Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 297
Filter
1.
Angew Chem Int Ed Engl ; 63(33): e202406542, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-38820076

ABSTRACT

Cytochrome P450 (P450, CYP) 19A1 is the steroid aromatase, the enzyme responsible for the 3-step conversion of androgens (androstenedione or testosterone) to estrogens. The final step is C-C bond scission (removing the 19-oxo group as formic acid) that proceeds via a historically controversial reaction mechanism. The two competing mechanistic possibilities involve a ferric peroxide anion (Fe3+O2 -, Compound 0) and a perferryl oxy species (FeO3+, Compound I). One approach to discern the role of each species in the reaction is with the use of oxygen-18 labeling, i.e., from 18O2 and H2 18O of the reaction product formic acid. We applied this approach, using several technical improvements, to study the deformylation of 19-oxo-androstenedione by human P450 19A1 and of a model secosteroid, 3-oxodecaline-4-ene-10-carboxaldehyde (ODEC), by rabbit P450 2B4. Both aldehyde substrates were sensitive to non-enzymatic acid-catalyzed deformylation, yielding 19-norsteroids, and conditions were established to avoid issues with artifactual generation of formic acid. The Compound 0 reaction pathway predominated (i.e., Fe3+O2 -) in both P450 19A1 oxidation of 19-oxo-androstenedione and P450 2B4 oxidation of ODEC. The P450 19A1 results contrast with our prior conclusions (J. Am. Chem. Soc. 2014, 136, 15016-16025), attributed to several technical modifications.


Subject(s)
Aromatase , Oxidation-Reduction , Aromatase/metabolism , Aromatase/chemistry , Humans , Peroxides/chemistry , Peroxides/metabolism , Animals , Anions/chemistry , Anions/metabolism , Ferric Compounds/chemistry , Ferric Compounds/metabolism , Cytochrome P450 Family 2/metabolism , Cytochrome P450 Family 2/chemistry , Rabbits , Steroids/chemistry , Steroids/metabolism , Androstenedione/chemistry , Androstenedione/metabolism
2.
Molecules ; 28(7)2023 Mar 29.
Article in English | MEDLINE | ID: mdl-37049810

ABSTRACT

Despite the significant outcomes attained by scientific research, breast cancer (BC) still represents the second leading cause of death in women. Estrogen receptor-positive (ER+) BC accounts for the majority of diagnosed BCs, highlighting the disruption of estrogenic signalling as target for first-line treatment. This goal is presently pursued by inhibiting aromatase (AR) enzyme or by modulating Estrogen Receptor (ER) α. An appealing strategy for fighting BC and reducing side effects and resistance issues may lie in the design of multifunctional compounds able to simultaneously target AR and ER. In this paper, previously reported flavonoid-related potent AR inhibitors were suitably modified with the aim of also targeting ERα. As a result, homoisoflavone derivatives 3b and 4a emerged as well-balanced submicromolar dual acting compounds. An extensive computational study was then performed to gain insights into the interactions the best compounds established with the two targets. This study highlighted the feasibility of switching from single-target compounds to balanced dual-acting agents, confirming that a multi-target approach may represent a valid therapeutic option to counteract ER+ BC. The homoisoflavone core emerged as a valuable natural-inspired scaffold for the design of multifunctional compounds.


Subject(s)
Aromatase Inhibitors , Aromatase , Breast Neoplasms , Drug Design , Estrogen Receptor alpha , Flavonoids , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Aromatase Inhibitors/chemical synthesis , Aromatase Inhibitors/chemistry , Aromatase Inhibitors/pharmacology , Flavonoids/chemical synthesis , Flavonoids/chemistry , Flavonoids/pharmacology , Humans , Female , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Molecular Dynamics Simulation , Aromatase/chemistry , Aromatase/metabolism , Thermodynamics , Inhibitory Concentration 50 , Molecular Docking Simulation
3.
Nat Chem Biol ; 16(3): 250-256, 2020 03.
Article in English | MEDLINE | ID: mdl-31932723

ABSTRACT

In plants, lineage-specific metabolites can be created by activities derived from the catalytic promiscuity of ancestral proteins, although examples of recruiting detoxification systems to biosynthetic pathways are scarce. The ubiquitous glyoxalase (GLX) system scavenges the cytotoxic methylglyoxal, in which GLXI isomerizes the α-hydroxy carbonyl in the methylglyoxal-glutathione adduct for subsequent hydrolysis. We show that GLXIs across kingdoms are more promiscuous than recognized previously and can act as aromatases without cofactors. In cotton, a specialized GLXI variant, SPG, has lost its GSH-binding sites and organelle-targeting signal, and evolved to aromatize cyclic sesquiterpenes bearing α-hydroxyketones to synthesize defense compounds in the cytosol. Notably, SPG is able to transform acetylated deoxynivalenol, the prevalent mycotoxin contaminating cereals and foods. We propose that detoxification enzymes are a valuable source of new catalytic functions and SPG, a standalone enzyme catalyzing complex reactions, has potential for toxin degradation, crop engineering and design of novel aromatics.


Subject(s)
Aromatase/metabolism , Lactoylglutathione Lyase/chemistry , Lactoylglutathione Lyase/metabolism , Aromatase/chemistry , Biological Products , Catalysis , Cytosol/metabolism , Glutathione/metabolism , Gossypium/metabolism , Multienzyme Complexes , Pyruvaldehyde/chemistry , Pyruvaldehyde/metabolism
4.
J Biol Chem ; 295(29): 9998-10007, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32493730

ABSTRACT

CYP51 enzymes (sterol 14α-demethylases) are cytochromes P450 that catalyze multistep reactions. The CYP51 reaction occurs in all biological kingdoms and is essential in sterol biosynthesis. It removes the 14α-methyl group from cyclized sterol precursors by first forming an alcohol, then an aldehyde, and finally eliminating formic acid with the introduction of a Δ14-15 double bond in the sterol core. The first two steps are typical hydroxylations, mediated by an electrophilic compound I mechanism. The third step, C-C bond cleavage, has been proposed to involve either compound I (i.e. FeO3+) or, alternatively, a proton transfer-independent nucleophilic ferric peroxo anion (compound 0, i.e. Fe3+O2-). Here, using comparative crystallographic and biochemical analyses of WT human CYP51 (CYP51A1) and its D231A/H314A mutant, whose proton delivery network is destroyed (as evidenced in a 1.98-Å X-ray structure in complex with lanosterol), we demonstrate that deformylation of the 14α-carboxaldehyde intermediate requires an active proton relay network to drive the catalysis. These results indicate a unified, compound I-based mechanism for all three steps of the CYP51 reaction, as previously established for CYP11A1 and CYP19A1. We anticipate that our approach can be applied to mechanistic studies of other P450s that catalyze multistep reactions, such as C-C bond cleavage.


Subject(s)
Protons , Sterol 14-Demethylase/chemistry , Aromatase/chemistry , Catalysis , Cholesterol Side-Chain Cleavage Enzyme/chemistry , Crystallography, X-Ray , Humans
5.
Org Biomol Chem ; 19(15): 3462-3468, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33899877

ABSTRACT

A simple, efficient, and transition metal-free approach to synthesize functionalized 2-(alkynyl)benzonitriles has been developed using suitably functionalized 2H-pyran-2-ones and 4-phenyl/trimethylsilanyl-but-3-yn-2-ones as precursors. The reaction proceeds in the presence of a base at room temperature to yield internal as well as terminal alkynes. The structure of the synthesized compound was confirmed by single-crystal X-ray analysis. The molecular docking study was performed to evaluate the binding mode of action of newly synthesized alkyne derivatives with known human breast cancer target receptor aromatase (PDB ID: 3EQM).


Subject(s)
Aromatase/metabolism , Molecular Docking Simulation , Nitriles/metabolism , Aromatase/chemistry , Breast Neoplasms/enzymology , Female , Humans , Molecular Structure , Nitriles/chemical synthesis , Nitriles/chemistry
6.
Bioorg Chem ; 113: 105017, 2021 08.
Article in English | MEDLINE | ID: mdl-34091288

ABSTRACT

Breast cancer, emerging malignancy is common among women due to overexpression of estrogen. Estrogens are biosynthesized from androgens by aromatase, a cytochrome P450 enzyme complex, and play a pivotal role in stimulating cell proliferation. Therefore, deprivation of estrogen by blocking aromatase is considered as the effective way for the inhibition and treatment of breast cancer. In recent years, various non-steroidal heterocyclic functionalities have been extensively developed and studied for their aromatase inhibition activity. This review provides information about the structural-activity relationship of heterocycles (Type II) towards aromatase. This aids the medicinal chemist around the significance of different heterocyclic moieties and helps to design potent aromatase inhibitors.


Subject(s)
Aromatase Inhibitors/chemistry , Aromatase/metabolism , Heterocyclic Compounds/chemistry , Aromatase/chemistry , Aromatase Inhibitors/metabolism , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Drug Design , Estrogens/metabolism , Female , Heterocyclic Compounds/metabolism , Heterocyclic Compounds/therapeutic use , Humans , Structure-Activity Relationship
7.
Int J Mol Sci ; 22(2)2021 Jan 10.
Article in English | MEDLINE | ID: mdl-33435208

ABSTRACT

Aromatase is the cytochrome P450 enzyme converting androgens into estrogen in the last phase of steroidogenesis. As estrogens are crucial in reproductive biology, aromatase is found in vertebrates and the invertebrates of the genus Branchiostoma, where it carries out the aromatization reaction of the A-ring of androgens that produces estrogens. Here, we investigate the molecular evolution of this unique and highly substrate-selective enzyme by means of structural, sequence alignment, and homology modeling, shedding light on its key role in species conservation. The alignments led to the identification of a core structure that, together with key and unique amino acids located in the active site and the substrate recognition sites, has been well conserved during evolution. Structural analysis shows what their roles are and the reason why they have been preserved. Moreover, the residues involved in the interaction with the redox partner and some phosphorylation sites appeared late during evolution. These data reveal how highly substrate-selective cytochrome P450 has evolved, indicating that the driving forces for evolution have been the optimization of the interaction with the redox partner and the introduction of phosphorylation sites that give the possibility of modulating its activity in a rapid way.


Subject(s)
Aromatase/genetics , Evolution, Molecular , Amino Acid Sequence , Animals , Aromatase/chemistry , Aromatase/metabolism , Catalytic Domain , Estrogens/metabolism , Humans , Models, Molecular , Sequence Alignment , Structure-Activity Relationship , Vertebrates/genetics , Vertebrates/metabolism
8.
Int J Mol Sci ; 22(1)2021 Jan 04.
Article in English | MEDLINE | ID: mdl-33406787

ABSTRACT

Breast cancer is the most common type of cancer. In the developmental stages of breast cancer, estrogens are strongly involved. As estrogen synthesis is regulated by the enzyme aromatase, targeting the activity of this enzyme represents a therapeutic option. The pineal hormone melatonin may exert a suppressive role on aromatase activity, leading to reduced estrogen biosynthesis. A melatonin-mediated decrease in the expression of aromatase promoters and associated genes would provide suitable evidence of this molecule's efficacy as an aromatase inhibitor. Furthermore, melatonin intensifies radiation-induced anti-aromatase effects and counteracts the unwanted disadvantages of chemotherapeutic agents. In this manner, this review summarizes the inhibitory role of melatonin in aromatase action, suggesting its role as a possible oncostatic molecule in breast cancer.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase/chemistry , Breast Neoplasms/drug therapy , Central Nervous System Depressants/pharmacology , Melatonin/pharmacology , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Female , Humans
9.
Molecules ; 26(13)2021 Jul 02.
Article in English | MEDLINE | ID: mdl-34279404

ABSTRACT

Herein, we present the synthesis and crystal structures determination of five 4-(1-phenyl-1H-1,2,3-triazol-4-yl)phenol derivatives containing halogen atoms, 6a-e, which may be used as an excellent mimic of steroids in the drug development process. Good quality crystals obtained for all of the synthesized compounds allowed the analysis of their molecular structures. Subsequently, the determined crystal structures were used to calculate the Hirshfeld surfaces for each of the synthesized compounds. Furthermore, results of our docking studies indicated that synthesized derivatives are able to bind effectively to the active sites of selected enzymes and receptors involved in the hormone biosynthesis and signaling pathways, analogously to the native steroids.


Subject(s)
Aromatase Inhibitors/chemical synthesis , Molecular Docking Simulation , Triazoles/chemical synthesis , Aromatase/chemistry , Aromatase/metabolism , Aromatase Inhibitors/pharmacology , Catalytic Domain , Crystallization , Halogens/chemistry , Phenols/chemistry , Protein Binding , Triazoles/pharmacology
10.
Biochemistry ; 59(33): 2999-3009, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32786398

ABSTRACT

Aromatase (CYP19A1) catalyzes the synthesis of estrogens from androgens and is an invaluable target of pharmacotherapy for estrogen-dependent cancers. CYP19A1 is also one of the most primordial human CYPs and, to the extent that its fundamental dynamics are conserved, is highly relevant to understanding those of the more recently evolved and promiscuous enzymes. A complementary approach employing molecular dynamics simulations and hydrogen-deuterium exchange mass spectrometry (HDX-MS) was employed to interrogate the changes in CYP19A1 dynamics coupled to binding androstenedione (ASD). Gaussian-accelerated molecular dynamics and HDX-MS agree that ASD globally suppresses CYP19A1 dynamics. Bimodal HDX patterns of the B'-C loop potentially arising from at least two conformations are present in free 19A1 only, supporting the possibility that conformational selection is operative. Random-acceleration molecular dynamics and adaptive biasing force simulations illuminate ASD's binding pathway, predicting ASD capture in the lipid headgroups and a pathway to the active site shielded from solvent. Intriguingly, the predicted access channel in 19A1 aligns well with the steroid binding sites of other human sterol-oxidizing CYPs.


Subject(s)
Androstenedione/pharmacokinetics , Aromatase/chemistry , Aromatase/metabolism , Membranes/metabolism , Androstenedione/metabolism , Catalytic Domain , Deuterium Exchange Measurement , Humans , Hydrogen Deuterium Exchange-Mass Spectrometry , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membranes/chemistry , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding
11.
J Recept Signal Transduct Res ; 40(2): 166-172, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32019395

ABSTRACT

In the present study, a new series of α-Aminophosphonates bearing 6-amino-1,3-dimethyluracil was synthesized in good to excellent yields (78-95%) by one-pot, three-component reaction of 6-amino-1,3-dimethyluracil, aromatic aldehydes and diethylphosphite via Kabachnik-Fields reaction by using an eco-friendly Eaton's reagent. All the compounds were screened for in vitro antioxidant studies by 2,2-diphenyl-1-picrylhydrazyl (DPPH) and hydrogen peroxide (H2O2) methods. Among the synthesized bioactive molecules, 4a, 4d, 4g, and 4h exhibited promising antioxidant activity compared with the standard drug Ascorbic acid. Furthermore, in order to support the biological results of the compounds, molecular docking studies were performed against Aromatase enzyme for four compounds which revealed that the compounds 4a, 4d, 4g, and 4h have significant binding modes, with docking scores of -8.6, -8.4, -8.1 and -8.1 respectively and the compound 4b specifically has equal dock score of -8.0 when compared with the standard drug Exemestane.


Subject(s)
Antioxidants/chemistry , Aromatase Inhibitors/chemistry , Organophosphonates/chemistry , Uracil/analogs & derivatives , Antioxidants/chemical synthesis , Aromatase/chemistry , Aromatase Inhibitors/chemical synthesis , Ascorbic Acid/chemistry , Biphenyl Compounds/chemistry , Computer Simulation , Humans , Hydrogen Peroxide/chemistry , In Vitro Techniques , Molecular Docking Simulation , Organophosphonates/chemical synthesis , Picrates/chemistry , Uracil/chemical synthesis , Uracil/chemistry
12.
Chemistry ; 26(28): 6214-6223, 2020 May 15.
Article in English | MEDLINE | ID: mdl-32049373

ABSTRACT

The hydroxylation of nonreactive C-H bonds can be easily catalyzed by a variety of metalloenzymes, especially cytochrome P450s (P450s). The mechanism of P450 mediated hydroxylation has been intensively studied, both experimentally and theoretically. However, understanding the regio- and stereoselectivities of substrates hydroxylated by P450s remains a great challenge. Herein, we use a multi-scale modeling approach to investigate the selectivity of testosterone (TES) and dihydrotestosterone (DHT) hydroxylation catalyzed by two important P450s, CYP3A4 and CYP19A1. For CYP3A4, two distinct binding modes for TES/DHT were predicted by dockings and molecular dynamics simulations, in which the experimentally identified sites of metabolism of TES/DHT can access to the catalytic center. The regio- and stereoselectivities of TES/DHT hydroxylation were further evaluated by quantum mechanical and ONIOM calculations. For CYP19A1, we found that sites 1ß, 2ß and 19 can access the catalytic center, with the intrinsic reactivity 2ß>1ß>19. However, our ONIOM calculations indicate that the hydroxylation is favored at site 19 for both TES and DHT, which is consistent with the experiments and reflects the importance of the catalytic environment in determining the selectivity. Our study unravels the mechanism underlying the selectivity of TES/DHT hydroxylation mediated by CYP3A4 and CYP19A1 and is helpful for understanding the selectivity of other substrates that are hydroxylated by P450s.


Subject(s)
Aromatase/metabolism , Cytochrome P-450 CYP3A/metabolism , Dihydrotestosterone/chemistry , Testosterone/metabolism , Aromatase/chemistry , Catalysis , Cytochrome P-450 CYP3A/chemistry , Humans , Hydroxylation , Kinetics , Oxidation-Reduction , Testosterone/chemistry
13.
Gen Comp Endocrinol ; 291: 113395, 2020 05 15.
Article in English | MEDLINE | ID: mdl-31981691

ABSTRACT

Duplicated cyp19a1 genes (cyp19a1a encoding aromatase a and cyp19a1b encoding aromatase b) have been identified in an increasing number of teleost species. Cyp19a1a is mainly expressed in the gonads, while cyp19a1b is mainly expressed in the brain, specifically in radial glial cells, as largely investigated by Kah and collaborators. The third round of whole-genome duplication that specifically occurred in the teleost lineage (TWGD or 3R) is likely at the origin of the duplicated cyp19a1 paralogs. In contrast to the situation in other teleosts, our previous studies identified a single cyp19a1 in eels (Anguilla), which are representative species of a basal group of teleosts, Elopomorpha. In the present study, using genome data mining and phylogenetic and synteny analyses, we confirmed that the whole aromatase genomic region was duplicated in eels, with most aromatase-neighboring genes being conserved in duplicate in eels, as in other teleosts. These findings suggest that specific gene loss of one of the 3R-duplicated cyp19a1 paralogs occurred in Elopomorpha after TWGD. Similarly, a single cyp19a1 gene was found in the arowana, which is a representative species of another basal group of teleosts, Osteoglossomorpha. In eels, the single cyp19a1 is expressed in both the brain and the gonads, as observed for the single CYP19A1 gene present in other vertebrates. The results of phylogenetic, synteny, closest neighboring gene, and promoter structure analyses showed that the single cyp19a1 of the basal teleosts shared conserved properties with both teleost cyp19a1a and cyp19a1b paralogs, which did not allow us to conclude which of the 3R-duplicated paralogs (cyp19a1a or cyp19a1b) was lost in Elopomorpha. Elopomorpha and Osteoglossomorpha cyp19a1 genes exhibited preserved ancestral functions, including expression in both the gonad and brain. We propose that the subfunctionalization of the 3R-duplicated cyp19a1 paralogs expressed specifically in the gonad or brain occurred in Clupeocephala, after the split of Clupeocephala from Elopomorpha and Osteoglossomorpha, which represented a driving force for the conservation of both 3R-duplicated paralogs in all extant Clupeocephala. In contrast, the functional redundancy of the undifferentiated 3R-duplicated cyp19a1 paralogs in elopomorphs and osteoglossomorphs would have favored the loss of one 3R paralog in basal teleosts.


Subject(s)
Aromatase/genetics , Evolution, Molecular , Fishes/genetics , Gene Duplication , Anguilla/genetics , Animals , Aromatase/chemistry , Aromatase/metabolism , Base Sequence , Biological Evolution , Conserved Sequence , Genome , Phylogeny , Promoter Regions, Genetic/genetics , Protein Domains , Synteny/genetics
14.
Biochem J ; 476(21): 3313-3331, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31652308

ABSTRACT

Aromatase CYP19A1 catalyzes the synthesis of estrogens in endocrine, reproductive and central nervous systems. Higher levels of 17ß-estradiol (E2) are associated with malignancies and diseases of the breast, ovary and endometrium, while low E2 levels increase the risk for osteoporosis, cardiovascular diseases and cognitive disorders. E2, the transcriptional activator of the estrogen receptors, is also known to be involved in non-genomic signaling as a neurotransmitter/neuromodulator, with recent evidence for rapid estrogen synthesis (RES) within the synaptic terminal. Although regulation of brain aromatase activity by phosphorylation/dephosphorylation has been suggested, it remains obscure in the endocrine and reproductive systems. RES and overabundance of estrogens could stimulate the genomic and non-genomic signaling pathways, and genotoxic effects of estrogen metabolites. Here, by utilizing biochemical, cellular, mass spectrometric, and structural data we unequivocally demonstrate phosphorylation of human placental aromatase and regulation of its activity. We report that human aromatase has multiple phosphorylation sites, some of which are consistently detectable. Phosphorylation of the residue Y361 at the reductase-coupling interface significantly elevates aromatase activity. Other sites include the active site residue S478 and several at the membrane interface. We present the evidence that two histidine residues are phosphorylated. Furthermore, oxidation of two proline residues near the active site may have implications in regulation. Taken together, the results demonstrate that aromatase activity is regulated by phosphorylation and possibly other post-translational modifications. Protein level regulation of aromatase activity not only represents a paradigm shift in estrogen-mediated biology, it could also explain unresolved clinical questions such as aromatase inhibitor resistance.


Subject(s)
Aromatase/metabolism , Placenta/enzymology , Amino Acid Motifs , Aromatase/chemistry , Aromatase/genetics , Estrogens/metabolism , Female , Humans , Phosphorylation , Placenta/metabolism , Pregnancy
15.
Molecules ; 25(3)2020 Feb 08.
Article in English | MEDLINE | ID: mdl-32046297

ABSTRACT

Aromatase is an enzyme member of the cytochrome P450 superfamily coded by the CYP19A1 gene. Its main action is the conversion of androgens into estrogens, transforming androstenedione into estrone and testosterone into estradiol. This enzyme is present in several tissues and it has a key role in the maintenance of the balance of androgens and estrogens, and therefore in the regulation of the endocrine system. With regard to chemical safety and human health, azoles, which are used as agrochemicals and pharmaceuticals, are potential endocrine disruptors due to their agonist or antagonist interactions with the human aromatase enzyme. This theoretical study investigated the active agonist and antagonist properties of "chemical classes of azoles" to determine the relationships of azole interaction with CYP19A1, using stereochemical and electronic properties of the molecules through classification and multilinear regression (MLR) modeling. The antagonist activities for the same substituent on diazoles and triazoles vary with its chemical composition and its position and both heterocyclic systems require aromatic substituents. The triazoles require the spherical shape and diazoles have to be in proper proportion of the branching index and the number of ring systems for the inhibition. Considering the electronic aspects, triazole antagonist activity depends on the electrophilicity index that originates from interelectronic exchange interaction (ωHF) and the LUMO energy ( E LUMO PM 7 ), and the diazole antagonist activity originates from the penultimate orbital ( E HOMONL PM 7 ) of diazoles. The regression models for agonist activity show that it is opposed by the static charges but favored by the delocalized charges on the diazoles and thiazoles. This study proposes that the electron penetration of azoles toward heme group decides the binding behavior and stereochemistry requirement for antagonist activity against CYP19A1 enzyme.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase/chemistry , Azoles/pharmacology , Cytochrome P-450 Enzyme Inducers/pharmacology , Electrons , Endocrine Disruptors/pharmacology , Models, Statistical , Aromatase/metabolism , Aromatase Inhibitors/chemistry , Azoles/chemistry , Cytochrome P-450 Enzyme Inducers/chemistry , Endocrine Disruptors/chemistry , Heme/chemistry , Heme/metabolism , Humans , Models, Chemical , Protein Binding , Quantum Theory , Static Electricity , Stereoisomerism , Structure-Activity Relationship , Thermodynamics
16.
Biophys J ; 116(3): 419-432, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30658838

ABSTRACT

Human cytochrome P450 (CYP) enzymes play an important role in the metabolism of drugs, steroids, fatty acids, and xenobiotics. Microsomal CYPs are anchored in the endoplasmic reticulum membrane by an N-terminal transmembrane (TM) helix that is connected to the globular catalytic domain by a flexible linker sequence. However, the structural and functional importance of the TM-helix is unclear because it has been shown that CYPs can still associate with the membrane and have enzymatic activity in reconstituted systems after truncation or modification of the N-terminal sequence. Here, we investigated the effect of mutations in the N-terminal TM-helix residues of two human steroidogenic enzymes, CYP 17A1 and CYP 19A1, that are major drug targets for cancer therapy. These mutations were originally introduced to increase the expression of the proteins in Escherichia coli. To investigate the effect of the mutations on protein-membrane interactions and function, we carried out coarse-grained and all-atom molecular dynamics simulations of the CYPs in a phospholipid bilayer. We confirmed the orientations of the globular domain in the membrane observed in the simulations by linear dichroism measurements in a Nanodisc. Whereas the behavior of CYP 19A1 was rather insensitive to truncation of the TM-helix, mutations in the TM-helix of CYP 17A1, especially W2A and E3L, led to a gradual drifting of the TM-helix out of the hydrophobic core of the membrane. This instability of the TM-helix could affect interactions with the allosteric redox partner, cytochrome b5, required for CYP 17A1's lyase activity. Furthermore, the simulations showed that the mutant TM-helix influenced the membrane interactions of the CYP 17A1 globular domain. In some simulations, the mutated TM-helix obstructed the substrate access tunnel from the membrane to the CYP active site, indicating a possible effect on enzyme function.


Subject(s)
Aromatase/chemistry , Aromatase/metabolism , Cell Membrane/metabolism , Mutation , Steroid 17-alpha-Hydroxylase/chemistry , Steroid 17-alpha-Hydroxylase/metabolism , Amino Acid Sequence , Molecular Dynamics Simulation , Protein Binding , Protein Conformation, alpha-Helical , Protein Domains , Steroid 17-alpha-Hydroxylase/genetics
17.
Mol Reprod Dev ; 86(5): 480-490, 2019 05.
Article in English | MEDLINE | ID: mdl-30779247

ABSTRACT

Cytochrome P450 aromatase (CYP19) catalyzes the conversion of androgens to estrogens and is critical in sex differentiation. CYP19 exists as the ovarian type and brain type. Herein, we cloned the full-length ovarian cyp19a gene from the Chinese soft-shelled turtle, Pelodiscus sinensis (pscyp19a). We determined the distribution of pscyp19a in adult tissue and evaluated its expression during embryonic development, following treatment with 17ß-estradiol (E2) or letrozole (LE). The pscyp19a complementary DNA is 2,285 bp in length and comprises a 1,512 bp open reading frame that encodes a protein of 503 AA. The nucleotide sequence and amino acid of pscyp19a shared significant identity with other vertebrate sequences. Expression of pscyp19a was high in the ovary (p < 0.01), and exhibited modest expression in the female brain and intestine. Expression of pscyp19a displayed significant differences between sexes during early embryo development stages; expression increased gradually during embryonic development in females, but the opposite trend was observed in males. Female embryos treated with different concentrations of E2 and LE displayed altered pscyp19a expression compared with untreated individuals, and E2 clearly induced pscyp19a expression. These results indicate that pscyp19a gene plays important roles in early developmental stages in Chinese soft-shelled turtle, and may assist future studies on sex differentiation and sex control in this and similar species.


Subject(s)
Aromatase , Estradiol/pharmacology , Gene Expression/drug effects , Letrozole/pharmacology , Turtles/genetics , Animals , Aromatase/analysis , Aromatase/chemistry , Aromatase/genetics , Aromatase/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Female , Male , Tissue Distribution , Turtles/embryology , Turtles/metabolism
18.
J Chem Inf Model ; 59(6): 2930-2940, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31033287

ABSTRACT

Phosphorylation by kinases enzymes is a widespread regulatory mechanism able of rapidly altering the function of target proteins. Among these are cytochrome P450s (CYP450), a superfamily of enzymes performing the oxidation of endogenous and exogenous substrates thanks to the electron supply of a redox partner. In spite of its pivotal role, the molecular mechanism by which phosphorylation modulates CYP450s metabolism remains elusive. Here by performing microsecond-long all-atom molecular dynamics simulations, we disclose how phosphorylation regulates estrogen biosynthesis, catalyzed by the Human Aromatase (HA) enzyme. Namely, we unprecedentedly propose that HA phosphorylation at Y361 markedly stabilizes its adduct with the flavin mononucleotide domain of CYP450s reductase (CPR), the redox partner of microsomal CYP450s, and a variety of other proteins. With CPR present at physiological conditions in a limiting ratio with respect to its multiple oxidative partners, the enhanced stability of the CPR/HA adduct may favor HA in the competition with the other proteins requiring CPR's electron supply, ultimately facilitating the electron transfer and estrogen biosynthesis. As a result, our work elucidates at atomic-level the post-translational regulation of CYP450s catalysis. Given the potential for rational clinical management of diseases associated with steroid metabolism disorders, unraveling this mechanism is of utmost importance, and raises the intriguing perspective of exploiting this knowledge to devise novel therapies.


Subject(s)
Aromatase/chemistry , Aromatase/metabolism , Molecular Dynamics Simulation , Protein Processing, Post-Translational , Binding Sites , Flavin Mononucleotide/metabolism , Flavin-Adenine Dinucleotide/metabolism , NADP/metabolism , Phosphorylation , Protein Conformation , Quantum Theory
19.
Bioorg Med Chem ; 27(19): 115040, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31416738

ABSTRACT

A library of bis-sulfonamides (9-26) were synthesized and tested for their aromatase inhibitory activities. Interestingly, all bis-sulfonamide derivatives inhibited the aromatase with IC50 range of 0.05-11.6 µM except for compound 23. The analogs 15 and 16 bearing hydrophobic chloro and bromo groups exhibited the potent aromatase inhibitory activity in sub-micromolar IC50 values (i.e., 50 and 60 nM, respectively) with high safety index. Molecular docking revealed that the chloro and bromo benzenesulfonamides (15 and 16) may play role in the hydrophobic interaction with Leu477 of the aromatase to mimic steroidal backbone of the natural substrate, androstenedione. QSAR study also revealed that the most potent activity of compounds was governed by van der Waals volume (GATS6v) and mass (Mor03m) descriptors. Finally, the two compounds (15 and 16) were highlighted as promising compounds to be further developed as novel aromatase inhibitors.


Subject(s)
Aromatase Inhibitors/pharmacology , Sulfonamides/pharmacology , Aromatase/chemistry , Aromatase/metabolism , Aromatase Inhibitors/chemical synthesis , Aromatase Inhibitors/metabolism , Binding Sites , Cell Line, Tumor , Humans , Molecular Docking Simulation , Molecular Structure , Protein Binding , Quantitative Structure-Activity Relationship , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Sulfonamides/chemical synthesis , Sulfonamides/metabolism
20.
Mol Divers ; 23(3): 723-738, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30560342

ABSTRACT

A series of new urea/thiourea derivatives 3a-j were synthesized by simple addition reaction of functionalized phenyl isocyanates/isothiocyanates 2a-j with N-(5-amino-2-methylphenyl)-4-(3-pyridyl)-2-pyrimidinamine (imatinib intermediate) (1) in the presence of 1,4-dimethyl piperazine (DMPZ) as a base, and another series of new sulfonamide/carbamate derivatives 5a-k were synthesized by reacting 1 with various substituted aromatic sulfonyl chlorides 4a-f and aromatic/aliphatic chloroformates 4g-k in the presence of DMPZ as a base. The title compounds 3a-j and 5a-k were characterized by IR, 1H, 13C NMR and mass spectral data. Antimicrobial, antioxidant and in silico molecular docking studies were made against aromatase.


Subject(s)
Carbamates/chemistry , Imatinib Mesylate/chemical synthesis , Imatinib Mesylate/pharmacology , Molecular Docking Simulation , Sulfonamides/chemistry , Thiourea/chemistry , Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/chemistry , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Antioxidants/chemical synthesis , Antioxidants/chemistry , Antioxidants/metabolism , Antioxidants/pharmacology , Aromatase/chemistry , Aromatase/metabolism , Chemistry Techniques, Synthetic , Imatinib Mesylate/chemistry , Imatinib Mesylate/metabolism , Protein Conformation , Spectrum Analysis , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL