Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
ChemMedChem ; 18(23): e202300457, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37872124

ABSTRACT

(R)-PFI-2 is a histone substrate-competitive inhibitor of the human histone lysine monomethyltransferase SETD7. Aimed at developing potent inhibitors of SETD7 that can also act as small molecule substrates, we replaced the pyrrolidine ring of (R)-PFI-2 with several side chains bearing nucleophilic functional groups. We explored the inhibitory activity of 20 novel (R)-PFI-2 analogues, and found that the most potent analogue has a hydroxyethyl side chain (7). SETD7's ability to catalyse methylation of (R)-PFI-2-based small molecules was evaluated by mass spectrometric assays, and we observed efficient methylation of analogues bearing lysine mimicking nucleophilic amines. The optimal side chain was found to be an aminoethyl group (1), which was surprisingly also dimethylated by SETD7. The work demonstrates that small molecules can act as both substrates and inhibitors of biomedically important SETD7.


Subject(s)
Histone-Lysine N-Methyltransferase , Histones , Humans , Lysine , Pyrrolidines/pharmacology , Pyrrolidines/chemistry
2.
Sci Rep ; 10(1): 3671, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111884

ABSTRACT

Methylation of lysine residues in histone proteins is catalyzed by S-adenosylmethionine (SAM)-dependent histone lysine methyltransferases (KMTs), a genuinely important class of epigenetic enzymes of biomedical interest. Here we report synthetic, mass spectrometric, NMR spectroscopic and quantum mechanical/molecular mechanical (QM/MM) molecular dynamics studies on KMT-catalyzed methylation of histone peptides that contain lysine and its sterically demanding analogs. Our synergistic experimental and computational work demonstrates that human KMTs have a capacity to catalyze methylation of slightly bulkier lysine analogs, but lack the activity for analogs that possess larger aromatic side chains. Overall, this study provides an important chemical insight into molecular requirements that contribute to efficient KMT catalysis and expands the substrate scope of KMT-catalyzed methylation reactions.


Subject(s)
Histone-Lysine N-Methyltransferase/chemistry , Lysine/chemistry , Catalysis , Catalytic Domain , Humans
3.
Chembiochem ; 21(3): 392-400, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31287209

ABSTRACT

Biomedicinally important histone lysine methyltransferases (KMTs) catalyze the transfer of a methyl group from S-adenosylmethionine (AdoMet) cosubstrate to lysine residues in histones and other proteins. Herein, experimental and computational investigations on human KMT-catalyzed ethylation of histone peptides by using S-adenosylethionine (AdoEth) and Se-adenosylselenoethionine (AdoSeEth) cosubstrates are reported. MALDI-TOF MS experiments reveal that, unlike monomethyltransferases SETD7 and SETD8, methyltransferases G9a and G9a-like protein (GLP) do have the capacity to ethylate lysine residues in histone peptides, and that cosubstrates follow the efficiency trend AdoMet>AdoSeEth>AdoEth. G9a and GLP can also catalyze AdoSeEth-mediated ethylation of ornithine and produce histone peptides bearing lysine residues with different alkyl groups, such as H3K9meet and H3K9me2et. Molecular dynamics and free energy simulations based on quantum mechanics/molecular mechanics potential supported the experimental findings by providing an insight into the geometry and energetics of the enzymatic methyl/ethyl transfer process.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Lysine/metabolism , Biocatalysis , Density Functional Theory , Histone-Lysine N-Methyltransferase/chemistry , Humans , Lysine/chemistry , Molecular Conformation , Molecular Dynamics Simulation
4.
Bioorg Med Chem Lett ; 29(17): 2516-2524, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31350126

ABSTRACT

Detailed structure activity relationship of two series of quinazoline EHMT1/EHMT2 inhibitors (UNC0224 and UNC0638) have been elaborated. New and active alternatives are presented for the ubiquitous substitution patterns found in literature for the linker to the lysine mimicking region and the lysine mimic itself. These findings could allow for advancing EHMT1/EHMT2 inhibitors of that type beyond tool compounds by fine-tuning physicochemical properties making these inhibitors more drug-like. .


Subject(s)
Enzyme Inhibitors/chemistry , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Binding Sites , Cell Line, Tumor , Drug Design , Enzyme Inhibitors/metabolism , Histocompatibility Antigens/genetics , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Inhibitory Concentration 50 , Lysine/chemistry , Molecular Docking Simulation , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Point Mutation , Quinazolines/chemistry , Quinazolines/metabolism , Structure-Activity Relationship
5.
J Org Chem ; 84(10): 6536-6545, 2019 May 17.
Article in English | MEDLINE | ID: mdl-31050295

ABSTRACT

We report an efficient catalytic Staudinger reduction at room temperature that enables the preparation of a structurally diverse set of amines from azides in excellent yields. The reaction is based on the use of catalytic amounts of triphenylphosphine as a phosphine source and diphenyldisiloxane as a reducing agent. Our catalytic Staudinger reduction exhibits a high chemoselectivity, as exemplified by reduction of azides over other common functionalities, including nitriles, alkenes, alkynes, esters, and ketones.

6.
Org Biomol Chem ; 17(23): 5693-5697, 2019 06 12.
Article in English | MEDLINE | ID: mdl-31134245

ABSTRACT

Histone lysine methyltransferases (KMTs) are biomedicinally important class of epigenetic enzymes that catalyse methylation of lysine residues in histones and other proteins. Enzymatic and computational studies on the simplest lysine analogues that possess a modified main chain demonstrate that the lysine's backbone contributes significantly to functional KMT binding and catalysis.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Histones/chemistry , Histones/metabolism , Lysine/chemistry , Lysine/metabolism , Histone-Lysine N-Methyltransferase/genetics , Models, Molecular , Molecular Structure , Thermodynamics
7.
Biochem J ; 476(7): 1109-1119, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30898847

ABSTRACT

The biologically important carnitine biosynthesis pathway in humans proceeds via four enzymatic steps. The first step in carnitine biosynthesis is catalyzed by trimethyllysine hydroxylase (TMLH), a non-heme Fe(II) and 2-oxoglutarate (2OG)-dependent oxygenase, which catalyzes the stereospecific hydroxylation of (2S)-Nε-trimethyllysine to (2S,3S)-3-hydroxy-Nε-trimethyllysine. Here, we report biocatalytic studies on human TMLH and its 19 variants introduced through site-directed mutagenesis. Amino acid substitutions at the sites involved in binding of the Fe(II) cofactor, 2OG cosubstrate and (2S)-Nε-trimethyllysine substrate provide a basic insight into the binding requirements that determine an efficient TMLH-catalyzed conversion of (2S)-Nε-trimethyllysine to (2S,3S)-3-hydroxy-Nε-trimethyllysine. This work demonstrates the importance of the recognition sites that contribute to the enzymatic activity of TMLH: the Fe(II)-binding H242-D244-H389 residues, R391-R398 involved in 2OG binding and several residues (D231, N334 and the aromatic cage comprised of W221, Y217 and Y234) associated with binding of (2S)-Nε-trimethyllysine.


Subject(s)
Mixed Function Oxygenases/chemistry , Amino Acid Sequence , Amino Acid Substitution , Biocatalysis , Carnitine/biosynthesis , Catalytic Domain/genetics , Humans , Kinetics , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , gamma-Butyrobetaine Dioxygenase/chemistry , gamma-Butyrobetaine Dioxygenase/genetics , gamma-Butyrobetaine Dioxygenase/metabolism
8.
ChemMedChem ; 13(14): 1405-1413, 2018 07 18.
Article in English | MEDLINE | ID: mdl-29869845

ABSTRACT

SETD7 is a histone H3K4 lysine methyltransferase involved in human gene regulation. Aberrant expression of SETD7 has been associated with various diseases, including cancer. Therefore, SETD7 is considered a good target for the development of new epigenetic drugs. To date, few selective small-molecule inhibitors have been reported that target SETD7, the most potent being (R)-PFI-2. Herein we report structure-activity relationship studies on (R)-PFI-2 and its analogues. A library of 29 structural analogues of (R)-PFI-2 was synthesized and evaluated for inhibition of recombinantly expressed human SETD7. The key interactions were found to be a salt bridge and a hydrogen bond formed between (R)-PFI-2's NH2+ group and SETD7's Asp256 and His252 residue, respectively.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Tetrahydroisoquinolines/chemistry , Tetrahydroisoquinolines/pharmacology , Enzyme Inhibitors/chemical synthesis , Epigenesis, Genetic/drug effects , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Humans , Molecular Docking Simulation , Pyrrolidines/chemical synthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Tetrahydroisoquinolines/chemical synthesis
9.
Bioorg Med Chem Lett ; 28(7): 1234-1238, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29519735

ABSTRACT

Histone lysine methyltransferases G9a and GLP are validated targets for the development of new epigenetic drugs. Most, if not all, inhibitors of G9a and GLP target the histone substrate binding site or/and the S-adenosylmethionine cosubstrate binding site. Here, we report an alternative approach for inhibiting the methyltransferase activity of G9a and GLP. For proper folding and enzymatic activity, G9a and GLP contain structural zinc fingers, one of them being adjacent to the S-adenosylmethionine binding site. Our work demonstrates that targeting these labile zinc fingers with electrophilic small molecules results in ejection of structural zinc ions, and consequently inhibition of the methyltransferase activity. Very effective Zn(II) ejection and inhibition of G9a and GLP was observed with clinically used ebselen, disulfiram and cisplatin.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Zinc Fingers/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Histocompatibility Antigens/chemistry , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Humans , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
10.
Org Biomol Chem ; 15(30): 6426-6432, 2017 Aug 02.
Article in English | MEDLINE | ID: mdl-28737181

ABSTRACT

Development of catalytic amide bond formation reactions has been the subject of the intensive investigations in the past decade. Herein we report an efficient organophosphorus-catalysed amidation reaction between unactivated carboxylic acids and amines. Poly(methylhydrosiloxane), a waste product of the silicon industry, is used as an inexpensive and green reducing agent for in situ reduction of phosphine oxide to phosphine. The reported method enables the synthesis of a wide range of secondary and tertiary amides in very good to excellent yields.

11.
Org Lett ; 18(9): 2086-9, 2016 05 06.
Article in English | MEDLINE | ID: mdl-27082825

ABSTRACT

A one-pot process for the synthesis of unsymmetrical sulfoxides using organometallic nucleophiles is described. Sulfur dioxide, delivered from the surrogate DABSO (DABCO-bis(sulfur dioxide)), acts as the initial electrophile and combines with the first organometallic reagent to generate a sulfinate intermediate. In situ conversion of the sulfinate to a sulfinate silyl ester, using TMS-Cl (trimethylsilyl chloride), generates a second electrophile, allowing addition of a second organometallic reagent. Organolithium or Grignard reagents can be employed, delivering sulfoxides in good to excellent yields.

12.
Chem Commun (Camb) ; 50(43): 5763-6, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24752820

ABSTRACT

Unactivated carboxylic acids and amines undergo organocatalytic Ph3P/CCl4-mediated amide bond formation by employing in situ reduction of triphenylphosphine oxide to triphenylphosphine in the presence of diethoxymethylsilane and bis(4-nitrophenyl)phosphate.


Subject(s)
Amides/chemistry , Amines/chemistry , Carboxylic Acids/chemistry , Organophosphorus Compounds/chemistry , Catalysis
SELECTION OF CITATIONS
SEARCH DETAIL
...