scholarly journals Discovery of small molecule antagonists of human Retinoblastoma Binding Protein 4 (RBBP4)

2022 ◽  
Author(s):  
Sumera Perveen ◽  
Carlos A Zepeda-Velazquez ◽  
David McLeod ◽  
Richard Marcellus ◽  
Mohammed Mohammed ◽  
...  

RBBP4 is a nuclear WD40 motif-containing protein widely implicated in various cancers and a putative drug target. It interacts with multiple proteins within diverse complexes such as nucleosome remodeling and deacetylase (NuRD) complex and polycomb repressive complex 2 (PRC2), as well as histone H3 and H4 through two distinct binding sites. B-cell lymphoma/leukemia 11A (BCL11A), friend of GATA-1 (FOG-1), plant homeodomain finger protein 6 (PHF6) and histone H3 bind to the top of the donut-shaped seven-bladed β-propeller fold of RBBP4, while suppressor of zeste 12 (SUZ12), metastasis associated protein 1 (MTA1) and histone H4 bind to a pocket on the side of the WD40 repeats of this protein. Here, we report the discovery of the first small molecule antagonists of the RBBP4 top pocket, competing with interacting peptides from proteins such as BCL11A and histone H3. We also determined the first crystal structure of RBBP4 in complex with a small molecule (OICR17251), paving the path for structure-guided design and optimization towards more potent antagonists.

2020 ◽  
Author(s):  
Sara A. Miller ◽  
Manashree Damle ◽  
Robert E. Kingston

AbstractPolycomb repressive complex 2 (PRC2) catalyzes trimethylation of histone H3 on lysine 27 and is required for normal development of complex eukaryotes. The requirement for H3K27me3 in various aspects of mammalian differentiation is not clear. Though associated with repressed genes, the modification is not sufficient to induce gene repression, and in some instances is not required. To examine the role of the modification in mammalian differentiation, we blocked trimethylation of H3K27 with both a small molecule inhibitor, GSK343, and by introducing a point mutation into EZH2, the catalytic subunit of PRC2. We found that cells with substantively decreased H3K27 tri-methylation were able to differentiate, which contrasts with EZH2 null cells. Different PRC2 targets had varied requirements for H3K27me3 in repressive regulation with a subset that maintained normal levels of repression in the absence of methylation. The primary cellular phenotype when H3K27 tri-methylation was blocked was an inability of the altered cells to maintain a differentiated state when challenged. This phenotype was determined by H3K27me3 deposition both in embryonic stem cells and in the first four days of differentiation. H3K27 tri-methylation therefore was not necessary for formation of differentiated cell states but was required to maintain a stable differentiated state.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 29-29
Author(s):  
Kadriye Nehir Cosgun ◽  
Mark E. Robinson ◽  
Lai N. Chan ◽  
Matthew Hur ◽  
Etienne Leveille ◽  
...  

Abstract Introduction: Oncogenic WNT/β-catenin signaling and activating mutations that increase stability and transcriptional activity of β-catenin are among the most frequent lesions throughout all main types of cancer (Morin et al. 1997; Rubinfeld et al. 1997). Results: Strikingly, pan-cancer analysis of activating mutations of β-catenin in patient samples and global proteomic analysis of human cancer cell lines revealed that B-cell malignancies are exempt from oncogenic activation of β-catenin. While nuclear accumulation of β-catenin was observed in >80% of cancer samples studied, nuclear β-catenin was consistently absent in normal and malignant B-lymphoid cells. To model the effects of inducible β-catenin activation in B-ALL, pre-B cells from Ctnnb1 ex3fl/+ mice were transformed with BCR-ABL1 or NRAS G12D oncogenes. Cre-mediated excision of exon 3 removes GSK3β and CK1 phosphorylation sites (S 33, S 37, T 41 and T 45) required for β-catenin degradation, therefore resulting in accumulation of β-catenin. Stabilization of β-catenin rapidly impaired competitive fitness of B-ALL cells, abolished colony forming ability and induced cell cycle arrest. Likewise, expression of constitutively active form of β-catenin in patient derived B-ALL cells (n=6) or B-cell lymphoma (n=4) compromised proliferation, clonal fitness, and induced cell death. Interestingly, activation of β-catenin signaling in myeloid leukemia (n=4) accelerated proliferation. Corroborating the lineage specific effects of β-catenin, CEBPα-mediated reprogramming of B-ALL cells into myeloid lineage leukemia cells, reversed the deleterious effect of inducible β-catenin-activation. In epithelial cell types, β-catenin/TCF complexes drive transcriptional activation of MYC (He et. al.1998). In striking contrast to epithelial cells, however, our transcriptomic analysis showed that inducible activation of β-catenin in B-cell malignancies resulted in transcriptional repression of Myc and its target genes. Surprisingly, proximity-based labeling and interactome studies in B-ALL cells revealed Ikzf1 and Ikzf3 as top-ranking interaction partners for β-catenin. Furthermore, several components of the Nucleosome Remodeling and Deacetylase (NuRD) complex including Mta1/2, Gatad2a/b, Chd4, Hdac1/2 were among the most significantly enriched proteins within the β-catenin interactome. Interaction of Ikzf1 and Ikzf3 with NuRD complex induced loss of histone H3 lysine 27 acetylation (H3K27Ac) and suppressed Myc expression. To test whether repressive complexes of β-catenin with Ikzf1, Ikzf3:NuRD subvert proliferation and survival by transcriptional repression of Myc, we introduced Cas9-RNP for genetic deletion of Ikzf1 and Ikzf3 in B-ALL cells. Interestingly, deletion of both Ikzf1 and Ikzf3 was required to restore clonal fitness, proliferation and Myc-driven transcriptional programs. Conversely, genetic deletion of β-catenin was sufficient to abrogate Ikzf1/Ikzf3-mediated tumor suppression and transcriptional repression of Myc. Together, these results suggest that engagement of b-catenin set the threshold for Ikzf1/Ikzf3-mediated tumor suppression in B-ALL and mature B-cell malignancies. Together with B-lineage-specific expression of Ikzf1 and Ikzf3, these findings provide a mechanistic explanation as to why activating lesions of β-catenin are not detected in B-cell malignancies. Therapeutic implication: To leverage the unique sensitivity of B-cell malignancies to β-catenin-activation, we tested pharmacological activation of β-catenin based on small molecule-inhibition of GSK3β, a central negative regulator of β-catenin. Testing six different GSK3β inhibitors, we identified LY2090314 as the most potent inhibitor in killing B-ALL cells and suppressing Myc (IC50=4.5 nM). Treatment of NSG mice bearing patient derived B-ALL xenografts with LY2090314 substantially reduced leukemia burden and significantly extended overall survival compared to vehicle treated mice (n=9, P= 6.5E-05). These findings suggest that β-catenin activation is a unique vulnerability and small molecule GSK3β inhibition represents a novel opportunity to overcome drug-resistance in refractory B-ALL. Disclosures No relevant conflicts of interest to declare.


2018 ◽  
Author(s):  
Megha Abbey ◽  
Viacheslav Trush ◽  
Elisa Gibson ◽  
Masoud Vedadi

AbstractRBBP4 and RBBP7 (RBBP4/7) are highly homologous nuclear WD40 motif containing proteins widely implicated in various cancers and are valuable drug targets. They interact with multiple proteins within diverse complexes such as NuRD and PRC2, as well as histone H3 and H4 through two distinct binding sites. FOG-1, PHF6 and histone H3 bind to the top of the donut shape seven-bladed β-propeller fold, while SUZ12, MTA1 and histone H4 bind to a pocket on the side of the WD40 repeats. Here, we briefly review these six interactions and present binding assays optimized for medium to high throughput screening. These assays enable screening of RBBP4/7 toward the discovery of novel cancer therapeutics.


2020 ◽  
Vol 6 (16) ◽  
pp. eaaz0356 ◽  
Author(s):  
Yongqiang Hou ◽  
Wei Liu ◽  
Xianfu Yi ◽  
Yang Yang ◽  
Dongxue Su ◽  
...  

TUDOR domain–containing proteins (TDRDs) are chiefly responsible for recognizing methyl-lysine/arginine residue. However, how TDRD dysregulation contributes to breast tumorigenesis is poorly understood. Here, we report that TUDOR domain–containing PHF20L1 as a H3K27me2 reader exerts transcriptional repression by recruiting polycomb repressive complex 2 (PRC2) and Mi-2/nucleosome remodeling and deacetylase (NuRD) complex, linking PRC2-mediated methylation and NuRD-mediated deacetylation of H3K27. Furthermore, PHF20L1 was found to serve as a potential MYC and hypoxia-driven oncogene, promoting glycolysis, proliferation, and metastasis of breast cancer cells by directly inhibiting tumor suppressors such as HIC1, KISS1, and BRCA1. PHF20L1 expression was also strongly correlated with higher histologic grades of breast cancer and markedly up-regulated in several cancers. Meanwhile, Phf20l1 deletion not only induces growth retardation and mammary ductal outgrowth delay but also inhibits tumorigenesis in vivo. Our data indicate that PHF20L1 promotes tumorigenesis, supporting the pursuit of PHF20L1 as a target for cancer therapy.


Genetics ◽  
2002 ◽  
Vol 162 (2) ◽  
pp. 973-976 ◽  
Author(s):  
Mirela Matecic ◽  
Shelagh Stuart ◽  
Scott G Holmes

Abstract We have identified histone H4 as a high-expression suppressor of Sir2-induced inviability in yeast cells. Overexpression of histone H3 does not suppress Sir2-induced lethality, nor does overexpression of histone H4 alleles associated with silencing defects. These results suggest a direct and specific interaction between Sir2 and H4 in the silencing mechanism.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Sara Busacca ◽  
Qi Zhang ◽  
Annabel Sharkey ◽  
Alan G. Dawson ◽  
David A. Moore ◽  
...  

AbstractWe hypothesized that small molecule transcriptional perturbation could be harnessed to target a cellular dependency involving protein arginine methyltransferase 5 (PRMT5) in the context of methylthioadenosine phosphorylase (MTAP) deletion, seen frequently in malignant pleural mesothelioma (MPM). Here we show, that MTAP deletion is negatively prognostic in MPM. In vitro, the off-patent antibiotic Quinacrine efficiently suppressed PRMT5 transcription, causing chromatin remodelling with reduced global histone H4 symmetrical demethylation. Quinacrine phenocopied PRMT5 RNA interference and small molecule PRMT5 inhibition, reducing clonogenicity in an MTAP-dependent manner. This activity required a functional PRMT5 methyltransferase as MTAP negative cells were rescued by exogenous wild type PRMT5, but not a PRMT5E444Q methyltransferase-dead mutant. We identified c-jun as an essential PRMT5 transcription factor and a probable target for Quinacrine. Our results therefore suggest that small molecule-based transcriptional perturbation of PRMT5 can leverage a mutation-selective vulnerability, that is therapeutically tractable, and has relevance to 9p21 deleted cancers including MPM.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Wendan Ren ◽  
Huitao Fan ◽  
Sara A. Grimm ◽  
Jae Jin Kim ◽  
Linhui Li ◽  
...  

AbstractDNA methylation and trimethylated histone H4 Lysine 20 (H4K20me3) constitute two important heterochromatin-enriched marks that frequently cooperate in silencing repetitive elements of the mammalian genome. However, it remains elusive how these two chromatin modifications crosstalk. Here, we report that DNA methyltransferase 1 (DNMT1) specifically ‘recognizes’ H4K20me3 via its first bromo-adjacent-homology domain (DNMT1BAH1). Engagement of DNMT1BAH1-H4K20me3 ensures heterochromatin targeting of DNMT1 and DNA methylation at LINE-1 retrotransposons, and cooperates with the previously reported readout of histone H3 tail modifications (i.e., H3K9me3 and H3 ubiquitylation) by the RFTS domain to allosterically regulate DNMT1’s activity. Interplay between RFTS and BAH1 domains of DNMT1 profoundly impacts DNA methylation at both global and focal levels and genomic resistance to radiation-induced damage. Together, our study establishes a direct link between H4K20me3 and DNA methylation, providing a mechanism in which multivalent recognition of repressive histone modifications by DNMT1 ensures appropriate DNA methylation patterning and genomic stability.


2013 ◽  
Vol 33 (16) ◽  
pp. 3286-3298 ◽  
Author(s):  
Zhongqi Ge ◽  
Devi Nair ◽  
Xiaoyan Guan ◽  
Neha Rastogi ◽  
Michael A. Freitas ◽  
...  

The best-characterized acetylation of newly synthesized histone H4 is the diacetylation of the NH2-terminal tail on lysines 5 and 12. Despite its evolutionary conservation, this pattern of modification has not been shown to be essential for either viability or chromatin assembly in any model organism. We demonstrate that mutations in histone H4 lysines 5 and 12 in yeast confer hypersensitivity to replication stress and DNA-damaging agents when combined with mutations in histone H4 lysine 91, which has also been found to be a site of acetylation on soluble histone H4. In addition, these mutations confer a dramatic decrease in cell viability when combined with mutations in histone H3 lysine 56. We also show that mutation of the sites of acetylation on newly synthesized histone H4 results in defects in the reassembly of chromatin structure that accompanies the repair of HO-mediated double-strand breaks. This defect is not due to a decrease in the level of histone H3 lysine 56 acetylation. Intriguingly, mutations that alter the sites of newly synthesized histone H4 acetylation display a marked decrease in levels of phosphorylated H2A (γ-H2AX) in chromatin surrounding the double-strand break. These results indicate that the sites of acetylation on newly synthesized histones H3 and H4 can function in nonoverlapping ways that are required for chromatin assembly, viability, and DNA damage response signaling.


Sign in / Sign up

Export Citation Format

Share Document