New support for structure-guided drug discovery coalition research on tropical diseases
Toronto, ON - Almost $2 million is being invested by the Bill & Melinda Gates Foundation to help fight major parasitic diseases of the developing world.
This probe is available from Sigma and Cayman Chemical.
The negative control GSK8573 is available from Sigma.
Probe | Negative control | |
![]() |
| ![]() |
GSK2801 |
| GSK8573 |
Biology of the BAZ2 Bromodomains
BAZ2A/B belong to a family of ubiquitously expressed bromodomain containing proteins. Proteins of the BAZ (bromodomain adjacent zinc finger) family are characterized by a carboxy-terminal bromodomain adjacent to a PHD finger and a WACZ motif. In addition four other conserved motifs are typically found in the N-terminus of BAZ family members, namely the LH motif (a leucine-rich helical domain), the ZB2 motif (conserved between ZK783.4 and BAZ2 proteins) and the BAZ 1 and BAZ 2 motifs (conserved between all BAZ proteins, Acf, and ZK783.4).
Little is known about the biological function of BAZ2B but it has been suggested to regulate nucleosome mobilization by the ATP-dependent chromatin remodelling factor ISWI. Interaction of BAZ2B with ISWI is mediated by the BAZ1 motif [1]. The BAZ2B locus has also been identified as associated with sudden cardiac death [2]. The related BAZ2A (also called TIP5) protein plays a central role in the NoRC (nucleolar remodelling complex). BAZ2A is essential for heterochromatin formation leading to transcriptional silencing of certain rRNAs [3].
GSK2801: A Chemical Probe for BAZ2A/B
GSK2801 a BAZ2A/B selective inhibitor is the result of a collaboration with GSK [4]. A second probe against this target, BAZ2-ICR, has also been reported [5].
An inactive control GSK8573 has also been reported (see properties tab).
Co-crystal structure
The co-crystal structure of GK2801 and BAZ2B has been solved (pdb id: 4RVR).
Potency Against Target Family
GSK2801 has been shown by ITC to bind to BAZ2A with a KD of 257 nM and to BAZ2B with a KD of 136 nM.
Selectivity
GSK2801 was screened at 10 μM concentration against 47 bromodomains using a thermal shift assay. Moderate activity was observed against TAF1L and BRD9 but GSK2801 was otherwise selective. The screened targets are labelled on the phylogenetic tree; targets that have not been screened are shown in grey. Temperature shifts are represented as spheres as indicated in the figure.
Selectivity Beyond Target Family
GSK2801 was screened in a binding assay against 55 receptors and ion channels and was found to be inactive against all but the Melatonin (MT-1) receptor.
Cellular Activity
GSK2801 accelerated FRAP half-recovery time to the same extent as observed for the mutant construct indicating that the compound was able to displace BAZ2A from chromatin FRAP assay.
GSK2801 |
![]() |
1-(1-(2-(methylsulfonyl)phenyl)-7-propoxyindolizin-3-yl)ethan-1-one |
Physical and chemical properties | |
---|---|
Molecular weight | 371.45 |
Molecular formula | C20H21NO4S |
IUPAC name | 1-(1-(2-(methylsulfonyl)phenyl)-7-propoxyindolizin-3-yl)ethan-1-one |
logP | 3.54 |
PSA | 63.68 |
No. of chiral centres | 0 |
No. of rotatable bonds | 6 |
No. of hydrogen bond acceptors | 5 |
No. of hydrogen bond donors | 0 |
Storage | Stable as solid in the dark at -20°C. NB making aliquots rather than freeze-thawing is recommended |
Solubility | Soluble in DMSO at least up to 50uM |
SMILES: CCCOC1=CC2=C(C3=C(S(=O)(C)=O)C=CC=C3)C=C(C(C)=O)N2C=C1
InChI: InChI=1S/C20H21NO4S/c1-4-11-25-15-9-10-21-18(14(2)22)13-17(19(21)12-15)16-7-5-6-8-20(16)26(3,23)24/h5-10,12-13H,4,11H2,1-3H3
InChI Key: KHWCPNJRJCNVRI-UHFFFAOYSA-N
The negative control GSK8573 has the following properties |
![]() |
1-(1-(3-methoxyphenyl)-7-propoxyindolizin-3-yl)ethan-1-one For SDF click here |
Physical and chemical properties | |
---|---|
Molecular weight | 323.39 |
Molecular formula | C20H21NO3 |
IUPAC name | 1-(1-(3-methoxyphenyl)-7-propoxyindolizin-3-yl)ethan-1-one |
logP | 3.10 |
PSA | 38.77 |
No. of chiral centres | 0 |
No. of rotatable bonds | 6 |
No. of hydrogen bond acceptors | 4 |
No. of hydrogen bond donors | 0 |
Storage | Stable as solid in the dark at -20°C. NB making aliquots rather than freeze-thawing is recommended |
Solubility | Soluble in DMSA at least up to to 50uM |
Potency against Target
GSK2801 has been shown by ITC to bind to BAZ2A with a KD of 257 nM and to BAZ2B with a KD of 136 nM. The ΔH for binding to BAZ2B is -10.69 (+/- 0.07) kcal/mol.
Selectivity Within Target family
GSK2801 showed significant thermal shifts of 4.1 and 2.7 °C for BAZ2A and BAZ2B respectively. GSK2801 did not show significant thermal shifts against all other bromodomains, except for BRD9 (ΔTm: 2.9 °C) and TAF1L (ΔTm: 3.4 °C) was observed. The KD of GSK2801 binding to BRD9 and TAF1L were determined to be 1.2 and 3.2 μM respectively by ITC.
Because of the limited sensitivity of BAZ2A/B in the thermal stability assay, the selectivity of GSK2801 was measured by BLI. Forty-two bromodomains modified with a Bir A biotin ligase targeting sequence were co-expressed in bacteria with the enzyme. The biotinylated proteins were used in BLI experiments probing the interaction of GSK2801 at two concentrations (0.2 and 1.0 μM). In agreement with the ΔTm data, BRD9 and TAF1(L) were detected as the major off-targets, while no other significant interactions were detected within the bromodomain family.
The structurally related GSK8573 proved to be inactive against BAZ2A/B and all other bromodomains except BRD9 in the BLI assay
Selectivity Beyond Target Family
GSK2801 (10 µM) was screened in a binding assay against 55 receptors and ion channels and was found to be inactive against all but the Melatonin (MT-1) receptor where inhibition was 4% of the control.
FRAP
To investigate whether GSK2801 can displace BAZ2 bromodomains from chromatin in living cells, we performed a fluorescence recovery after photobleaching (FRAP) assay utilizing GFP-tagged BAZ2A full length protein transfected into human osteosarcoma cells (U2OS). Mutant BAZ2A (N1873F) was used as a control that does not bind acetylated lysine containing peptides and therefore mirrors the behaviour of inhibitor bound BAZ2A. The histone deacetylase (HDAC) inhibitor suberoylanilide hydroxamic acid (SAHA) was used to increase overall levels of histone acetylation, resulting in a sufficient assay window to enable the measurement of differences in recovery time and demonstrating the acetylation dependence of the FRAP experiments. GSK2801 at 1 μM GSK2801 accelerated FRAP half-recovery time to the same extent as observed for the mutant construct indicating that the compound was able to displace BAZ2A from chromatin. Conversely, the inactive GSK-8573 did not have any effect on the half-recovery time of GFP-BAZ2A
Cytotoxicity
GSK2801 showed no significant toxicity against U02S or HeLa cells
A Novel Family of Bromodomain Genes,
Jones, M. H.; Hamana, N.; Nezu, Ji.; Shimane, M., Genomics, 63, 40– 45 (2000)
Arking, D. E.; Junttila, M. J.; Goyette, P.; Huertas-Vazquez, A.; Eijgelsheim, M.; Blom, M. T.; Newton-Cheh, C.; Reinier, K.; Teodorescu, C.; Uy-Evanado, A.; Carter-Monroe, N.; Kaikkonen, K. S.; Kortelainen, M. L.; Boucher, G.; Lagace, C.; Moes, A.; Zhao, X.; Kolodgie, F.; Rivadeneira, F.; Hofman, A.; Witteman, J. C.; Uitterlinden, A. G.; Marsman, R. F.; Pazoki, R.; Bardai, A.; Koster, R. W.; Dehghan, A.; Hwang, S. J.; Bhatnagar, P.; Post, W.; Hilton, G.; Prineas, R. J.; Li, M.; Kottgen, A.; Ehret, G.; Boerwinkle, E.; Coresh, J.; Kao, W. H.; Psaty, B. M.; Tomaselli, G. F.; Sotoodehnia, N.; Siscovick, D. S.; Burke, G. L.; Marban, E.; Spooner, P. M.; Cupples, L. A.; Jui, J.; Gunson, K.; Kesaniemi, Y. A.; Wilde, A. A.; Tardif, J. C.; O’Donnell, C. J.; Bezzina, C. R.; Virmani, R.; Stricker, B. H.; Tan, H. L.; Albert, C. M.; Chakravarti, A.; Rioux, J. D.; Huikuri, H. V.; Chugh, S. S., PLoS Genet., 7, e1002158 (2011)
Guetg, C.; Lienemann, P.; Sirri, V.; Grummt, I.; Hernandez‐Verdun, D.; Hottiger, M.O.; Fussenegger, M.; Santoro, R., The NoRC complex mediates the heterochromatin formation and stability of silent rRNA genes and centromeric repeats, Embo J., 29, 2135-2146, (2010)
The pharmacokinetic parameters GSK2801 for in vivo experiments were measured after intraperitoneal and oral dosing in male CD1 mice. This PK study showed that GSK2801 has reasonable in vivo exposure after oral dosing, modest clearance, and reasonable plasma stability. These properties should allow GSK2801 to be used as a BAZ2A/B bromodomain inhibitor in vivo.
IP 30 mg/kg | PO 30 mg/kg | |
Tmax (h) | 0.25 | 1.0 |
Cmax (ng/mL) | 1670 | 435 |
Terminal t½ (h) | 4.5 | 1.5 |
The co-crystal structure of GSK2801 and BAZ2B has been solved (pdb id: 4RVR).
Isothermal Titration Calorimetry
Experiments were carried out on a VP-ITC titration microcalorimeter from MicroCalTM, LLC (Northampton, MA), at 15 °C while stirring at 295 rpm, in ITC buffer (50 mM HEPES, 150 mM NaCl). All titrations were conducted using an initial injection of 2 µl followed by 34 identical injections of 8 µl with a duration of 16 sec/injection and a spacing of 250 sec between injections. The heat of dilution was determined by independent titrations (protein into buffer) and was subtracted from the experimental data. MicroCalTM Origin software was used to calculate enthalpies of binding (ΔH) and binding constants (KB). Thermodynamic parameters were calculated (ΔG = ΔH - TΔS = -RTlnKB, where ΔG, ΔH and ΔS are the changes in free energy, enthalpy and entropy of binding respectively). In all cases a single binding site model was employed.
Differential Scanning Fluorimetry (DSF)
Thermal melting experiments were carried out using an Mx3005p Real Time PCR machine (Stratagene). Proteins were buffered (10 mM HEPES, 500 mM NaCl) and assayed at a final concentration of 2 µM. Compounds were added at a final concentration of 10 µM. SYPRO Orange (Molecular Probes) was added as a fluorescence probe at a dilution of 1:1000. Excitation and emission filters were set to 465 nm and 590 nm respectively. The temperature was raised with a step of 3 °C per minute from 25 °C to 96 °C and fluorescence readings were taken at each interval. Data was analysed as previously reported [6]
Biolayer interference assays
BAZ2A and BAZ2B were expressed in frame with a C-terminal Avi tag and a His-TEV tag enabling enzymatic conjugation of a single biotin using the biotin ligase (BirA) which had been co-expressed in E. coli. Cells grown in the presence of 0.1 mM biotin were lysed and purified using a similar procedure. Incorporation of biotin was confirmed by ESI-MS spectroscopy. The biotin labelled BAZ2B and BAZ2A was immobilized on SuperStreptavidin BLI sensors. BioLayer Interferometry (BLI) experiments were performed on a 16-channel ForteBio Octet RED384 instrument at 25°C (25 mM HEPES, pH 7.5, 0.05% Tween and 100 mM NaCl buffer). The reference sensors without attached bromodomain were used to subtract background binding to SuperStreptavidin sensors. Data were processed and analysed using ForteBio Analysis software.
CEREP assay
GSK2801 (10 µm) was screened against a panel of 55 ligand receptors, ion channels and transports using an established and widely utilized commercial assay platform (ExpresSProfile; CEREP, Paris, FRANCE).
Fluorescence Recovery After Photobleaching (FRAP) Assay
FRAP studies were performed using U20S cells expressing full-length BAZ2A or BRD4 protein fused with an N-terminal eGFP as previously described [4]. Six hours after transfection 2.5uM SAHA (to increase global histone acetylation) was added and GSK2801 was added 1 hour before imaging. Imaging was carried out 24 hours after transfection. Percent inhibition was calculated between the DMSO treated (0%) and N1873F expressing mutant (100%)
Cytotoxicity Assay
U20S cells were harvested from exponential phase cultures and plated in a 96-well opaque flat bottomed plates at a cell density of 3 x 103 cells/well (100uL). Compounds were dissolved in DMSO at 10 uM and serial dilutions performed. 5 uL of compound solution was added to each well and incubated for 24 or 72 hours at 37˚C in a humidified atmosphere containing CO2 (5%). 10 uL of WST-1 (Roche) was added and the plates returned to the incubator. Plates were read on a plate reader at 450 nm after 2 h for cells treated with GSK2801 for 24 h. Results plotted as % of DMSO control.
Pharmacokinetics
These PK studies were performed by Shanghai ChemPartner Co. Ltd. The IP and PO dosing solutions were prepared in 0.5% CMC+1% Tween 80. No clinical symptoms were observed during the entire in-life study. Male CD-1 mice, 28-32g were used (with free access to food and water). A dose of 30 mg/kg (10 mL/kg) was used via intraperitoneal injection (N=9) and for oral dosing 30 mg/kg (10 mL/kg) via gavage (N=9). The animals were restrained manually for blood collection and approx. 110 μL blood/time point was collected, centrifuged at 4°C (2000 g, 5min) to obtain plasma within 15 min after sample collection. Plasma samples were stored at -70℃ until analysis. An UPLC/MS-MS-002 (API-4000) was used for sample analysis, with Dexamethasone as internal standard. An aliquot of 20 μL sample was added with 200 μL ACN containing 50 ng/mL dexamethasone. The mixture was vortexed for 2 min and centrifuged at 14000 rpm for 5 min. An aliquot of 1 μL supernatant was injected for LC-MS/MS analysis.
Toronto, ON - Almost $2 million is being invested by the Bill & Melinda Gates Foundation to help fight major parasitic diseases of the developing world.
The SGC-CHDI open access patent-free partnership for Huntington’s disease research has been selected as a featured Innovator to present on November 17th at the 6th annual
On Friday August 22nd, 2014, sixteen members of SGC at University of Toronto completed the ALS Ice Bucket Challenge outside the MaRS building (video here). As part of the challenge, SGC's Director, Aled Edwards nominated Takeda's General Manager Tetsu Maruyama.
ChemNET trainees on their final day at SGC-Toronto (L to R): Zafar Qureshi, Venugopal R
The probe is available from Sigma and Cayman Chemical.
Probe |
![]() |
OF-1 |
A chemical probe for the bromodomains of the BRPF (BRomodomain and PHD Finger containing) family of proteins (BRPF1/2/3) has been discovered by the SGC.
BRPF1, BRPF2 (BRD1) and BRPF3 are scaffolding proteins, assembling HAT complexes of the MOZ/MORF family (MOZ, Ybf2/Sas3, Sas2, and Tip60) (1). These MYST complexes have a tetrameric core containing BRPF, the tumour suppressor ING and Eaf6/EPC (enhancer of polycomb)-related scaffold subunits. MYST complexes play crucial roles in DNA repair, recombination, and replication as well as in transcription activation (2,3). MOZ is frequently translocated in AML (acute myeloid leukemia) and is required for HSC proliferation (4).
Two BRPF1 isoforms (isoform A and B) can be generated by alternative splicing. In contrast to BRPF1B, the isoform A harbours a residue insertion in the ZA-loop that prevents binding to acetylated histone peptides (5).
Phylogenetic tree of bromodomains and detailed view at the BRPF family.
OF-1 has been shown to bind to BRPF1B with a KD of 100 nM (ITC), to BRPF2 with a KD of 500 nM (ITC) and to BRPF3 with a KD of 2.4 µM (ITC). Selectivity against other BRDs is very good, in general >100-fold. The closest off-target effects are against BRD4 (39-fold selectivity) and TIF1α (50% inhibition at 20 µM in the alpha screen). OF-1 increases thermal stability in the cellular thermal shift assay (CETSA) of full length BRPF1B at 1 µM and also demonstrates accelerated FRAP recovery at 5 µM in the BRPF2 FRAP assay. It shows modest general cytotoxicity.
Bromodomain | Kd/nM (ITC) | IC50/nM (Alpha Screen) | TM Shift °C |
BRPF1B | 100 | 270 | 6.5±0.4 |
BRPF2 (BRD1) | 500 | 2200 | 3.1±0.3 |
BRPF3 | 2400 | ND | 3.3±0.2 |
BRD4 (1) | 4000 | >10,000 | 2.0±0.2 |
OF-1 is a chemical probe with potent binding affinity for BRPF1 (KD of 100nM), BRPF2 (KD of 500 nM) and BRPF3 (KD of 2400 nM) as determined by isothermal titration calorimetry (ITC). Alpha screen biochemical assay confirmed OF-1 as a potent inhibitor of BRPF1 (IC50 of 270 nM).
OF-1 induced significant ΔTm shifts within the BRPF family. Weak interactions (2.1 °C) were also observed for BRD4(1), however alpha screen did not reveal strong interactions of OF-1 with BRD4 (IC50 of >10000 nM) and a KD of 4000nM for BRD4(1) reveals 39-fold selectivity when compared to BRPF1B isoform.
OF-1 shows no significant inhibition of protein kinases (<20% inhibition at 10 μM for 40 kinases screened).
We recommend to use the BRPF inhibitor NI-57 in parallel to confirm the result. Please do not apply OF-1 higher than 5 μM to avoid effects on BRD4 and verify the result by using NI-57.
OF-1 increases thermal stability in the CETSA of full length BRPF1B at 1 µM.
In a NanoBRETTM assay, BRPF1B, but not BRPF1A isoform shows dose-dependent displacement from histone H3.3, with IC50 of 0.08 μM and the FRAP assay reveals strong inhibition of BRPF2 (BRD1) at 5 µM concentration of OF-1.
OF-1 attenuates RANKL/MCSF induced differentiation of primary human monocytes into osteoclasts.
Probe |
![]() |
OF-1 |
Physical and chemical properties | |
---|---|
Molecular weight | 440.31 |
Molecular formula | C17H18BrN3O4S |
IUPAC name | 4-bromo-N-(6-methoxy-1,3-dimethyl-2-oxo-2,3-dihydro-1H-benzo[d]imidazol-5-yl)-2-methylbenzenesulfonamide |
clogP | 2.55 |
PSA | 78.95 |
No. of chiral centres | 0 |
No. of rotatable bonds | 4 |
No. of hydrogen bond acceptors | 4 |
No. of hydrogen bond donors | 1 |
Storage | Stable as solid in the dark at -20°C. NB making aliquots rather than freeze-thawing is recommended |
Dissolution | Soluble in DMSO |
Temperature Shift Assay
Selectivity screening of chemical probe OF-1 determined by temperature shift assay.
The temperature shifts mapped onto the phylogenetic tree using red circles corresponding to ΔTm as indicated in the figure.
CESTA
Thermal-stability of BRPF1A/B determined by CESTA at 1 µM OF-1.
NanoBRET
Dose-dependent displacement of BRPF1A/B from histone H3.3 following treatment with OF-1.
FRAP Assay
Half-times of fluorescence recovery (t1/2) after photo bleaching measured for BRPF2 (BRD1) after treatment with OF-1 at different concentrations with or without SAHA.
TRAP staining for osteoclasts and measurement of pits depth.
Work on this probe has been published in 'Selective targeting of Bromodomain-PHD fingers family (BRPF) bromodomains impairs osteoclast differentiation'.
Isothermal Titration Calorimetry (ITC)
All calorimetric experiments were performed on a VP-ITC micro-calorimeter (MicroCalTM, LLC Northampton, MA). Protein solutions were buffer exchanged by gel filtration or dialysis into buffer (20 mM Hepes pH 7.5, 150 mM NaCl, and 0.5 mM tris (2-carboxyethyl) phosphine (TCEP). All measurements were carried out at 288.15 K. All injections were performed using an initial injection of 2 µL followed by injections of 8 µL. The data were analysed with the MicroCal ORIGIN software package employing a single binding site model. The first data point was excluded from the analysis.
Temperature shift assay
Thermal melting experiments were carried out using a Stratagene Mx3005p Real Time PCR machine (Agilent Technologies). OF-1 was added at a final concentration of 10 µM. SYPRO Orange (Molecular Probes) was added as a fluorescence probe at a dilution of 1:1000 as described (6).
AlphaScreen Assay
Assays were performed as described previously with minor modifications (7). All reagents were diluted in 25 mM HEPES, 100 mM NaCl, 0.1 % BSA, pH 7.4 supplemented with 0.05 % CHAPS and allowed to equilibrate to room temperature prior to addition to plates. An 11-point 1:2.0 serial dilutions of the ligands was prepared on lowvolume 384-well plates (ProxiPlateTM-384 Plus, PerkinElmer, USA), using LabCyte Eco liquid handler. Plates filled with 5 µL of the assay buffer followed by 7 µL of biotinylated peptide [H-YSGRGKacGGKacGLGKacGGAKacRHRK(Biotin)-OH for BRD1, BRD4, BRPF1B and BRPF3 or YQTARKSTGGK(ac)APRKQLATKAK(biotin)-OH for TIF1α] and Histagged protein to achieve final assay concentrations of 25-100 nM depending on the dose-response curve for each individual protein. Plates were sealed and incubated for a further 30 minutes, before the addition of 8 µM of the mixture of streptavidin-coated donor beads (12.5 µg/mL) and nickel chelate acceptor beads (12.5 µg/mL) under low light conditions. Plates were foil-sealed to protect from light, incubated at room temperature for 60 minutes and read on a PHERAstar FS plate reader (BMG Labtech, Germany) using an AlphaScreen 680 excitation/570 emission filter set. IC50 values were calculated in Prism 5 (GraphPad Software, USA) after normalization against corresponding DMSO controls.
Fluorescence Recovery After Photobleaching (FRAP) Assay
FRAP studies were performed using U20S cells expressing full-length BRPF2 (BRD1). Six hours after transfection 2.5 µM SAHA (to increase global histone acetylation) was added and cells were treated with 1 µM or 5 µM of OF-1 1 hour before imaging and half recovery times from the fluorescence signal of the bleached U2OS nuclei were plotted.
NanoBRET
U2OS cells were co-transfected with Histone H3.3-HaloTag and NanoLuc-BRPF1. Twenty hours post-transfection cells were collected, washed with PBS, and exchanged into media containing phenol red-free DMEM and 4% FBS in the absence (control sample) or the presence (experimental sample) of 100 nM NanoBRET 618 fluorescent ligand (Promega). Cells were then treated with an increasing dose of OF-1. Five minutes prior to reading, NanoBRET furimazine substrate (Promega) was added to both control and experimental samples and plates were read on a CLARIOstar (BMG) equipped with 450/80 nm bandpass and 610 nm longpass filters with a 0.5 sec reading setting. A corrected BRET ratio was calculated and is defined as the ratio of the emission at 610 nm/450 nm for experimental samples (i.e. those treated with NanoBRET fluorescent ligand) subtracted by and the emission at 610 nm/450 nm for control samples (not treated with NanoBRET fluorescent ligand). BRET ratios are expressed as milliBRET units (mBU), where 1 mBU corresponds to the corrected BRET ratio multiplied by 1000. Relative IC50 values were estimated by non-linear regression analysis of (log) concentration of each inhibitor versus milliBRET ratios (GraphPad Prism).
Human osteoclast differentiation
Primary human peripheral blood mononuclear cells (PBMCs) were collected from a Histopaque generated buffy coat after gradient centrifugation at 20 min and 500g, brakes off. The CD14+ monocyte fraction was obtained by on-column CD14+-MACS bead isolation, washed twice with MACS buffer, and seeded at a density of 50 000 c/mL in αMEM/10%FCS supplemented with 25 ng/mL MCSF. After 6 days at 37 °C, 5% CO2 treatment with OF-1 with and without 50 ng/mL RANKL was started. Media were changed with fresh compound every 3−4 days. After 14−21 days, cells were fixed and stained for TRAP.
Bone Resorption Assays.
PBMCs were isolated and seeded onto self-cut dentine slices from ivory. After 14 days of differentiation, cells were removed from dentine slices. Dentine pits were imaged with confocal microscopy.
Toronto, Canada & New York City, USA — The Structural Genomics Consortium (SGC) and CHDI Foundation have entered into a unique open-access research collaboration to discover and characterize new drug targets for Huntington’s disease (HD) using structural and chemical biology.
SGC707 is available from Cayman Chemical and Sigma and from Tocris
XY1 (the negative control of SGC 707) is available from Tocris and from Cayman Chemical.
Probe | Negative control | |
![]() |
| ![]() |
SGC707 (IC50=31 nM) |
| XY1 (IC50>100,000 nM) |
A collaboration between the SGC, China Novartis Institutes for Biomedical Research, and the center for Integrative Chemical Biology and Drug Discovery (CICBDD) at the University of North Carolina has resulted in the discovery of SGC707, a chemical probe for PRMT3, a protein which catalyzes the asymmetrical di-methylation of arginine residues. SGC707 is a potent allosteric inhibitor of PRMT3 (IC50 = 50 nM) with >100-fold selectivity over other methyltransferases, and other non-epigenetic targets. SGC707 has been shown to bind to PRMT3 with KD of 50 nM (ITC), and inhibits the methylation of histones in cells with IC50 value below 1 µM.
Probe | Negative control | |
![]() |
| ![]() |
SGC707 (IC50=31 nM) |
| XY1 (IC50>100,000 nM) |
Click here to download SDF file
Physical and chemical properties | |
---|---|
Molecular weight | 298.1430 |
Molecular formula | C16H18N4O2 |
IUPAC name | 2-((3-aza-bicyclo[4.4.0]deca-1,3,5,7,9-pentaen-8-ylamino)-formylamino)-1-(pyrrolidin-1-yl)-ethanone |
logP | 1.8 |
PSA | 59.9 A |
(C) Effect of SGC707 on the activity of 27 protein methyltransferases as well as DNMT1, DNMT3A-3L, DNMT3B-3L and BCDIN3D (an RNA-methyltransferase) were assessed.
(D) IC50 values were determined at saturating concentration of peptide substrate (1.5 µM) and SAM concentrations equal to 0.25, 0.5, 1, 2, 4, 8, 10 and 12x of the Km for SAM. No change in IC50 values at varying SAM concentrations was consistent with a noncompetitive pattern of inhibition with respect to SAM. (E) To determine the competition with peptide, the SAM concentration was kept at saturation (70 µM) and IC50 values were determined at different peptide concentrations (0.5, 1, 2, 4, 8, 12, 16, and 20 ×Km). A similar noncompetitive pattern was observed for SGC707 with respect to peptide confirming the allosteric mode of inhibition.
Western blot analysis of H4R3me2a levels. HEK293 cells were co-transfected with FLAG tagged PRMT3 (WT) or its catalytically dead mutant E335Q (Mut) and treated with different concentrations of SGC707, as indicated. Total cell lysates were collected 24 h post inhibitor treatment and analysed for H4R3me2a levels. The total levels of exogenous histone H4 and overexpressed PRMT3 were determined with anti-GFP, anti-H4 and anti-FLAG antibodies, respectively
Kaniskan, H. Ü., Szewczyk, M. M., Yu, Z., Eram, M. S., Yang, X., Schmidt, K., Luo, X., Dai, M., He, F., Zang, I., Lin, Y., Kennedy, S., Li, F., Dobrovetsky, E., Dong, A., Smil, D., Min, S.-J., Landon, M., Lin-Jones, J., Huang, X.-P., Roth, B. L., Schapira, M., Atadja, P., Barsyte-Lovejoy, D., Arrowsmith, C. H., Brown, P. J., Zhao, K., Jin, J. and Vedadi, M. (2015), A Potent, Selective and Cell-Active Allosteric Inhibitor of Protein Arginine Methyltransferase 3 (PRMT3).
Angew. Chem.. doi: 10.1002/ange.201412154
The Group led by Professor Chas Bountra, Chief Scientist of SGC Oxford, has been awarded the Royal Society of Chemistry's Rita and John Cornforth Award 2014 for "... world leading collaborative research across the disciplines of structural biology, medicinal chemistry, chemical biology and enzymology towards understanding and exploiting the potential of epigenetics as a target family for future drug discovery".
This probe is available from Cayman Chemical , Tocris and Sigma-Aldrich.
The control is available from Cayman Chemical.
Probe | Negative control | |
![]() |
| ![]() |
OICR-9429 |
| OICR-0547 |
A collaboration between the SGC and The Drug Discovery Program at the Ontario Institute for Cancer Research (OICR) has resulted in the discovery of OICR-9429, a chemical probe for WDR5, a component of several chromatin regulatory complexes including the MLL1 (Mixed Lineage Leukemia 1) complex. OICR-9429 is a potent antagonist of the interaction of WDR5 with peptide regions of MLL and Histone 3 with >100-fold selectivity over other chromatin “reader” domains, methyltransferases, and other non-epigenetic targets. OICR-9429 has been shown to bind to WDR5 with KD values of 24 nM (Biacore) and 52 nM (ITC). OICR-9429 also disrupts the interaction of WDR5 with MLL1 and RbBP5 in cells with IC50 values below 1 µM.
A closely-related compound, OICR-0547, does not show WDR5 antagonism, and can therefore be used as a negative control in phenotypic assays.
Probe | Negative control | |
![]() |
| ![]() |
OICR-9429 |
| OICR-0547 |
Physical and chemical properties for OICR-9429 | |
Molecular weight | 555.2 |
Molecular formula | C29H32F3N5O3 |
IUPAC name | 5-((2-(4-methyl-piperazin-1-yl)-5-(3-((morpholin-4-yl)-methyl)-phenyl)-phenylamino)-formyl)-4-(trifluoro-methyl)-1,2-dihydro-pyridin-2-one |
MollogP | 3.382 |
PSA | 65.34 |
No. of chiral centres | 0 |
No. of rotatable bonds | 8 |
No. of hydrogen bond acceptors | 7 |
No. of hydrogen bond donors | 2 |
Physical and chemical properties for OICR-0547 (Negative Control) | |
Molecular weight | 542.2 |
Molecular formula | C28H29F3N4O4 |
IUPAC name | 5-((2-(morpholin-4-yl)-5-(3-((morpholin-4-yl)-methyl)-phenyl)-phenylamino)-formyl)-4-(trifluoro-methyl)-1,2-dihydro-pyridin-2-one |
MollogP | 3.331 |
PSA | 69.77 |
No. of chiral centres | 0 |
No. of rotatable bonds | 8 |
No. of hydrogen bond acceptors | 7 |
No. of hydrogen bond donors | 2 |
Pharmacological targeting of the Wdr5-MLL interaction in C/EBPa N-terminal leukemia ,Florian Grebien, Masoud Vedadi, Matthäus Getlik, Roberto Giambruno, Amit Grover, Roberto Avellino, Anna Skucha, Sarah Vittori, Ekaterina Kuznetsova, David Smil, Dalia Barsyte-Lovejoy, Fengling Li, Gennadiy Poda, Matthieu Schapira, Hong Wu, Aiping Dong, Guillermo Senisterra, Alexey Stukalov, Kilian V M Huber,Andreas Schönegger, Richard Marcellus, Martin Bilban, Christoph Bock, Peter J Brown, Johannes Zuber, Keiryn L Bennett, Rima Al-awar, Ruud Delwel, Claus Nerlov, Cheryl H Arrowsmith & Giulio Superti-Furga. Nature Chemical Biology. 10.1038/nchembio.1859
Main features