Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
SLAS Discov ; 25(2): 137-147, 2020 02.
Article in English | MEDLINE | ID: mdl-31566060

ABSTRACT

Thermal shift assays (TSAs) can reveal changes in protein structure, due to a resultant change in protein thermal stability. Since proteins are often stabilized upon binding of ligand molecules, these assays can provide a readout for protein target engagement. TSA has traditionally been applied using purified proteins and more recently has been extended to study target engagement in cellular environments with the emergence of cellular thermal shift assays (CETSAs). The utility of CETSA in confirming molecular interaction with targets in a more native context, and the desire to apply this technique more broadly, has fueled the emergence of higher-throughput techniques for CETSA (HT-CETSA). Recent studies have demonstrated that HT-CETSA can be performed in standard 96-, 384-, and 1536-well microtiter plate formats using methods such as beta-galactosidase and NanoLuciferase reporters and AlphaLISA assays. HT-CETSA methods can be used to select and characterize compounds from high-throughput screens and to prioritize compounds in lead optimization by facilitating dose-response experiments. In conjunction with cellular and biochemical activity assays for targets, HT-CETSA can be a valuable addition to the suite of assays available to characterize molecules of interest. Despite the successes in implementing HT-CETSA for a diverse set of targets, caveats and challenges must also be recognized to avoid overinterpretation of results. Here, we review the current landscape of HT-CETSA and discuss the methodologies, practical considerations, challenges, and applications of this approach in research and drug discovery. Additionally, a perspective on potential future directions for the technology is presented.


Subject(s)
Biomedical Research/trends , Drug Discovery/trends , High-Throughput Screening Assays/methods , Humans , Ligands
2.
Sci Rep ; 9(1): 13078, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31511536

ABSTRACT

Significant resource is spent by drug discovery project teams to generate numerous, yet unique target constructs for the multiple platforms used to drive drug discovery programs including: functional assays, biophysical studies, structural biology, and biochemical high throughput screening campaigns. To improve this process, we developed Modular Protein Ligation (MPL), a combinatorial reagent platform utilizing Expressed Protein Ligation to site-specifically label proteins at the C-terminus with a variety of cysteine-lysine dipeptide conjugates. Historically, such proteins have been chemically labeled non-specifically through surface amino acids. To demonstrate the feasibility of this approach, we first applied MPL to proteins of varying size in different target classes using different recombinant protein expression systems, which were then evaluated in several different downstream assays. A key advantage to the implementation of this paradigm is that one construct can generate multiple final products, significantly streamlining the reagent generation for multiple early drug discovery project teams.


Subject(s)
Drug Discovery/methods , Proteins/metabolism , Animals , Feasibility Studies , Humans , Ligands , Mice , Models, Molecular , Protein Conformation , Proteins/chemistry
3.
Anal Biochem ; 559: 30-33, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30142329

ABSTRACT

Aggregated compounds can promiscuously and nonspecifically associate with proteins resulting in either false inhibition or activation of many different protein target classes. We developed a high-content imaging assay in a 384-well format using fluorescently labeled target proteins and an Operetta cell imager to screen for compound aggregates that interact with target proteins. The high-throughput assay can not only directly detect the interaction between compound aggregators and the target of interest, but also determine the critical aggregation concentration (CAC) of a given promiscuous small molecule.


Subject(s)
Fluorescent Dyes/chemistry , High-Throughput Screening Assays , Optical Imaging , Proteins/chemistry , Humans , Particle Size , Protein Aggregates , Surface Properties
4.
SLAS Discov ; 23(1): 34-46, 2018 01.
Article in English | MEDLINE | ID: mdl-28957646

ABSTRACT

A persistent problem in early small-molecule drug discovery is the frequent lack of rank-order correlation between biochemical potencies derived from initial screens using purified proteins and the diminished potency and efficacy observed in subsequent disease-relevant cellular phenotypic assays. The introduction of the cellular thermal shift assay (CETSA) has bridged this gap by enabling assessment of drug target engagement directly in live cells based on ligand-induced changes in protein thermal stability. Initial success in applying CETSA across multiple drug target classes motivated our investigation into replacing the low-throughput, manually intensive Western blot readout with a quantitative, automated higher-throughput assay that would provide sufficient capacity to use CETSA as a primary hit qualification strategy. We introduce a high-throughput dose-response cellular thermal shift assay (HTDR-CETSA), a single-pot homogenous assay adapted for high-density microtiter plate format. The assay features titratable BacMam expression of full-length target proteins fused to the DiscoverX 42 amino acid ePL tag in HeLa suspension cells, facilitating enzyme fragment complementation-based chemiluminescent quantification of ligand-stabilized soluble protein. This simplified format can accommodate determination of full-dose CETSA curves for hundreds of individual compounds/analyst/day in replicates. HTDR-CETSA data generated for substrate site and alternate binding mode inhibitors of the histone-lysine N-methyltransferase SMYD3 in HeLa suspension cells demonstrate excellent correlation with rank-order potencies observed in cellular mechanistic assays and direct translation to target engagement of endogenous Smyd3 in cancer-relevant cell lines. We envision this workflow to be generically applicable to HTDR-CETSA screening spanning a wide variety of soluble intracellular protein target classes.


Subject(s)
Drug Discovery/methods , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Cell Culture Techniques , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Activation , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Small Molecule Libraries , Workflow
5.
Protein Expr Purif ; 97: 9-16, 2014 May.
Article in English | MEDLINE | ID: mdl-24534072

ABSTRACT

The mammalian pyruvate dehydrogenase complex (PDC) is a multi-component mitochondrial enzyme that plays a key role in the conversion of pyruvate to acetyl-CoA connecting glycolysis to the citric acid cycle. Recent studies indicate that targeting the regulation of PDC enzymatic activity might offer therapeutic opportunities by inhibiting cancer cell metabolism. To facilitate drug discovery in this area, a well defined PDC sample is needed. Here, we report a new method of producing functional, recombinant, high quality human PDC complex. All five components were co-expressed in the cytoplasm of baculovirus-infected SF9 cells by deletion of the mitochondrial localization signal sequences of all the components and E1a was FLAG-tagged to facilitate purification. The protein FLAG tagged E1a complex was purified using FLAG-M2 affinity resin, followed by Superdex 200 sizing chromatography. The E2 and E3BP components were then Lipoylated using an enzyme based in vitro process. The resulting PDC is over 90% pure and homogenous. This non-phosphorylated, lipoylated human PDC was demonstrated to produce a robust detection window when used to develop an enzyme coupled assay of PDHK.


Subject(s)
Baculoviridae/genetics , Pyruvate Dehydrogenase Complex/genetics , Sf9 Cells/metabolism , Animals , Cloning, Molecular , Gene Expression , Humans , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Pyruvate Dehydrogenase Complex/isolation & purification , Pyruvate Dehydrogenase Complex/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
6.
J Am Chem Soc ; 136(9): 3370-3, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24533830

ABSTRACT

Proteins as well as small molecules have demonstrated success as therapeutic agents, but their pharmacologic properties sometimes fall short against particular drug targets. Although the adenosine 2a receptor (A(2A)R) has been identified as a promising target for immunotherapy, small molecule A(2A)R agonists have suffered from short pharmacokinetic half-lives and the potential for toxicity by modulating nonimmune pathways. To overcome these limitations, we have tethered the A(2A)R agonist CGS-21680 to the immunoglobulin Fc domain using expressed protein ligation with Sf9 cell secreted protein. The protein small molecule conjugate Fc-CGS retained potent Fc receptor and A(2A)R interactions and showed superior properties as a therapeutic for the treatment of a mouse model of autoimmune pneumonitis. This approach may provide a general strategy for optimizing small molecule therapeutics.


Subject(s)
Adenosine/analogs & derivatives , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/pharmacology , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Phenethylamines/chemistry , Adenosine/chemistry , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Mice , Models, Molecular , Protein Conformation
7.
Structure ; 19(2): 221-31, 2011 Feb 09.
Article in English | MEDLINE | ID: mdl-21256037

ABSTRACT

Yeast Rtt109 promotes nucleosome assembly and genome stability by acetylating K9, K27, and K56 of histone H3 through interaction with either of two distinct histone chaperones, Vps75 or Asf1. We report the crystal structure of an Rtt109-AcCoA/Vps75 complex revealing an elongated Vps75 homodimer bound to two globular Rtt109 molecules to form a symmetrical holoenzyme with a ∼12 Å diameter central hole. Vps75 and Rtt109 residues that mediate complex formation in the crystals are also important for Rtt109-Vps75 interaction and H3K9/K27 acetylation both in vitro and in yeast cells. The same Rtt109 residues do not participate in Asf1-mediated Rtt109 acetylation in vitro or H3K56 acetylation in yeast cells, demonstrating that Asf1 and Vps75 dictate Rtt109 substrate specificity through distinct mechanisms. These studies also suggest that Vps75 binding stimulates Rtt109 catalytic activity by appropriately presenting the H3-H4 substrate within the central cavity of the holoenzyme to promote H3K9/K27 acetylation of new histones before deposition.


Subject(s)
Acetyl Coenzyme A/metabolism , Cell Cycle Proteins/metabolism , Histone Acetyltransferases/metabolism , Histones/metabolism , Molecular Chaperones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Acetylation , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Chromatin Assembly and Disassembly , Crystallography, X-Ray , Gene Expression , Genomic Instability , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/genetics , Histones/genetics , Humans , Lysine/metabolism , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Mutagenesis, Site-Directed , Protein Binding , Recombinant Fusion Proteins , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Substrate Specificity
8.
J Am Chem Soc ; 132(29): 9986-7, 2010 Jul 28.
Article in English | MEDLINE | ID: mdl-20608637

ABSTRACT

Protein acetylation on Lys residues is recognized as a significant post-translational modification in cells, but it is often difficult to discern the direct structural and functional effects of individual acetylation events. Here we describe a new tool, methylthiocarbonyl-aziridine, to install acetyl-Lys mimics site-specifically into peptides and proteins by alkylation of Cys residues. We demonstrate that the resultant thiocarbamate modification can be recognized by the Brdt bromodomain and site-specific antiacetyl-Lys antibodies, is resistant to histone deacetylase cleavage, and can confer activation of the histone acetyltransferase Rtt109 by simulating autoacetylation. We also use this approach to obtain functional evidence that acetylation of CK2 protein kinase on Lys102 can stimulate its catalytic activity.


Subject(s)
Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Cysteine/metabolism , Lysine/metabolism , Peptides/metabolism , Proteins/metabolism , Acetylation , Alkylation , Animals , Aziridines/chemistry , Aziridines/metabolism , Binding Sites , Histones/chemistry , Histones/metabolism , Peptides/chemistry , Proteins/chemistry , Substrate Specificity
9.
Chem Biol ; 17(5): 471-82, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20534345

ABSTRACT

The histone acetyltransferase (HAT) p300/CBP is a transcriptional coactivator implicated in many gene regulatory pathways and protein acetylation events. Although p300 inhibitors have been reported, a potent, selective, and readily available active-site-directed small molecule inhibitor is not yet known. Here we use a structure-based, in silico screening approach to identify a commercially available pyrazolone-containing small molecule p300 HAT inhibitor, C646. C646 is a competitive p300 inhibitor with a K(i) of 400 nM and is selective versus other acetyltransferases. Studies on site-directed p300 HAT mutants and synthetic modifications of C646 confirm the importance of predicted interactions in conferring potency. Inhibition of histone acetylation and cell growth by C646 in cells validate its utility as a pharmacologic probe and suggest that p300/CBP HAT is a worthy anticancer target.


Subject(s)
Benzoates/chemistry , Enzyme Inhibitors/chemistry , Histone Acetyltransferases/antagonists & inhibitors , Pyrazoles/chemistry , p300-CBP Transcription Factors/antagonists & inhibitors , Acetylation , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzoates/pharmacology , Binding Sites , Binding, Competitive , Catalytic Domain , Cell Line, Tumor , Computer Simulation , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Histone Acetyltransferases/metabolism , Ligands , Mice , Pyrazoles/pharmacology , Pyrazolones/chemistry , Small Molecule Libraries/chemistry , Structure-Activity Relationship , p300-CBP Transcription Factors/metabolism
10.
Nat Struct Mol Biol ; 15(7): 738-45, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18568037

ABSTRACT

Rtt109, also known as KAT11, is a recently characterized fungal-specific histone acetyltransferase (HAT) that modifies histone H3 lysine 56 (H3K56) to promote genome stability. Rtt109 does not show sequence conservation with other known HATs and depends on association with either of two histone chaperones, Asf1 or Vps75, for HAT activity. Here we report the X-ray crystal structure of an Rtt109-acetyl coenzyme A complex and carry out structure-based mutagenesis, combined with in vitro biochemical studies of the Rtt109-Vps75 complex and studies of Rtt109 function in vivo. The Rtt109 structure reveals noteworthy homology to the metazoan p300/CBP HAT domain but exhibits functional divergence, including atypical catalytic properties and mode of cofactor regulation. The structure reveals a buried autoacetylated lysine residue that we show is also acetylated in the Rtt109 protein purified from yeast cells. Implications for understanding histone substrate and chaperone binding by Rtt109 are discussed.


Subject(s)
Histone Acetyltransferases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Structural Homology, Protein , p300-CBP Transcription Factors/chemistry , Acetyl Coenzyme A/chemistry , Acetylation/drug effects , Animals , Binding Sites , Crystallography, X-Ray , Histones/metabolism , Lysine/metabolism , Models, Molecular , Mutagenesis , Mutagens/pharmacology , Mutant Proteins/metabolism , Protein Structure, Secondary , Saccharomyces cerevisiae/drug effects , Structure-Activity Relationship
11.
J Med Chem ; 50(22): 5330-8, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17924613

ABSTRACT

Serotonin N-acetyltransferase (arylalkylamine N-acetyltransferase, AANAT) is a member of the GCN5 N-acetyltransferase (GNAT) superfamily and catalyzes the penultimate step in the biosynthesis of melatonin; a large daily rhythm in AANAT activity drives the daily rhythm in circulating melatonin. We have used a structure-based computational approach to identify the first druglike and selective inhibitors of AANAT. Approximately 1.2 million compounds were virtually screened by 3D high-throughput docking into the active site of X-ray structures for AANAT, and in total 241 compounds were tested as inhibitors. One compound class, containing a rhodanine scaffold, exhibited low micromolar competitive inhibition against acetyl-CoA (AcCoA) and proved to be effective in blocking melatonin production in pineal cells. Compounds from this class are predicted to bind as bisubstrate inhibitors through interactions with the AcCoA and serotonin binding sites. Overall, this study demonstrates the feasibility of using virtual screening to identify small molecules that are selective inhibitors of AANAT.


Subject(s)
Arylalkylamine N-Acetyltransferase/antagonists & inhibitors , Arylalkylamine N-Acetyltransferase/chemistry , Enzyme Inhibitors/chemistry , Models, Molecular , Quantitative Structure-Activity Relationship , Acetyl Coenzyme A/antagonists & inhibitors , Acetyl Coenzyme A/chemistry , Animals , Arylalkylamine N-Acetyltransferase/biosynthesis , Binding Sites , Cells, Cultured , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Melatonin/antagonists & inhibitors , Melatonin/biosynthesis , Pineal Gland/cytology , Protein Binding , Protein Conformation , Rats , Rhodanine/analogs & derivatives , Rhodanine/chemistry , Rhodanine/pharmacology , Tryptamines/chemistry , Tryptamines/pharmacology
12.
J Biol Chem ; 282(50): 36603-13, 2007 Dec 14.
Article in English | MEDLINE | ID: mdl-17925393

ABSTRACT

The human monocytic leukemia zinc finger (MOZ) protein is an essential transcriptional coactivator and histone acetyltransferase (HAT) that plays a primary role in the differentiation of erythroid and myeloid cells and is required to maintain hematopoietic stem cells. Chromosomal translocations involving the HAT-encoded region are also associated with acute myeloid leukemia. Here we present the x-ray crystal structure of the MOZ HAT domain and related biochemical studies. We find that the HAT domain contains a central region that is structurally and functionally conserved with the yeast MYST HAT protein Esa1, but contains more divergent N- and C-terminal regions harboring a TFIIIA-type zinc finger and helix-turn-helix DNA-binding motifs. Solution DNA-binding and acetyltransferase activity assays, in concert with mutagenesis, confirm that the MOZ HAT domain binds strongly to DNA through the zinc finger and helix-turn-helix motifs and that DNA binding and catalysis are not mutually exclusive. Consistent with the DNA-binding properties of MOZ, we also show that MOZ is able to acetylate nucleosomes and free histones equally well, whereas other HATs prefer free histones. Our results reveal, for the first time, that enzymatic and DNA-targeting activities can be contained within the same chromatin regulatory domain.


Subject(s)
Histone Acetyltransferases/chemistry , Acetylation , Amino Acid Motifs/physiology , Animals , Cell Differentiation/physiology , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Histone Acetyltransferases/metabolism , Histones/metabolism , Humans , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Nucleosomes/metabolism , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Structure-Activity Relationship , Translocation, Genetic/physiology , Xenopus laevis
13.
Mol Cell ; 24(6): 841-51, 2006 Dec 28.
Article in English | MEDLINE | ID: mdl-17189187

ABSTRACT

The ability of p53 to induce apoptosis plays an important role in tumor suppression. Here, we describe a previously unknown posttranslational modification of the DNA-binding domain of p53. This modification, acetylation of lysine 120 (K120), occurs rapidly after DNA damage and is catalyzed by the MYST family acetyltransferases hMOF and TIP60. Mutation of K120 to arginine, as occurs in human cancer, debilitates K120 acetylation and diminishes p53-mediated apoptosis without affecting cell-cycle arrest. The K120R mutation selectively blocks the transcription of proapoptotic target genes such as BAX and PUMA while the nonapoptotic targets p21 and hMDM2 remain unaffected. Consistent with this, depletion of hMOF and/or TIP60 inhibits the ability of p53 to activate BAX and PUMA transcription. Furthermore, the acetyllysine 120 (acetyl-K120) form of p53 specifically accumulates at proapoptotic target genes. These data suggest that K120 acetylation may help distinguish the cell-cycle arrest and apoptotic functions of p53.


Subject(s)
Apoptosis , Histone Acetyltransferases/physiology , Tumor Suppressor Protein p53/metabolism , Acetylation , Amino Acid Sequence , Apoptosis Regulatory Proteins/genetics , Binding Sites , Cell Cycle , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , Lysine Acetyltransferase 5 , Molecular Sequence Data , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , bcl-2-Associated X Protein/genetics
14.
Curr Opin Struct Biol ; 16(6): 668-75, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17085043

ABSTRACT

Protein tyrosine kinases (PTKs) are enzymes that catalyze the phosphorylation of tyrosyl residues. They are important in physiological and pathophysiological processes. Protein substrates of PTKs are often difficult to discern, but recently reported methods have helped to identify targets and characterize their structural interactions with kinases. A mechanism-based bisubstrate analog strategy has given X-ray crystallographic insights into how several topical PTKs, including the insulin receptor, Abl and epidermal growth factor receptor, interact with tyrosine-containing peptide substrates. These PTK co-crystal structures reveal both conserved and specialized features of recognition that probably contribute to substrate selection and the individual functions of these key enzymes.


Subject(s)
Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Catalytic Domain , ErbB Receptors/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , In Vitro Techniques , Models, Molecular , Molecular Sequence Data , Protein Array Analysis , Protein Conformation , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Receptor, Insulin/chemistry , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
15.
Curr Opin Struct Biol ; 15(6): 673-80, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16263263

ABSTRACT

The post-translational modification of histones plays an important role in chromatin regulation, a process that insures the fidelity of gene expression and other DNA transactions. Equally important as the enzymes that generate these modifications are the enzymes that remove them. Recent studies have identified some of the enzymes that remove histone modifications and have characterized their activities. In addition, structural and biochemical studies of these enzymes have focused on the histone lysine deacetylases HDAC8 and sirtuins, and on the arginine and lysine demethylases PAD and BHC110/LSD1, respectively. These new findings may be used as a context to present new information that contributes to our understanding of chromatin regulation, and to pose remaining questions pertaining to the activities of these enzymes and the roles they play in chromatin regulation.


Subject(s)
Histone Deacetylases/metabolism , Histones/metabolism , Protein Processing, Post-Translational , Repressor Proteins/metabolism , Sirtuins/metabolism , Animals , Histone Deacetylases/chemistry , Histone Demethylases , Humans , Methylation , Models, Molecular , Oxidoreductases, N-Demethylating/chemistry , Oxidoreductases, N-Demethylating/metabolism , Protein Conformation , Repressor Proteins/chemistry , Sirtuins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...