Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
1.
Nat Chem Biol ; 18(1): 56-63, 2022 01.
Article in English | MEDLINE | ID: mdl-34782742

ABSTRACT

Nuclear receptor-binding SET domain-containing 2 (NSD2) is the primary enzyme responsible for the dimethylation of lysine 36 of histone 3 (H3K36), a mark associated with active gene transcription and intergenic DNA methylation. In addition to a methyltransferase domain, NSD2 harbors two proline-tryptophan-tryptophan-proline (PWWP) domains and five plant homeodomains (PHDs) believed to serve as chromatin reading modules. Here, we report a chemical probe targeting the N-terminal PWWP (PWWP1) domain of NSD2. UNC6934 occupies the canonical H3K36me2-binding pocket of PWWP1, antagonizes PWWP1 interaction with nucleosomal H3K36me2 and selectively engages endogenous NSD2 in cells. UNC6934 induces accumulation of endogenous NSD2 in the nucleolus, phenocopying the localization defects of NSD2 protein isoforms lacking PWWP1 that result from translocations prevalent in multiple myeloma (MM). Mutations of other NSD2 chromatin reader domains also increase NSD2 nucleolar localization and enhance the effect of UNC6934. This chemical probe and the accompanying negative control UNC7145 will be useful tools in defining NSD2 biology.


Subject(s)
Cell Nucleolus/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Molecular Probes/chemistry , Protein Domains , Repressor Proteins/metabolism , Methylation , Multiple Myeloma/metabolism , Nucleosomes/metabolism
2.
J Am Chem Soc ; 145(14): 8176-8188, 2023 04 12.
Article in English | MEDLINE | ID: mdl-36976643

ABSTRACT

Nuclear receptor-binding SET domain-containing 2 (NSD2) plays important roles in gene regulation, largely through its ability to dimethylate lysine 36 of histone 3 (H3K36me2). Despite aberrant activity of NSD2 reported in numerous cancers, efforts to selectively inhibit the catalytic activity of this protein with small molecules have been unsuccessful to date. Here, we report the development of UNC8153, a novel NSD2-targeted degrader that potently and selectively reduces the cellular levels of both NSD2 protein and the H3K36me2 chromatin mark. UNC8153 contains a simple warhead that confers proteasome-dependent degradation of NSD2 through a novel mechanism. Importantly, UNC8153-mediated reduction of H3K36me2 through the degradation of NSD2 results in the downregulation of pathological phenotypes in multiple myeloma cells including mild antiproliferative effects in MM1.S cells containing an activating point mutation and antiadhesive effects in KMS11 cells harboring the t(4;14) translocation that upregulates NSD2 expression.


Subject(s)
Chromatin , Histones , Histones/metabolism , Gene Expression Regulation , Cell Line, Tumor , Down-Regulation
3.
J Chem Inf Model ; 63(13): 4070-4078, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37350740

ABSTRACT

DCAF1 functions as a substrate recruitment subunit for the RING-type CRL4DCAF1 and the HECT family EDVPDCAF1 E3 ubiquitin ligases. The WDR domain of DCAF1 serves as a binding platform for substrate proteins and is also targeted by HIV and SIV lentiviral adaptors to induce the ubiquitination and proteasomal degradation of antiviral host factors. It is therefore attractive both as a potential therapeutic target for the development of chemical inhibitors and as an E3 ligase that could be recruited by novel PROTACs for targeted protein degradation. In this study, we used a proteome-scale drug-target interaction prediction model, MatchMaker, combined with cheminformatics filtering and docking to identify ligands for the DCAF1 WDR domain. Biophysical screening and X-ray crystallographic studies of the predicted binders confirmed a selective ligand occupying the central cavity of the WDR domain. This study shows that artificial intelligence-enabled virtual screening methods can successfully be applied in the absence of previously known ligands.


Subject(s)
Artificial Intelligence , Carrier Proteins , Ligands , Carrier Proteins/chemistry , Ubiquitin-Protein Ligases/metabolism , Machine Learning
4.
Nat Chem Biol ; 15(8): 822-829, 2019 08.
Article in English | MEDLINE | ID: mdl-31285596

ABSTRACT

Here, we report the fragment-based discovery of BI-9321, a potent, selective and cellular active antagonist of the NSD3-PWWP1 domain. The human NSD3 protein is encoded by the WHSC1L1 gene located in the 8p11-p12 amplicon, frequently amplified in breast and squamous lung cancer. Recently, it was demonstrated that the PWWP1 domain of NSD3 is required for the viability of acute myeloid leukemia cells. To further elucidate the relevance of NSD3 in cancer biology, we developed a chemical probe, BI-9321, targeting the methyl-lysine binding site of the PWWP1 domain with sub-micromolar in vitro activity and cellular target engagement at 1 µM. As a single agent, BI-9321 downregulates Myc messenger RNA expression and reduces proliferation in MOLM-13 cells. This first-in-class chemical probe BI-9321, together with the negative control BI-9466, will greatly facilitate the elucidation of the underexplored biological function of PWWP domains.


Subject(s)
Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , CRISPR-Cas Systems , Cell Line , Cell Proliferation/drug effects , Cell Survival , Gene Expression Regulation/drug effects , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Domains , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
5.
Phys Rev Lett ; 125(9): 090401, 2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32915619

ABSTRACT

Alice and Bob each have half of a pair of entangled qubits. Bob measures his half and then passes his qubit to a second Bob who measures again and so on. The goal is to maximize the number of Bobs that can have an expected violation of the Clauser-Horne-Shimony-Holt (CHSH) Bell inequality with the single Alice. This scenario was introduced in [R. Silva et al., Phys. Rev. Lett. 114, 250401 (2015)PRLTAO0031-900710.1103/PhysRevLett.114.250401] where the authors mentioned evidence that when the Bobs act independently and with unbiased inputs then at most two of them can expect to violate the CHSH inequality with Alice. Here we show that, contrary to this evidence, arbitrarily many independent Bobs can have an expected CHSH violation with the single Alice. Our proof is constructive and our measurement strategies can be generalized to work with a larger class of two-qubit states that includes all pure entangled two-qubit states. Since violation of a Bell inequality is necessary for device-independent tasks, our work represents a step towards an eventual understanding of the limitations on how much device-independent randomness can be robustly generated from a single pair of qubits.

6.
J Biol Chem ; 293(9): 3410-3420, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29321206

ABSTRACT

Short-chain acylation of lysine residues has recently emerged as a group of reversible posttranslational modifications in mammalian cells. The diversity of acylation further broadens the landscape and complexity of the proteome. Identification of regulatory enzymes and effector proteins for lysine acylation is critical to understand functions of these novel modifications at the molecular level. Here, we report that the MYST family of lysine acetyltransferases (KATs) possesses strong propionyltransferase activity both in vitro and in cellulo Particularly, the propionyltransferase activity of MOF, MOZ, and HBO1 is as strong as their acetyltransferase activity. Overexpression of MOF in human embryonic kidney 293T cells induced significantly increased propionylation in multiple histone and non-histone proteins, which shows that the function of MOF goes far beyond its canonical histone H4 lysine 16 acetylation. We also resolved the X-ray co-crystal structure of MOF bound with propionyl-coenzyme A, which provides a direct structural basis for the propionyltransferase activity of the MYST KATs. Our data together define a novel function for the MYST KATs as lysine propionyltransferases and suggest much broader physiological impacts for this family of enzymes.


Subject(s)
Histone Acetyltransferases/metabolism , Protein Processing, Post-Translational , Acetylation , Amino Acid Sequence , HEK293 Cells , Histone Acetyltransferases/chemistry , Humans , Lysine/metabolism , Models, Molecular , Protein Conformation , Proteomics
7.
Nat Chem Biol ; 13(3): 317-324, 2017 03.
Article in English | MEDLINE | ID: mdl-28114273

ABSTRACT

Protein lysine methyltransferases (PKMTs) regulate diverse physiological processes including transcription and the maintenance of genomic integrity. Genetic studies suggest that the PKMTs SUV420H1 and SUV420H2 facilitate proficient nonhomologous end-joining (NHEJ)-directed DNA repair by catalyzing the di- and trimethylation (me2 and me3, respectively) of lysine 20 on histone 4 (H4K20). Here we report the identification of A-196, a potent and selective inhibitor of SUV420H1 and SUV420H2. Biochemical and co-crystallization analyses demonstrate that A-196 is a substrate-competitive inhibitor of both SUV4-20 enzymes. In cells, A-196 induced a global decrease in H4K20me2 and H4K20me3 and a concomitant increase in H4K20me1. A-196 inhibited 53BP1 foci formation upon ionizing radiation and reduced NHEJ-mediated DNA-break repair but did not affect homology-directed repair. These results demonstrate the role of SUV4-20 enzymatic activity in H4K20 methylation and DNA repair. A-196 represents a first-in-class chemical probe of SUV4-20 to investigate the role of histone methyltransferases in genomic integrity.


Subject(s)
Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Genomic Instability/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Cell Line, Tumor , Crystallography, X-Ray , DNA Repair/drug effects , Enzyme Inhibitors/chemistry , Heterocyclic Compounds, 4 or More Rings/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Humans , Methylation/drug effects , Models, Molecular , Molecular Structure
8.
Bioorg Med Chem ; 27(17): 3866-3878, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31327677

ABSTRACT

SET domain bifurcated protein 1 (SETDB1) is a human histone-lysine methyltransferase which is amplified in human cancers and was shown to be crucial in the growth of non-small and small cell lung carcinoma. In addition to its catalytic domain, SETDB1 harbors a unique tandem tudor domain which recognizes histone sequences containing both methylated and acetylated lysines, and likely contributes to its localization on chromatin. Using X-ray crystallography and NMR spectroscopy fragment screening approaches, we have identified the first small molecule fragment hits that bind to histone peptide binding groove of the Tandem Tudor Domain (TTD) of SETDB1. Herein, we describe the binding modes of these fragments and analogues and the biophysical characterization of key compounds. These confirmed small molecule fragments will inform the development of potent antagonists of SETDB1 interaction with histones.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Histone-Lysine N-Methyltransferase/isolation & purification , Histone-Lysine N-Methyltransferase/metabolism , Histones/antagonists & inhibitors , Histones/metabolism , Humans , Models, Molecular , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Tudor Domain/drug effects
9.
J Biol Chem ; 292(51): 20947-20959, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29074623

ABSTRACT

UHRF1 is a key mediator of inheritance of epigenetic DNA methylation patterns during cell division and is a putative target for cancer therapy. Recent studies indicate that interdomain interactions critically influence UHRF1's chromatin-binding properties, including allosteric regulation of its histone binding. Here, using an integrative approach that combines small angle X-ray scattering, NMR spectroscopy, and molecular dynamics simulations, we characterized the dynamics of the tandem tudor domain-plant homeodomain (TTD-PHD) histone reader module, including its 20-residue interdomain linker. We found that the apo TTD-PHD module in solution comprises a dynamic ensemble of conformers, approximately half of which are compact conformations, with the linker lying in the TTD peptide-binding groove. These compact conformations are amenable to cooperative, high-affinity histone binding. In the remaining conformations, the linker position was in flux, and the reader adopted both extended and compact states. Using a small-molecule fragment screening approach, we identified a compound, 4-benzylpiperidine-1-carboximidamide, that binds to the TTD groove, competes with linker binding, and promotes open TTD-PHD conformations that are less efficient at H3K9me3 binding. Our work reveals a mechanism by which the dynamic TTD-PHD module can be allosterically targeted with small molecules to modulate its histone reader function for therapeutic or experimental purposes.


Subject(s)
CCAAT-Enhancer-Binding Proteins/chemistry , CCAAT-Enhancer-Binding Proteins/metabolism , Allosteric Regulation , Crystallography, X-Ray , Epigenesis, Genetic , Histones/metabolism , Humans , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Protein Domains , Scattering, Small Angle , Ubiquitin-Protein Ligases , X-Ray Diffraction
10.
Nat Chem Biol ; 12(3): 180-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26807715

ABSTRACT

We report the design and characterization of UNC3866, a potent antagonist of the methyllysine (Kme) reading function of the Polycomb CBX and CDY families of chromodomains. Polycomb CBX proteins regulate gene expression by targeting Polycomb repressive complex 1 (PRC1) to sites of H3K27me3 via their chromodomains. UNC3866 binds the chromodomains of CBX4 and CBX7 most potently, with a K(d) of ∼100 nM for each, and is 6- to 18-fold selective as compared to seven other CBX and CDY chromodomains while being highly selective over >250 other protein targets. X-ray crystallography revealed that UNC3866's interactions with the CBX chromodomains closely mimic those of the methylated H3 tail. UNC4195, a biotinylated derivative of UNC3866, was used to demonstrate that UNC3866 engages intact PRC1 and that EED incorporation into PRC1 is isoform dependent in PC3 prostate cancer cells. Finally, UNC3866 inhibits PC3 cell proliferation, consistent with the known ability of CBX7 overexpression to confer a growth advantage, whereas UNC4219, a methylated negative control compound, has negligible effects.


Subject(s)
Oligopeptides/pharmacology , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , Animals , Biological Availability , Biotinylation , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Gene Expression Regulation/genetics , Humans , Isomerism , Ligases , Male , Methylation , Mice , Models, Molecular , Polycomb Repressive Complex 1/biosynthesis , Polycomb Repressive Complex 1/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Substrate Specificity , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
11.
Nat Chem Biol ; 11(8): 571-578, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26167872

ABSTRACT

The CEBPA gene is mutated in 9% of patients with acute myeloid leukemia (AML). Selective expression of a short (30-kDa) CCAAT-enhancer binding protein-α (C/EBPα) translational isoform, termed p30, represents the most common type of CEBPA mutation in AML. The molecular mechanisms underlying p30-mediated transformation remain incompletely understood. We show that C/EBPα p30, but not the normal p42 isoform, preferentially interacts with Wdr5, a key component of SET/MLL (SET-domain/mixed-lineage leukemia) histone-methyltransferase complexes. Accordingly, p30-bound genomic regions were enriched for MLL-dependent H3K4me3 marks. The p30-dependent increase in self-renewal and inhibition of myeloid differentiation required Wdr5, as downregulation of the latter inhibited proliferation and restored differentiation in p30-dependent AML models. OICR-9429 is a new small-molecule antagonist of the Wdr5-MLL interaction. This compound selectively inhibited proliferation and induced differentiation in p30-expressing human AML cells. Our data reveal the mechanism of p30-dependent transformation and establish the essential p30 cofactor Wdr5 as a therapeutic target in CEBPA-mutant AML.


Subject(s)
Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Dihydropyridines/pharmacology , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Myeloid-Lymphoid Leukemia Protein/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Amino Acid Sequence , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Molecular Docking Simulation , Molecular Sequence Data , Molecular Targeted Therapy , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Signal Transduction , Tumor Cells, Cultured
12.
Bioorg Med Chem ; 25(16): 4414-4423, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28662962

ABSTRACT

Given the high homology between the protein lysine methyltransferases G9a-like protein (GLP) and G9a, it has been challenging to develop potent and selective inhibitors for either enzyme. Recently, we reported two quinazoline compounds, MS0124 and MS012, as GLP selective inhibitors. To further investigate the structure-activity relationships (SAR) of the quinazoline scaffold, we designed and synthesized a range of analogs bearing different 2-amino substitutions and evaluated their inhibition potencies against both GLP and G9a. These studies led to the identification of two new GLP selective inhibitors, 13 (MS3748) and 17 (MS3745), with 59- and 65-fold higher potency for GLP over G9a, which were confirmed by isothermal titration calorimetry (ITC). Crystal structures of GLP and G9a in complex with 13 and 17 provide insight into the interactions of the inhibitors with both proteins. In addition, we generated GLP selective inhibitors bearing a quinoline core instead of the quinazoline core.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Quinazolines/pharmacology , Quinolines/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Histone-Lysine N-Methyltransferase/isolation & purification , Histone-Lysine N-Methyltransferase/metabolism , Humans , Models, Molecular , Molecular Structure , Quinazolines/chemical synthesis , Quinazolines/chemistry , Quinolines/chemical synthesis , Quinolines/chemistry , Structure-Activity Relationship
13.
Proc Natl Acad Sci U S A ; 111(35): 12853-8, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25136132

ABSTRACT

SET domain containing (lysine methyltransferase) 7 (SETD7) is implicated in multiple signaling and disease related pathways with a broad diversity of reported substrates. Here, we report the discovery of (R)-PFI-2-a first-in-class, potent (Ki (app) = 0.33 nM), selective, and cell-active inhibitor of the methyltransferase activity of human SETD7-and its 500-fold less active enantiomer, (S)-PFI-2. (R)-PFI-2 exhibits an unusual cofactor-dependent and substrate-competitive inhibitory mechanism by occupying the substrate peptide binding groove of SETD7, including the catalytic lysine-binding channel, and by making direct contact with the donor methyl group of the cofactor, S-adenosylmethionine. Chemoproteomics experiments using a biotinylated derivative of (R)-PFI-2 demonstrated dose-dependent competition for binding to endogenous SETD7 in MCF7 cells pretreated with (R)-PFI-2. In murine embryonic fibroblasts, (R)-PFI-2 treatment phenocopied the effects of Setd7 deficiency on Hippo pathway signaling, via modulation of the transcriptional coactivator Yes-associated protein (YAP) and regulation of YAP target genes. In confluent MCF7 cells, (R)-PFI-2 rapidly altered YAP localization, suggesting continuous and dynamic regulation of YAP by the methyltransferase activity of SETD7. These data establish (R)-PFI-2 and related compounds as a valuable tool-kit for the study of the diverse roles of SETD7 in cells and further validate protein methyltransferases as a druggable target class.


Subject(s)
Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Pyrrolidines/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , Tetrahydroisoquinolines/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Fibroblasts/drug effects , Hippo Signaling Pathway , Histone-Lysine N-Methyltransferase/genetics , Humans , MCF-7 Cells , Methyltransferases/antagonists & inhibitors , Methyltransferases/metabolism , Mutation , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Pyrrolidines/chemistry , Structure-Activity Relationship , Sulfonamides/chemistry , Tetrahydroisoquinolines/chemistry , Transcription Factors , YAP-Signaling Proteins
14.
J Biol Chem ; 290(22): 13641-53, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25825497

ABSTRACT

SMYD2 is a lysine methyltransferase that catalyzes the monomethylation of several protein substrates including p53. SMYD2 is overexpressed in a significant percentage of esophageal squamous primary carcinomas, and that overexpression correlates with poor patient survival. However, the mechanism(s) by which SMYD2 promotes oncogenesis is not understood. A small molecule probe for SMYD2 would allow for the pharmacological dissection of this biology. In this report, we disclose LLY-507, a cell-active, potent small molecule inhibitor of SMYD2. LLY-507 is >100-fold selective for SMYD2 over a broad range of methyltransferase and non-methyltransferase targets. A 1.63-Å resolution crystal structure of SMYD2 in complex with LLY-507 shows the inhibitor binding in the substrate peptide binding pocket. LLY-507 is active in cells as measured by reduction of SMYD2-induced monomethylation of p53 Lys(370) at submicromolar concentrations. We used LLY-507 to further test other potential roles of SMYD2. Mass spectrometry-based proteomics showed that cellular global histone methylation levels were not significantly affected by SMYD2 inhibition with LLY-507, and subcellular fractionation studies indicate that SMYD2 is primarily cytoplasmic, suggesting that SMYD2 targets a very small subset of histones at specific chromatin loci and/or non-histone substrates. Breast and liver cancers were identified through in silico data mining as tumor types that display amplification and/or overexpression of SMYD2. LLY-507 inhibited the proliferation of several esophageal, liver, and breast cancer cell lines in a dose-dependent manner. These findings suggest that LLY-507 serves as a valuable chemical probe to aid in the dissection of SMYD2 function in cancer and other biological processes.


Subject(s)
Antineoplastic Agents/chemistry , Benzamides/chemistry , Enzyme Inhibitors/chemistry , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Neoplasms/enzymology , Pyrrolidines/chemistry , Cell Line, Tumor , Cell Proliferation , Chromatin/chemistry , Computational Biology , Crystallization , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Epigenesis, Genetic , Histones/chemistry , Humans , Mass Spectrometry , Neoplasms/drug therapy , Peptides/chemistry , Protein Denaturation , Proteomics , Tumor Suppressor Protein p53/metabolism
16.
J Vet Med Educ ; 43(1): 13-20, 2016.
Article in English | MEDLINE | ID: mdl-26752020

ABSTRACT

Virtual microscopy using digitized slides has become more widespread in teaching in recent years. There have been no direct comparisons of the use of virtual microscopy and the use of microscopes and glass slides. Third-year veterinary students from two different schools completed a simple objective test, covering aspects of histology and histopathology, before and after a practical class covering relevant material presented as either glass slides viewed with a microscope or as digital slides. There was an overall improvement in performance by students at both veterinary schools using both practical formats. Neither format was consistently better than the other, and neither school consistently outperformed the other. In a comparison of student appraisal of use of digital slides and microscopes, the digital technology was identified as having many advantages.


Subject(s)
Computer-Assisted Instruction , Education, Veterinary/methods , Histology/education , Microscopy , Pathology/education
17.
Histopathology ; 67(2): 167-75, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25557923

ABSTRACT

AIMS: UK faecal occult blood test screening has tripled the proportion of pT1 colorectal cancers. The risk of metastasis is predicted by depth of invasion, suggesting that access to deep lymphovascular vessels is important. The aim of this study was to quantify the distribution and size of the submucosal vasculature, and generate a novel three-dimensional (3D) model to validate the findings. METHODS AND RESULTS: Thirty samples of normal large bowel wall were immunostained with CD31, a vascular endothelium marker, to identify blood vessels, which were quantified and digitally analysed for their number, circumference, area and diameter in the deep mucosa and submucosa (Sm1, Sm2, and Sm3). The model required serial sections, a double immunostain (using CD31 and D2-40), and 3D reconstruction. Significant differences were shown between submucosal layers in the number, circumference and area of vessels (P < 0.001). Blood vessels were most numerous in the mucosa (11.79 vessels/0.2 mm(2)) but smaller [median area of 247 µm(2) , interquartile range (IQR) 162-373 µm(2)] than in Sm2, where they were fewer in number (6.92 vessels/0.2 mm(2)) but considerably larger (2086 µm(2), IQR 1007-4784 µm(2)). The 3D model generated novel observations on lymphovascular structures. CONCLUSIONS: The number and size of blood vessels do not increase with depth of submucosa, as hypothesized. The distribution of vessels suggests that we should investigate the area or volume of submucosal invasion rather than the depth.


Subject(s)
Colorectal Neoplasms/blood supply , Colorectal Neoplasms/pathology , Neovascularization, Pathologic/pathology , Aged , Biomarkers, Tumor/metabolism , Computer Simulation , Early Detection of Cancer/methods , Endothelium, Vascular/metabolism , Female , Humans , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Intestinal Mucosa/pathology , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness , Neovascularization, Pathologic/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Risk Factors
18.
Nat Chem Biol ; 9(3): 184-91, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23292653

ABSTRACT

We describe the discovery of UNC1215, a potent and selective chemical probe for the methyllysine (Kme) reading function of L3MBTL3, a member of the malignant brain tumor (MBT) family of chromatin-interacting transcriptional repressors. UNC1215 binds L3MBTL3 with a K(d) of 120 nM, competitively displacing mono- or dimethyllysine-containing peptides, and is greater than 50-fold more potent toward L3MBTL3 than other members of the MBT family while also demonstrating selectivity against more than 200 other reader domains examined. X-ray crystallography identified a unique 2:2 polyvalent mode of interaction between UNC1215 and L3MBTL3. In cells, UNC1215 is nontoxic and directly binds L3MBTL3 via the Kme-binding pocket of the MBT domains. UNC1215 increases the cellular mobility of GFP-L3MBTL3 fusion proteins, and point mutants that disrupt the Kme-binding function of GFP-L3MBTL3 phenocopy the effects of UNC1215 on localization. Finally, UNC1215 was used to reveal a new Kme-dependent interaction of L3MBTL3 with BCLAF1, a protein implicated in DNA damage repair and apoptosis.


Subject(s)
Benzamides/pharmacology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Drug Discovery , Lysine/analogs & derivatives , Molecular Probes/pharmacology , Piperidines/pharmacology , Benzamides/chemistry , Benzamides/metabolism , Binding, Competitive/drug effects , Crystallography, X-Ray , DNA-Binding Proteins/antagonists & inhibitors , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Lysine/antagonists & inhibitors , Lysine/chemistry , Lysine/metabolism , Models, Molecular , Molecular Probes/chemistry , Molecular Probes/metabolism , Molecular Structure , Piperidines/chemistry , Piperidines/metabolism , Protein Structure, Tertiary , Repressor Proteins/metabolism , Structure-Activity Relationship , Tumor Suppressor Proteins/metabolism
19.
Angew Chem Int Ed Engl ; 54(13): 3927-31, 2015 Mar 23.
Article in English | MEDLINE | ID: mdl-25650139

ABSTRACT

Methods for the rapid and inexpensive discovery of hit compounds are essential for pharmaceutical research and DNA-encoded chemical libraries represent promising tools for this purpose. We here report on the design and synthesis of DAL-100K, a DNA-encoded chemical library containing 103 200 structurally compact compounds. Affinity screening experiments and DNA-sequencing analysis provided ligands with nanomolar affinities to several proteins, including prostate-specific membrane antigen and tankyrase 1. Correlations of sequence counts with binding affinities and potencies of enzyme inhibition were observed and enabled the identification of structural features critical for activity. These results indicate that libraries of this type represent a useful source of small-molecule binders for target proteins of pharmaceutical interest and information on structural features important for binding.


Subject(s)
DNA Probes/chemical synthesis , DNA/chemistry , DNA Fingerprinting , DNA Probes/chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , Ligands , Prostate-Specific Antigen/drug effects , Serum Albumin/chemistry , Small Molecule Libraries , Structure-Activity Relationship , Tankyrases/antagonists & inhibitors
20.
Angew Chem Int Ed Engl ; 54(17): 5166-70, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25728001

ABSTRACT

PRMT3 catalyzes the asymmetric dimethylation of arginine residues of various proteins. It is essential for maturation of ribosomes, may have a role in lipogenesis, and is implicated in several diseases. A potent, selective, and cell-active PRMT3 inhibitor would be a valuable tool for further investigating PRMT3 biology. Here we report the discovery of the first PRMT3 chemical probe, SGC707, by structure-based optimization of the allosteric PRMT3 inhibitors we reported previously, and thorough characterization of this probe in biochemical, biophysical, and cellular assays. SGC707 is a potent PRMT3 inhibitor (IC50 =31±2 nM, KD =53±2 nM) with outstanding selectivity (selective against 31 other methyltransferases and more than 250 non-epigenetic targets). The mechanism of action studies and crystal structure of the PRMT3-SGC707 complex confirm the allosteric inhibition mode. Importantly, SGC707 engages PRMT3 and potently inhibits its methyltransferase activity in cells. It is also bioavailable and suitable for animal studies. This well-characterized chemical probe is an excellent tool to further study the role of PRMT3 in health and disease.


Subject(s)
Enzyme Inhibitors/chemistry , Isoquinolines/chemistry , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Allosteric Regulation , Binding Sites , Calorimetry , Cell Line, Tumor , Enzyme Inhibitors/metabolism , HEK293 Cells , Histones , Humans , Isoquinolines/metabolism , Methylation , Molecular Dynamics Simulation , Mutagenesis , Protein Binding , Protein Structure, Tertiary , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL