Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem ; 68: 116875, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35716588

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) is an important biomolecule with essential roles at the intersection of energy metabolism, epigenetic regulation and cell signalling. Synthetic analogues of NAD+ are therefore of great interest as chemical tools for medicinal chemistry, chemical biology and drug discovery. Herein, we report the chemical synthesis and full analytical characterisation of three stereoisomers of 2″-amino NAD+, and their biochemical evaluation against two classes of NAD+-consuming enzymes: the human sirtuins 1-3, and the bacterial toxin TccC3. To rationalise the observed activities, molecular docking experiments were carried out with SIRT1 and SIRT2, which identified the correct orientation of the pyrophosphate linkage as a major determinant for activity in this series. These results, together with results from stability tests and a conformational analysis, allow, for the first time, a side-by-side comparison of the chemical and biochemical features, and analytical properties, of different 2″-amino NAD+ stereoisomers. Our findings provide insight into the recognition of co-substrate analogues by sirtuins, and will greatly facilitate the application of these important NAD+ analogues as chemical tool compounds for mechanistic studies with these as well as other NAD+-dependent enyzmes.


Subject(s)
Sirtuins , Adenosine Diphosphate , Epigenesis, Genetic , Humans , Molecular Docking Simulation , NAD/metabolism , Sirtuin 2/metabolism , Sirtuins/metabolism , Stereoisomerism , Transferases/metabolism
2.
J Med Chem ; 62(5): 2582-2597, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30785747

ABSTRACT

Lysine acetyltransferases (KATs, also termed histone acetyltransferases, HATs) catalyze the acetylation of substrate lysine residues by employing the cofactor acetyl-coenzyme A (AcCoA), thereby providing a dynamic control mechanism of protein function. Because of their major involvement in cell development and homeostasis, small-molecule modulators of KAT activity are urgently needed to assess their therapeutic potential and for probing their underlying biology. Recent advances in the field suggest that targeting the cofactor binding site represents a promising strategy for identifying potent and selective ligands. Here, we present the synthesis of two functional cofactor-based chemical probes and their usage as mechanistic tools in a broadly applicable assay platform. A fluorescence polarization (FP)-based binding assay was combined with biolayer interferometry competition analysis and a FP competition activity immunoassay to enable easy, reliable, and profound evaluation of ligands that target the KAT cofactor binding site.


Subject(s)
Acetyl Coenzyme A/metabolism , Lysine Acetyltransferases/metabolism , Molecular Probes/metabolism , Catalytic Domain , Fluorescence Polarization Immunoassay , Ligands , Protein Binding
3.
Nat Genet ; 50(10): 1442-1451, 2018 10.
Article in English | MEDLINE | ID: mdl-30224647

ABSTRACT

The etiological spectrum of ultra-rare developmental disorders remains to be fully defined. Chromatin regulatory mechanisms maintain cellular identity and function, where misregulation may lead to developmental defects. Here, we report pathogenic variations in MSL3, which encodes a member of the chromatin-associated male-specific lethal (MSL) complex responsible for bulk histone H4 lysine 16 acetylation (H4K16ac) in flies and mammals. These variants cause an X-linked syndrome affecting both sexes. Clinical features of the syndrome include global developmental delay, progressive gait disturbance, and recognizable facial dysmorphism. MSL3 mutations affect MSL complex assembly and activity, accompanied by a pronounced loss of H4K16ac levels in vivo. Patient-derived cells display global transcriptome alterations of pathways involved in morphogenesis and cell migration. Finally, we use histone deacetylase inhibitors to rebalance acetylation levels, alleviating some of the molecular and cellular phenotypes of patient cells. Taken together, we characterize a syndrome that allowed us to decipher the developmental importance of MSL3 in humans.


Subject(s)
Genetic Diseases, X-Linked/genetics , Histones/metabolism , Mutation , Neurodevelopmental Disorders/genetics , Transcription Factors/genetics , Acetylation , Adolescent , Animals , Case-Control Studies , Cells, Cultured , Child , Child, Preschool , Chromosomal Proteins, Non-Histone , Cohort Studies , DNA-Binding Proteins , Female , Genes, X-Linked , HEK293 Cells , Histone Acetyltransferases/metabolism , Humans , Infant , Male , Mice , Mice, Transgenic , Neurodevelopmental Disorders/metabolism , Protein Processing, Post-Translational/genetics , Syndrome
4.
Chem Rec ; 18(12): 1701-1707, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29931800

ABSTRACT

Reversible lysine deacetylation is exerted by both zinc and NAD+ -dependent deacetylases. It is an important factor in epigenetic regulation and more generally in the posttranslational regulation of protein stability, association and activity. Some of these enzymes can also cleave off fatty acids or dicarboxylic acids from lysines in proteins. The NAD+ -dependent deacetylases are termed Sirtuins and are implicated in the pathogenesis of different diseases. For the isotype Sirt2 highly selective inhibitors have been identified in the last few years. Many of those Sirt2 selective compounds, like the Sirtuin rearranging ligands (SirReals) discovered in our group, have been shown or are postulated to bind to the so-called selectivity pocket. This binding site is not observed in crystal structures of the apo-enzyme but can be opened up by long chain fatty acid substrates respectively suitable inhibitors. Recently, this unique feature of Sirt2 was exploited to provide highly potent and selective tools for the chemical biology of Sirtuins. Here, we shortly review Sirtuin biology, present inhibitors that have either been confirmed or postulated to bind to the selectivity pocket, their applications and an outlook regarding mechanistic investigations.


Subject(s)
Sirtuin 2/chemistry , Sirtuin 2/metabolism , Binding Sites , Epigenomics , Humans , Ligands , Lysine/chemistry , Lysine/metabolism , Molecular Dynamics Simulation , Protein Structure, Tertiary , Pyrimidines/chemistry , Pyrimidines/metabolism , Sirtuin 2/antagonists & inhibitors , Substrate Specificity
5.
Article in English | MEDLINE | ID: mdl-29685963

ABSTRACT

Sirtuins are NAD+-dependent protein deacylases capable of cleaving off acetyl as well as other acyl groups from the ɛ-amino group of lysines in histones and other substrate proteins. They have been reported as promising drug targets, and thus modulators of their activity are needed as molecular tools to uncover their biological function and as potential therapeutics. Here, we present new assay formats that complement existing assays for sirtuin biochemistry and cellular target engagement. Firstly, we report the development of a homogeneous fluorescence-based activity assay using unlabelled acylated peptides. Upon deacylation, the free lysine residue reacts with fluorescamine to form a fluorophore. Secondly, using click chemistry with a TAMRA-azide on a propargylated sirtuin inhibitor, we prepared the first fluorescently labelled small-molecule inhibitor of Sirt2. This is used in a binding assay, which is based on fluorescence polarization. We used it successfully to map potential inhibitor-binding sites and also to show cellular Sirt2 engagement. By means of these new assays, we were able to identify and characterize novel Sirt2 inhibitors out of a focused library screen. The binding of the identified Sirt2 inhibitors was rationalized by molecular docking studies. These new chemical tools thus can enhance further sirtuin research.This article is part of a discussion meeting issue 'Frontiers in epigenetic chemical biology'.


Subject(s)
Molecular Docking Simulation , NAD/chemistry , Sirtuin 2/chemistry , Fluorescence , Humans
6.
Med Res Rev ; 38(1): 147-200, 2018 01.
Article in English | MEDLINE | ID: mdl-28094444

ABSTRACT

Sirtuins are NAD+ -dependent protein deacylases that cleave off acetyl, as well as other acyl groups, from the ε-amino group of lysines in histones and other substrate proteins. Seven sirtuin isotypes (Sirt1-7) have been identified in mammalian cells. As sirtuins are involved in the regulation of various physiological processes such as cell survival, cell cycle progression, apoptosis, DNA repair, cell metabolism, and caloric restriction, a dysregulation of their enzymatic activity has been associated with the pathogenesis of neoplastic, metabolic, infectious, and neurodegenerative diseases. Thus, sirtuins are promising targets for pharmaceutical intervention. Growing interest in a modulation of sirtuin activity has prompted the discovery of several small molecules, able to inhibit or activate certain sirtuin isotypes. Herein, we give an update to our previous review on the topic in this journal (Schemies, 2010), focusing on recent developments in sirtuin biology, sirtuin modulators, and their potential as novel therapeutic agents.


Subject(s)
Group III Histone Deacetylases/antagonists & inhibitors , Group III Histone Deacetylases/metabolism , Histone Deacetylase Inhibitors/pharmacology , Animals , Humans , Molecular Targeted Therapy
7.
Future Med Chem ; 8(13): 1537-51, 2016 09.
Article in English | MEDLINE | ID: mdl-27572962

ABSTRACT

AIM: The histone kinase PRK1 has been identified as a potential target to combat prostate cancer but selective PRK1 inhibitors are lacking. The US FDA -approved JAK1-3 inhibitor tofacitinib also potently inhibits PRK1 in vitro. RESULTS: We show that tofacitinib also inhibits PRK1 in a cellular setting. Using tofacitinib as a starting point for structure-activity relationship studies, we identified a more potent and another more selective PRK1 inhibitor compared with tofacitinib. Furthermore, we found two potential PRK1/JAK3-selectivity hotspots. CONCLUSION: The identified inhibitors and the selectivity hotspots lay the basis for the development of selective PRK1 inhibitors. The identification of PRK1, but also of other cellular tofacitinib targets, has implications on its clinical use and on future development of tofacitinib-like JAK inhibitors. [Formula: see text].


Subject(s)
Piperidines/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Piperidines/chemical synthesis , Piperidines/chemistry , Protein Kinase C/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Structure-Activity Relationship
8.
ChemMedChem ; 11(18): 2084-94, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27472906

ABSTRACT

Despite the considerable interest in protein kinase C-related kinase 1 (PRK1) as a target in cancer research, there is still a lack of PRK1 inhibitors with suitable selectivity profiles and physicochemical properties. To identify new PRK1 inhibitors we applied a virtual screening approach, which combines ensemble docking, minimization of the protein-ligand complex, binding free energy calculations, and application of quantitative structure-activity relationship (QSAR) models for predicting in vitro activity. The developed approach was then applied in a prospective manner to screen available libraries of kinase inhibitors from Selleck and GlaxoSmithKline (GSK). Compounds that showed favorable prediction were then tested in vitro for PRK1 inhibition. Some of the hits were found to inhibit PRK1 in the low-nanomolar range. Three in vitro hits were additionally tested in a mass-spectrometry-based cellular kinase profiling assay to examine selectivity. Our findings show that nanomolar and drug-like inhibitors can be identified by the virtual screening approach presented herein. The identified inhibitors are valuable tools for gaining a better understanding of PRK1 inhibition, and the identified hits can serve as starting points for further chemical optimization.


Subject(s)
Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Thermodynamics , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Molecular Structure , Protein Kinase C/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Quantitative Structure-Activity Relationship
9.
Angew Chem Int Ed Engl ; 55(6): 2252-6, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26748890

ABSTRACT

Sirtuins are NAD(+)-dependent protein deacylases that cleave off acetyl groups, as well as other acyl groups, from the ɛ-amino group of lysines in histones and other substrate proteins. Dysregulation of human Sirt2 activity has been associated with the pathogenesis of cancer, inflammation, and neurodegeneration, thus making Sirt2 a promising target for pharmaceutical intervention. Here, based on a crystal structure of Sirt2 in complex with an optimized sirtuin rearranging ligand (SirReal) that shows improved potency, water solubility, and cellular efficacy, we present the development of the first Sirt2-selective affinity probe. A slow dissociation of the probe/enzyme complex offers new applications for SirReals, such as biophysical characterization, fragment-based screening, and affinity pull-down assays. This possibility makes the SirReal probe an important tool for studying sirtuin biology.


Subject(s)
Molecular Probes/analysis , Molecular Probes/chemistry , Sirtuin 2/analysis , Sirtuin 2/chemistry , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Molecular Probes/chemical synthesis , Molecular Structure , Sirtuin 2/metabolism , Solubility , Structure-Activity Relationship
10.
J Med Chem ; 59(4): 1599-612, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26696402

ABSTRACT

Sirtuins are NAD(+)-dependent protein deacylases that cleave off acetyl but also other acyl groups from the ε-amino group of lysines in histones and other substrate proteins. Dysregulation of human Sirt2 (hSirt2) activity has been associated with the pathogenesis of cancer, inflammation, and neurodegeneration, which makes the modulation of hSirt2 activity a promising strategy for pharmaceutical intervention. The sirtuin rearranging ligands (SirReals) have recently been discovered by us as highly potent and isotype-selective hSirt2 inhibitors. Here, we present a well-defined structure-activity relationship study, which rationalizes the unique features of the SirReals and probes the limits of modifications on this scaffold regarding inhibitor potency. Moreover, we present a crystal structure of hSirt2 in complex with an optimized SirReal derivative that exhibits an improved in vitro activity. Lastly, we show cellular hyperacetylation of the hSirt2 targeted tubulin caused by our improved lead structure.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Sirtuin 2/antagonists & inhibitors , Thiazoles/chemistry , Thiazoles/pharmacology , Amination , Crystallography, X-Ray , HeLa Cells , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Sirtuin 2/chemistry , Sirtuin 2/metabolism , Structure-Activity Relationship , Tubulin/metabolism
11.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 12): 1498-510, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26625292

ABSTRACT

Sirtuins constitute a family of NAD(+)-dependent enzymes that catalyse the cleavage of various acyl groups from the ℇ-amino group of lysines. They regulate a series of cellular processes and their misregulation has been implicated in various diseases, making sirtuins attractive drug targets. To date, only a few sirtuin modulators have been reported that are suitable for cellular research and their development has been hampered by a lack of structural information. In this work, microseed matrix seeding (MMS) was used to obtain crystals of human Sirt3 in its apo form and of human Sirt2 in complex with ADP ribose (ADPR). Crystal formation using MMS was predictable, less error-prone and yielded a higher number of crystals per drop than using conventional crystallization screening methods. The crystals were used to solve the crystal structures of apo Sirt3 and of Sirt2 in complex with ADPR at an improved resolution, as well as the crystal structures of Sirt2 in complex with ADPR and the indoles EX527 and CHIC35. These Sirt2-ADPR-indole complexes unexpectedly contain two indole molecules and provide novel insights into selective Sirt2 inhibition. The MMS approach for Sirt2 and Sirt3 may be used as the basis for structure-based optimization of Sirt2/3 inhibitors in the future.


Subject(s)
Crystallography, X-Ray , Sirtuin 2/chemistry , Sirtuin 3/chemistry , Adenosine Diphosphate Ribose/chemistry , Amino Acid Sequence , Catalytic Domain , Crystallization , Enzyme Inhibitors/pharmacology , Humans , Indoles/pharmacology , Models, Molecular , Molecular Sequence Data , NAD/metabolism , Protein Structure, Secondary
12.
Nat Commun ; 6: 6263, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25672491

ABSTRACT

Sirtuins are a highly conserved class of NAD(+)-dependent lysine deacylases. The human isotype Sirt2 has been implicated in the pathogenesis of cancer, inflammation and neurodegeneration, which makes the modulation of Sirt2 activity a promising strategy for pharmaceutical intervention. A rational basis for the development of optimized Sirt2 inhibitors is lacking so far. Here we present high-resolution structures of human Sirt2 in complex with highly selective drug-like inhibitors that show a unique inhibitory mechanism. Potency and the unprecedented Sirt2 selectivity are based on a ligand-induced structural rearrangement of the active site unveiling a yet-unexploited binding pocket. Application of the most potent Sirtuin-rearranging ligand, termed SirReal2, leads to tubulin hyperacetylation in HeLa cells and induces destabilization of the checkpoint protein BubR1, consistent with Sirt2 inhibition in vivo. Our structural insights into this unique mechanism of selective sirtuin inhibition provide the basis for further inhibitor development and selective tools for sirtuin biology.


Subject(s)
Sirtuin 2/antagonists & inhibitors , Sirtuin 2/chemistry , Acetamides/chemistry , Acetamides/pharmacology , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Ligands , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Thiazoles/chemistry , Thiazoles/pharmacology
13.
J Mol Biol ; 426(20): 3442-53, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-24657767

ABSTRACT

Schistosomiasis, caused by the parasitic flatworm Schistosoma mansoni and related species, is a tropical disease that affects over 200 million people worldwide. A new approach for targeting eukaryotic parasites is to tackle their dynamic epigenetic machinery that is necessary for the extensive phenotypic changes during the life cycle of the parasite. Recently, we identified S. mansoni histone deacetylase 8 (smHDAC8) as a potential target for antiparasitic therapy. Here, we present results on the investigations of a focused set of HDAC (histone deacetylase) inhibitors on smHDAC8. Besides several active hydroxamates, we identified a thiol-based inhibitor that inhibited smHDAC8 activity in the micromolar range with unexpected selectivity over the human isotype, which has not been observed so far. The crystal structure of smHDAC8 complexed with the thiol derivative revealed that the inhibitor is accommodated in the catalytic pocket, where it interacts with both the catalytic zinc ion and the essential catalytic tyrosine (Y341) residue via its mercaptoacetamide warhead. To our knowledge, this is the first complex crystal structure of any HDAC inhibited by a mercaptoacetamide inhibitor, and therefore, this finding offers a rationale for further improvement. Finally, an ester prodrug of the thiol HDAC inhibitor exhibited antiparasitic activity on cultured schistosomes in a dose-dependent manner.


Subject(s)
Antiparasitic Agents/chemistry , Helminth Proteins/chemistry , Histone Deacetylases/chemistry , Schistosoma mansoni/enzymology , Thioacetamide/chemistry , Animals , Antiparasitic Agents/metabolism , Antiparasitic Agents/pharmacology , Apoptosis/drug effects , Biocatalysis/drug effects , Crystallography, X-Ray , Helminth Proteins/antagonists & inhibitors , Helminth Proteins/metabolism , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/metabolism , Hydroxamic Acids/pharmacology , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Protein Binding , Protein Structure, Tertiary , Schistosoma mansoni/drug effects , Schistosoma mansoni/physiology , Schistosomiasis mansoni/parasitology , Thioacetamide/metabolism , Thioacetamide/pharmacology , Vorinostat
14.
J Chem Inf Model ; 54(1): 138-50, 2014 Jan 27.
Article in English | MEDLINE | ID: mdl-24377786

ABSTRACT

Protein kinase C Related Kinase 1 (PRK1) has been shown to be involved in the regulation of androgen receptor signaling and has been identified as a novel potential drug target for prostate cancer therapy. Since there is no PRK1 crystal structure available to date, multiple PRK1 homology models were generated in order to address the protein flexibility. An in-house library of compounds tested on PRK1 was docked into the ATP binding site of the generated models. In most cases a correct pose of the inhibitors could be identified by ensemble docking, while there is still a challenge of finding a reasonable scoring function that is able to rank compounds according to their biological activity. We estimated the binding free energy for our data set of structurally diverse PRK1 inhibitors using the MM-PB(GB)SA and QM/MM-GBSA methods. The obtained results demonstrate that a correlation between calculated binding free energies and experimental IC50 values was found to be usually higher than using docking scores. Furthermore, the developed approach was tested on a set of diverse PRK1 inhibitors taken from literature, which resulted in a significant correlation. The developed method is computationally inexpensive and can be applied as a postdocking filter in virtual screening as well as for optimization of PRK1 inhibitors in order to prioritize compounds for further biological characterization.


Subject(s)
Drug Evaluation, Preclinical/methods , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Binding Sites , Computational Biology , Databases, Pharmaceutical , Drug Evaluation, Preclinical/statistics & numerical data , Humans , Male , Models, Molecular , Molecular Dynamics Simulation , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Protein Conformation , Protein Kinase C/chemistry , Quantitative Structure-Activity Relationship , Software , Structural Homology, Protein , User-Computer Interface
15.
PLoS One ; 7(4): e34973, 2012.
Article in English | MEDLINE | ID: mdl-22532837

ABSTRACT

BACKGROUND: Epigenetics is defined as heritable changes in gene expression that are not based on changes in the DNA sequence. Posttranslational modification of histone proteins is a major mechanism of epigenetic regulation. The kinase PRK1 (protein kinase C related kinase 1, also known as PKN1) phosphorylates histone H3 at threonine 11 and is involved in the regulation of androgen receptor signalling. Thus, it has been identified as a novel drug target but little is known about PRK1 inhibitors and consequences of its inhibition. METHODOLOGY/PRINCIPAL FINDING: Using a focused library screening approach, we identified the clinical candidate lestaurtinib (also known as CEP-701) as a new inhibitor of PRK1. Based on a generated 3D model of the PRK1 kinase using the homolog PKC-theta (protein kinase c theta) protein as a template, the key interaction of lestaurtinib with PRK1 was analyzed by means of molecular docking studies. Furthermore, the effects on histone H3 threonine phosphorylation and androgen-dependent gene expression was evaluated in prostate cancer cells. CONCLUSIONS/SIGNIFICANCE: Lestaurtinib inhibits PRK1 very potently in vitro and in vivo. Applied to cell culture it inhibits histone H3 threonine phosphorylation and androgen-dependent gene expression, a feature that has not been known yet. Thus our findings have implication both for understanding of the clinical activity of lestaurtinib as well as for future PRK1 inhibitors.


Subject(s)
Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Histones/metabolism , Prostatic Neoplasms/enzymology , Protein Kinase C/antagonists & inhibitors , Androgens/physiology , Cell Line, Tumor , Cells, Cultured , Furans , Histones/genetics , Humans , Male , Phosphorylation/drug effects , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
16.
ACS Med Chem Lett ; 3(12): 1050-3, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-24900427

ABSTRACT

NAD(+)-dependent histone deacetylases (sirtuins) play important roles in epigenetic regulation but also through nonhistone substrates for other key cellular events and have been linked to the pathogenesis of cancer, neurodegeneration, and metabolic diseases. The subtype Sirt5 has been shown recently to act as a desuccinylating and demalonylating enzyme. We have established an assay for biochemical testing of Sirt5 using a small labeled succinylated lysine derivative. We present a comparative study on the profiling of several established sirtuin inhibitors on Sirt1-3 as well as Sirt5 and also present initial results on a screening for new compounds that block Sirt5. Thiobarbiturates were identified as new Sirt5 inhibitors in the low micromolar range, which are selective over Sirt3 that can be found in the same cell compartment as Sirt5.

17.
Aging (Albany NY) ; 3(9): 818, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21937769
18.
J Med Chem ; 54(10): 3492-9, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21528845

ABSTRACT

We report the design and concise synthesis, in two steps from commercially available material, of novel, bioactive derivatives of the enzyme cofactor nicotinamide adenine dinucleotide (NAD). The new synthetic dinucleotides act as sirtuin (SIRT) inhibitors and show isoform selectivity for SIRT2 over SIRT1. An NMR-based conformational analysis suggests that the conformational preferences of individual analogues may contribute to their isoform selectivity.


Subject(s)
NAD/chemistry , Sirtuin 1/chemistry , Sirtuin 2/chemistry , Chemistry, Pharmaceutical/methods , Drug Design , Humans , Hydrogen Bonding , Magnetic Resonance Spectroscopy/methods , Models, Chemical , Oxidation-Reduction , Protein Binding , Protein Conformation , Protein Isoforms , Recombinant Fusion Proteins/chemistry , Sirtuin 1/antagonists & inhibitors , Sirtuin 2/antagonists & inhibitors
19.
Bioorg Med Chem ; 19(12): 3669-77, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21315612

ABSTRACT

Small molecules interfering with posttranslational modification of histones are of interest as tools to study epigenetic regulation of gene transcription. Specifically, drugs that interfere with histone deacetylation could be useful to induce differentiation, growth arrest as well as apoptotic cell death in tumor cells. One class of histone deacetylases is known as sirtuins some of which (Saccharomyces cerevisiae Sir2) are for example inhibited by the lactone splitomicin leading to telomeric silencing in yeast. However, splitomicin is only a micromolar inhibitor of yeast Sir2 and does not inhibit human subtypes and the lactone is prone to hydrolytic ring opening. In preliminary SAR-studies, splitomicin analogs lacking this hydrolytically labile ring were described as inactive while the naphthalene moiety could successfully be replaced by smaller aromatic rings in a fragment-like dihydrocoumarin. Here we report the synthesis and biological activity of a series of hydrolytically stable analogs with activity against human SIRT1 and 2. These comparatively small compounds characterized by high ligand efficiency are used as a starting point toward the development of specific inhibitors of histone deacetylases from the class of sirtuins.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Naphthalenes/chemical synthesis , Naphthalenes/pharmacology , Pyrones/chemical synthesis , Pyrones/pharmacology , Sirtuins/antagonists & inhibitors , Drug Delivery Systems , Enzyme Inhibitors/chemistry , Humans , Inhibitory Concentration 50 , Models, Molecular , NAD/chemistry , Naphthalenes/chemistry , Pyrones/chemistry
20.
Biochim Biophys Acta ; 1799(10-12): 726-39, 2010.
Article in English | MEDLINE | ID: mdl-20601279

ABSTRACT

Histone deacetylases (HDACs) are enzymes that cleave acetyl groups from acetyl-lysine residues in histones and various nonhistone proteins. Unlike the other three of the four classes of HDACs that have been identified in humans, which are zinc-dependent amidohydrolases, class III HDACs depend on nicotinamide adenine dinucleotide (NAD(+)) for their catalytic activity. The seven members of the class III HDACs are also named sirtuins for their homology to Sir2p, a yeast histone deacetylase. Sirtuin inhibitors have been critical for the linkage of sirtuin activity to many physiological and pathological processes, and sirtuin activity has been associated with the pathogenesis of cancer, HIV, and metabolic and neurological diseases. Presented here is an overview of the many sirtuin inhibitors that have provided insight into the biological role of sirtuins.


Subject(s)
Histone Deacetylase Inhibitors , NAD , Sirtuins , Animals , HIV Infections/drug therapy , HIV Infections/enzymology , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/therapeutic use , Humans , Metabolic Diseases/drug therapy , Metabolic Diseases/enzymology , NAD/chemistry , NAD/metabolism , Neoplasms/drug therapy , Neoplasms/enzymology , Nervous System Diseases/drug therapy , Nervous System Diseases/enzymology , Sirtuins/chemistry , Sirtuins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...