt the same conformation as Glu(185).


Assuntos
Epinefrina/biossíntese , Norepinefrina/metabolismo , Feniletanolamina N-Metiltransferase/química , Feniletanolamina N-Metiltransferase/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Humanos , Ligação Proteica , Especificidade por Substrato
14.
J Med Chem ; 50(20): 4845-53, 2007 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-17845018

RESUMO

Shape complementarity is a fundamental principle of inhibitor design. Here we show that an enzyme for which the crystal structure has been determined (phenylethanolamine N-methyltransferase, PNMT) conceals a cryptic binding site. This site is revealed upon binding of inhibitors that are double the size of the physiological substrate. These large inhibitors are not predicted to bind in that they protrude through the accessible surface calculated from a PNMT/7-aminosulfonyl-1,2,3,4-tetrahydroisoquinoline (SK&F 29661) crystal structure, yet they are potent inhibitors of PNMT. We determined structures of the enzyme complexed with large inhibitors and found that the volume of the active site increases by 140 A3 upon binding. Changes in active site size and shape are brought about by unfavorable side chain conformations and rigid body helix motions. The energetic cost is modest, estimated at 2-3 kcal/mol from mutational analyses. Our findings further underline the importance of protein flexibility in structure-based inhibitor design studies.


Assuntos
Isoquinolinas/química , Modelos Moleculares , Feniletanolamina N-Metiltransferase/antagonistas & inibidores , Feniletanolamina N-Metiltransferase/química , Sítios de Ligação , Cristalização , Cristalografia por Raios X , Estrutura Molecular , Mutação , Feniletanolamina N-Metiltransferase/genética , Ligação Proteica , Conformação Proteica , Termodinâmica
15.
J Med Chem ; 49(18): 5424-33, 2006 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-16942016

RESUMO

3-Fluoromethyl-7-(N-substituted aminosulfonyl)-1,2,3,4-tetrahydroisoquinolines (14, 16, and 18-22) are highly potent and selective inhibitors of phenylethanolamine N-methyltransferase (PNMT). Molecular modeling studies with 3-fluoromethyl-7-(N-alkyl aminosulfonyl)-1,2,3,4-tetrahydroisoquinolines, such as 16, suggested that the sulfonamide -NH- could form a hydrogen bond with the side chain of Lys57. However, SAR studies and analysis of the crystal structure of human PNMT (hPNMT) in complex with 7 indicated that the sulfonamide oxygens, and not the sulfonamide -NH-, formed favorable interactions with the enzyme. Thus, we hypothesized that replacement of the sulfonamide -NH- with a methylene group could result in compounds that would retain potency at PNMT and that would have increased lipophilicity, thus increasing the likelihood they will cross the blood brain barrier. A series of 3-fluoromethyl-7-sulfonyl-1,2,3,4-tetrahydroisoquinolines (23-30) were synthesized and evaluated for their PNMT inhibitory potency and affinity for the alpha2-adrenoceptor. A comparison of these compounds with their isosteric sulfonamides (14, 16, and 18-22) showed that the sulfones were more lipophilic but less potent than their corresponding sulfonamides. Sulfone 24 (hPNMT Ki = 1.3 microM) is the most potent compound in this series and is quite selective for PNMT versus the alpha2-adrenoceptor, but 24 is less potent than the corresponding sulfonamide, 16 (hPNMT Ki = 0.13 microM). We also report the crystal structure of hPNMT in complex with sulfonamide 15, from which a potential hydrogen bond acceptor within the hPNMT active site has been identified, the main chain carbonyl oxygen of Asn39. The interaction of this residue with the sulfonamide -NH- is likely responsible for much of the enhanced inhibitory potency of the sulfonamides versus the sulfones.


Assuntos
Isoquinolinas/síntese química , Feniletanolamina N-Metiltransferase/antagonistas & inibidores , Sulfonamidas/síntese química , Sulfonas/síntese química , Animais , Sítios de Ligação , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Cristalografia por Raios X , Humanos , Ligação de Hidrogênio , Técnicas In Vitro , Isoquinolinas/química , Isoquinolinas/farmacologia , Masculino , Modelos Moleculares , Estrutura Molecular , Feniletanolamina N-Metiltransferase/química , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 2/metabolismo , Estereoisomerismo , Relação Estrutura-Atividade , Sulfonamidas/química , Sulfonamidas/farmacologia , Sulfonas/química , Sulfonas/farmacologia
16.
J Med Chem ; 49(10): 2939-52, 2006 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-16686536

RESUMO

3-Methyl-1,2,3,4-tetrahydroisoquinolines (3-methyl-THIQs) are potent inhibitors of phenylethanolamine N-methyltransferase (PNMT), but are not selective due to significant affinity for the alpha(2)-adrenoceptor. Fluorination of the methyl group lowers the pK(a) of the THIQ amine from 9.53 (CH(3)) to 7.88 (CH(2)F), 6.42 (CHF(2)), and 4.88 (CF(3)). This decrease in pK(a) results in a reduction in affinity for the alpha(2)-adrenoceptor. However, increased fluorination also results in a reduction in PNMT inhibitory potency, apparently due to steric and electrostatic factors. Biochemical evaluation of a series of 3-fluoromethyl-THIQs and 3-trifluoromethyl-THIQs showed that the former were highly potent inhibitors of PNMT, but were often nonselective due to significant affinity for the alpha(2)-adrenoceptor, while the latter were devoid of alpha(2)-adrenoceptor affinity, but also lost potency at PNMT. 3-Difluoromethyl-7-substituted-THIQs have the proper balance of both steric and pK(a) properties and thus have enhanced selectivity versus the corresponding 3-fluoromethyl-7-substituted-THIQs and enhanced PNMT inhibitory potency versus the corresponding 3-trifluoromethyl-7-substituted-THIQs. Using the "Goldilocks Effect" analogy, the 3-fluoromethyl-THIQs are too potent (too hot) at the alpha(2)-adrenoceptor and the 3-trifluoromethyl-THIQs are not potent enough (too cold) at PNMT, but the 3-difluoromethyl-THIQs are just right. They are both potent inhibitors of PNMT and highly selective due to low affinity for the alpha(2)-adrenoceptor. This seems to be the first successful use of the beta-fluorination of aliphatic amines to impart selectivity to a pharmacological agent while maintaining potency at the site of interest.


Assuntos
Flúor , Feniletanolamina N-Metiltransferase/antagonistas & inibidores , Feniletanolamina N-Metiltransferase/química , Tetra-Hidroisoquinolinas/síntese química , Animais , Encéfalo/metabolismo , Cristalografia por Raios X , Desenho de Fármacos , Humanos , Masculino , Modelos Moleculares , Estrutura Molecular , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 2/metabolismo , Relação Estrutura-Atividade , Tetra-Hidroisoquinolinas/química , Tetra-Hidroisoquinolinas/farmacologia
17.
Biochemistry ; 44(51): 16875-85, 2005 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-16363801

RESUMO

Here we report three crystal structure complexes of human phenylethanolamine N-methyltransferase (PNMT), one bound with a substrate that incorporates a flexible ethanolamine side chain (p-octopamine), a second bound with a semirigid analogue substrate [cis-(1R,2S)-2-amino-1-tetralol, cis-(1R,2S)-AT], and a third with trans-(1S,2S)-2-amino-1-tetralol [trans-(1S,2S)-AT] that acts as an inhibitor of PNMT rather than a substrate. A water-mediated interaction between the critical beta-hydroxyl of the flexible ethanolamine group of p-octopamine and an acidic residue, Asp267, is likely to play a key role in positioning the side chain correctly for methylation to occur at the amine. A second interaction with Glu219 may play a lesser role. Catalysis likely occurs via deprotonation of the amine through the action of Glu185; mutation of this residue significantly reduced the kcat without affecting the Km. The mode of binding of cis-(1R,2S)-AT supports the notion that this substrate is a conformationally restrained analogue of flexible PNMT substrates, in that it forms interactions with the enzyme similar to those observed for p-octopamine. By contrast, trans-(1S,2S)-AT, an inhibitor rather than a substrate, binds in an orientation that is flipped by 180 degrees compared with cis-(1R,2S)-AT. A consequence of this flipped binding mode is that the interactions between the hydroxyl and Asp267 and Glu219 are lost. However, the amines of inhibitor trans-(1S,2S)-AT and substrate cis-(1R,2S)-AT are both within methyl transfer distance of the cofactor. These results suggest that PNMT catalyzes transfer of methyl to ligand amines only when "anchor" interactions, such as those identified for the beta-hydroxyls of p-octopamine and cis-AT, are present.


Assuntos
Feniletanolamina N-Metiltransferase/química , Substituição de Aminoácidos , Catálise , Domínio Catalítico/genética , Cristalografia por Raios X , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Epinefrina/biossíntese , Glutamina/química , Glutamina/genética , Humanos , Isoquinolinas/química , Isoquinolinas/metabolismo , Cinética , Modelos Químicos , Modelos Moleculares , Conformação Molecular , Estrutura Molecular , Mutação/genética , Octopamina/química , Octopamina/metabolismo , Feniletanolamina N-Metiltransferase/genética , Feniletanolamina N-Metiltransferase/metabolismo , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Eletricidade Estática , Tetralonas/química
18.
J Med Chem ; 48(23): 7243-52, 2005 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-16279783

RESUMO

The X-ray structure of human phenylethanolamine N-methyltransferase (hPNMT) complexed with its product, S-adenosyl-L-homocysteine (4), and the most potent inhibitor reported to date, SK&F 64139 (7), was used to identify the residues involved in inhibitor binding. Four of these residues, Val53, Lys57, Glu219 and Asp267, were replaced, in turn, with alanine. All variants had increased Km values for phenylethanolamine (10), but only D267A showed a noteworthy (20-fold) decrease in its kcat value. Both WT hPNMT and D267A had similar kcat values for a rigid analogue, anti-9-amino-6-(trifluoromethyl)benzonorbornene (12), suggesting that Asp267 plays an important role in positioning the substrate but does not participate directly in catalysis. The Ki values for the binding of inhibitors such as 7 to the E219A and D267A variants increased by 2-3 orders of magnitude. Further, the inhibitors were shown to bind up to 50-fold more tightly in the presence of S-adenosyl-L-methionine (3), suggesting that the binding of the latter brings about a conformational change in the enzyme.


Assuntos
2-Hidroxifenetilamina/química , Feniletanolamina N-Metiltransferase/antagonistas & inibidores , Feniletanolamina N-Metiltransferase/química , S-Adenosil-Homocisteína/química , Tetra-Hidroisoquinolinas/química , Sítios de Ligação , Catálise , Cristalografia por Raios X , Humanos , Cinética , Modelos Moleculares , Mutação , Feniletanolamina N-Metiltransferase/genética , Ligação Proteica , S-Adenosilmetionina/química
19.
Biochim Biophys Acta ; 1750(1): 82-92, 2005 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15893506

RESUMO

The crystal structure of human phenylethanolamine N-methyltransferase (hPNMT) reveals a disulfide-linked dimer, despite the presence of reducing agent in the crystallisation conditions. By removing the reducing agent, hPNMT crystals grow more rapidly and at lower protein concentrations. However, it was unclear whether the disulfide bonds are only present in the crystal form or whether these affect enzyme activity. The solution oligomeric state of hPNMT was investigated using biochemical techniques and activity assays. We found that in the absence of reducing agent, hPNMT forms dimers in solution. Furthermore, the solution dimer of hPNMT incorporates disulfide bonds, since this form is sensitive to reducing agent. The C48A and C139A mutants of hPNMT, which are incapable of forming the disulfide bond observed in the crystal structure, have a decreased propensity to form dimer in solution. Those dimers that do form are also sensitive to reducing agent. Further, the C48A/C139A double mutant shows only monomeric behaviour. Both dimeric and monomeric hPNMT, as well as mutants have wildtype enzyme activity. These results show that a variety of disulfides, including those observed in the crystal structure, can form in solution. In addition, disulfide-linked dimers are as active as the monomeric enzyme indicating that the crystal structure of the protein is a valid target for inhibitor design.


Assuntos
Dissulfetos/química , Feniletanolamina N-Metiltransferase/química , Feniletanolamina N-Metiltransferase/metabolismo , Sítios de Ligação , Catálise , Cristalização , Cristalografia por Raios X , Cisteína/genética , Dimerização , Ditiotreitol/química , Ditiotreitol/metabolismo , Humanos , Modelos Moleculares , Feniletanolamina N-Metiltransferase/genética , Conformação Proteica , Substâncias Redutoras/química , Substâncias Redutoras/metabolismo , Soluções , Ultracentrifugação/métodos
20.
J Med Chem ; 48(6): 1806-12, 2005 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-15771426

RESUMO

A series of (R)-(+)-3-fluoromethyl-7-(N-substituted aminosulfonyl)-1,2,3,4-tetrahydroisoquinolines has been synthesized and evaluated as inhibitors of PNMT and for their affinity for the alpha(2)-adrenoceptor. Compounds (R)-8 and (R)-9 are remarkably potent and selective inhibitors of PNMT and are predicted to penetrate the blood-brain barrier on the basis of their calculated log P values. Conformational analysis and docking studies were performed in order to examine why the (R)-enantiomer of these 3-fluoromethyl-7-(N-substituted aminosulfonyl)-1,2,3,4-tetrahydroisoquinolines is more potent than the (S)-enantiomer and to determine the likely bound ring conformer of the (R)-enantiomer. It appears that the (R)-enantiomer participates in a water-mediated hydrogen bond in which the (S)-enantiomer cannot. The likely favored ring conformation for (R)-3-fluoromethyl-7-(N-substituted aminosulfonyl)-1,2,3,4-tetrahydroisoquinolines in the PNMT active site is similar to the ring conformation of (R)-5a as determined by gas-phase ab initio calculations.


Assuntos
Encéfalo/metabolismo , Epinefrina/biossíntese , Isoquinolinas/síntese química , Feniletanolamina N-Metiltransferase/antagonistas & inibidores , Feniletanolamina N-Metiltransferase/química , Sulfonamidas/síntese química , Sulfonas/síntese química , Animais , Sítios de Ligação , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Ligação de Hidrogênio , Técnicas In Vitro , Isoquinolinas/química , Isoquinolinas/farmacologia , Masculino , Modelos Moleculares , Conformação Molecular , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/metabolismo , Estereoisomerismo , Relação Estrutura-Atividade , Sulfonamidas/química , Sulfonamidas/farmacologia , Sulfonas/química , Sulfonas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...