Convergent Alterations of a Protein Hub Produce Divergent Effects within a Binding SiteClick to copy article linkArticle link copied!
- Ali ImranAli ImranDepartment of Physics, Syracuse University, 201 Physics Building, Syracuse, New York 13244-1130, United StatesMore by Ali Imran
- Brandon S. MoyerBrandon S. MoyerIchor Life Sciences, Inc., 2651 US Route 11, LaFayette, New York 13084, United StatesMore by Brandon S. Moyer
- Dan KalinaDan KalinaIchor Life Sciences, Inc., 2651 US Route 11, LaFayette, New York 13084, United StatesDepartment of Chemistry, State University of New York College of Environmental Science and Forestry, 1 Forestry Dr., Syracuse, New York 13210, United StatesMore by Dan Kalina
- Thomas M. DuncanThomas M. DuncanDepartment of Biochemistry and Molecular Biology, State University of New York Upstate Medical University, 4249 Weiskotten Hall, 766 Irving Avenue, Syracuse, New York 13210, United StatesMore by Thomas M. Duncan
- Kelsey J. MoodyKelsey J. MoodyDepartment of Physics, Syracuse University, 201 Physics Building, Syracuse, New York 13244-1130, United StatesIchor Life Sciences, Inc., 2651 US Route 11, LaFayette, New York 13084, United StatesDepartment of Chemistry, State University of New York College of Environmental Science and Forestry, 1 Forestry Dr., Syracuse, New York 13210, United StatesLewis School of Health Sciences, Clarkson University, 8 Clarkson Avenue, Potsdam, New York 13699, United StatesMore by Kelsey J. Moody
- Aaron J. WolfeAaron J. WolfeDepartment of Physics, Syracuse University, 201 Physics Building, Syracuse, New York 13244-1130, United StatesIchor Life Sciences, Inc., 2651 US Route 11, LaFayette, New York 13084, United StatesDepartment of Chemistry, State University of New York College of Environmental Science and Forestry, 1 Forestry Dr., Syracuse, New York 13210, United StatesLewis School of Health Sciences, Clarkson University, 8 Clarkson Avenue, Potsdam, New York 13699, United StatesMore by Aaron J. Wolfe
- Michael S. Cosgrove*Michael S. Cosgrove*Email: [email protected]Department of Biochemistry and Molecular Biology, State University of New York Upstate Medical University, 4249 Weiskotten Hall, 766 Irving Avenue, Syracuse, New York 13210, United StatesMore by Michael S. Cosgrove
- Liviu Movileanu*Liviu Movileanu*Email: [email protected]Department of Physics, Syracuse University, 201 Physics Building, Syracuse, New York 13244-1130, United StatesDepartment of Biomedical and Chemical Engineering, Syracuse University, 329 Link Hall, Syracuse, New York 13244, United StatesThe BioInspired Institute, Syracuse University, Syracuse, New York 13244, United StatesMore by Liviu Movileanu
Abstract
Progress in tumor sequencing and cancer databases has created an enormous amount of information that scientists struggle to sift through. While several research groups have created computational methods to analyze these databases, much work still remains in distinguishing key implications of pathogenic mutations. Here, we describe an approach to identify and evaluate somatic cancer mutations of WD40 repeat protein 5 (WDR5), a chromatin-associated protein hub. This multitasking protein maintains the functional integrity of large multi-subunit enzymatic complexes of the six human SET1 methyltransferases. Remarkably, the somatic cancer mutations of WDR5 preferentially distribute within and around an essential cavity, which hosts the WDR5 interaction (Win) binding site. Hence, we assessed the real-time binding kinetics of the interactions of key clustered WDR5 mutants with the Win motif peptide ligands of the SET1 family members (SET1Win). Our measurements highlight that this subset of mutants exhibits divergent perturbations in the kinetics and strength of interactions not only relative to those of the native WDR5 but also among various SET1Win ligands. These outcomes could form a fundamental basis for future drug discovery and other developments in medical biotechnology.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Figure 1
Figure 1. Two binding sites of WDR5 and the structure of the WDR5-MLL3Win complex. (a) Representations of the Win and WBM binding sites of WDR5. Orientations of WDR5 in the two cartoons are 180° with respect to each other. (b) Top view of the WDR5-MLL3Win complex. (c) Side view of the WDR5-MLL3Win complex. (d) Side view of the interaction sites between MLL3Win (green) and WDR5 (gray). All presented residues are within 5 Å of the other binding partner. The residues corresponding to the WDR5 mutations explored in this study are marked in dark gray. MLL3Win residues are labeled as well. Potential hydrogen bonds between the two binding partners are shown as yellow dotted lines. (e) Key residues of the WDR5 binding cavity involved in hydrogen bonding with the evolutionarily conserved Arg residue (R4710) of MLL3Win at position P0 (Table S1). The hydrogen bonds are indicated by thick dashed lines marked in yellow. The cutoff distance for identifying these hydrogen bonds was 4.0 Å. WDR5 was represented using pdb entry 4ERY. (35)
Results and Discussion
Use of CLUMPS for the Identification of Mutation Clustering in WDR5
N | m | WAP score | P-value |
---|---|---|---|
<10 000 | 51 | 2.258 | 0.403 |
<5000 | 33 | 0.931 | 0.206 |
<1000 | 11 | 0.095 | 0.111 |
<500 | 8 | 0.072 | 0.025 |
WAP scores were calculated using four different subsets of mutations divided on the basis of the genetic damage, N, in their corresponding tumors. The P-values were calculated by comparison to configurations with random permutations of the distribution of mutations. 106 configurations were used for each subset. N is the total number of mutations in a given tumor sample. m is the total number of mutations that met the condition N < Nmax, where Nmax is the upper limit of the number of mutations in a given tumor sample. Nmax values are listed below for four data subsets on the first column. m was kept constant for all configurations of a subset.
Biolayer Interferometry Measurements
Figure 2
Figure 2. Label-free optical BLI sensorgrams of the WDR5 mutant-MLL3Win interactions. (a) Locations of the surface and cavity WDR5 mutations are shown in blue using surface and cross-sectional views of WDR5, respectively. (b) BLI sensorgrams showing the association and dissociation phases. For each WDR5 mutation, sensors with immobilized MLL3Win ligand were immersed in buffers containing different WDR5 concentrations (listed on sensorgrams) to monitor association kinetics. Sensors were then transferred to buffer alone to monitor dissociation kinetics.
Surface Mutants
Figure 3
Figure 3. Normalized dissociation rate constants of the WDR5 mutant-SET1Win interactions using BLI sensorgrams. The koff values for each SET1Win ligand’s interaction with mutants have been divided by the koff of that SET1Win ligand’s interaction with the native WDR5 protein. (a) MLL2Win, (b) MLL3Win, (c) MLL4Win, (d) SETd1AWin, and (e) SETd1BWin. ND stands for “Not Determined”. Using a BLI measurement, the interaction between F133L and MLL4Win was detectable, but not quantifiable.
Figure 4
Figure 4. Normalized KD of the WDR5 mutant-SET1Win interactions using BLI sensorgrams. The KD values for each SET1Win ligand’s interaction with WDR5 mutants have been divided by the KD of that SET1Win ligand’s interaction with the native WDR5 protein. (a) MLL2Win, (b) MLL3Win, (c) MLL4Win, (d) SETd1AWin, and (e) SETd1BWin. ND stands for “Not Determined”. Using a BLI measurement, the interaction between F133L and MLL4Win was detectable, but not quantifiable.
Cavity Mutants
Validations of BLI Data and Qualitative Comparisons between Competing Techniques
Figure 5
Figure 5. Normalized KD of the WDR5 mutant-SET1Win interactions using steady-state FP spectroscopy. The KD values for each SET1Win ligand’s interaction with WDR5 mutants have been divided by the KD of that SET1Win ligand’s interaction with the native WDR5 protein. (a) MLL1Win, (b) MLL2Win, (c) MLL3Win, (d) MLL4Win, (e) SETd1AWin, and (f) SETd1BWin. For vertical bars marked by “*”, the KD of those interactions could not be determined. Those values represent the lower-limit of the KD based on the highest WDR5 mutant concentrations used in this study.
Figure 6
Figure 6. Quantitative comparison between affinity data resulting from BLI and FP measurements. (a) 3D graph of the ratio of the normalized KD-BLI to the normalized KD-FP. (b) Two-dimensional heat map of the ratio of the normalized KD-BLI to the normalized KD-FP. Normalized KD values are the KD measured for a specific WDR5 mutant-SET1Win interaction pair divided by the KD value corresponding to the native WDR5 protein.
Implications of Win Binding Site Mutants
Concluding Remarks
Materials and Methods
Clustering of Mutations in Protein Structures


Protein Expression and Purification
Peptide Synthesis, Labeling, Purification, and Analysis
Biolayer Interferometry



Steady-State Fluorescence Polarization (Steady-State FP) Measurements
Molecular Graphics
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acschembio.2c00273.
Sequence of SET1Win peptide ligands; location of somatic cancer mutations from tumor samples with N < 500; results of mutation clustering for different mutation subsets; list of tumor locations associated with missense WDR5 mutations within and around the Win binding site; list of noncovalent bonds at the WDR5-SET1Win interface; location of surface WDR5 mutations within the A and B pockets; BLI measurements of very weak interactions; kinetic rate constants of association and dissociation of WDR5 mutants with SET1Win ligands; equilibrium dissociation constants of WDR5 mutants with SET1Win ligands; structural information on the effect of the S175L mutation; steady-state FP spectroscopy curves for the interactions of WDR5 mutants with SET1win ligands; equilibrium dissociation constants of WDR5 mutants with SET1Win ligands using steady-state FP spectroscopy; quantitative comparisons of affinity data acquired with BLI and FP; and supporting references (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
We are grateful to our colleagues in the Movileanu and Cosgrove laboratories and at Ichor Life Sciences Laboratories for their comments on the paper and stimulating discussions as well as for their assistance during the early stage of this project. This work was supported by the National Cancer Institute of the U.S. National Institutes of Health grant R01 CA140522 (to M.S.C.) and by the National Institute of General Medical Sciences of the U.S. National Institutes of Health grant R01 GM129429 (to L.M.).
References
This article references 76 other publications.
- 1Xu, C.; Min, J. Structure and function of WD40 domain proteins. Protein Cell 2011, 2, 202– 214, DOI: 10.1007/s13238-011-1018-1Google Scholar1Structure and function of WD40 domain proteinsXu, Chao; Min, JinrongProtein & Cell (2011), 2 (3), 202-214CODEN: PCREFB; ISSN:1674-800X. (Higher Education Press)A review. The WD40 domain exhibits a β-propeller architecture, often comprising seven blades. The WD40 domain is one of the most abundant domains and also among the top interacting domains in eukaryotic genomes. In this review, we will discuss the identification, definition and architecture of the WD40 domains. WD40 domain proteins are involved in a large variety of cellular processes, in which WD40 domains function as a protein-protein or protein-DNA interaction platform. WD40 domain mediates mol. recognition events mainly through the smaller top surface, but also through the bottom surface and sides. So far, no WD40 domain has been found to display enzymic activity. We will also discuss the different binding modes exhibited by the large versatile family of WD40 domain proteins. In the last part of this review, we will discuss how post-translational modifications are recognized by WD40 domain proteins.
- 2Zhang, C.; Zhang, F. The Multifunctions of WD40 Proteins in Genome Integrity and Cell Cycle Progression. J. Genomics 2015, 3, 40– 50, DOI: 10.7150/jgen.11015Google Scholar2The Multifunctions of WD40 Proteins in Genome Integrity and Cell Cycle ProgressionZhang Caiguo; Zhang FanJournal of genomics (2015), 3 (), 40-50 ISSN:1839-9940.Eukaryotic genome encodes numerous WD40 repeat proteins, which generally function as platforms of protein-protein interactions and are involved in numerous biological process, such as signal transduction, gene transcriptional regulation, protein modifications, cytoskeleton assembly, vesicular trafficking, DNA damage and repair, cell death and cell cycle progression. Among these diverse functions, genome integrity maintenance and cell cycle progression are extremely important as deregulation of them is clinically linked to uncontrolled proliferative diseases such as cancer. Thus, we mainly summarize and discuss the recent understanding of WD40 proteins and their molecular mechanisms linked to genome stability and cell cycle progression in this review, thereby demonstrating their pervasiveness and importance in cellular networks.
- 3Afanasieva, E.; Chaudhuri, I.; Martin, J.; Hertle, E.; Ursinus, A.; Alva, V.; Hartmann, M. D.; Lupas, A. N. Structural diversity of oligomeric β-propellers with different numbers of identical blades. eLife 2019, 8, e49853 DOI: 10.7554/eLife.49853Google Scholar3Structural diversity of oligomeric β-propellers with different numbers of identical bladesAfanasieva, Evgenia; Chaudhuri, Indronil; Martin, Joerg; Hertle, Eva; Ursinus, Astrid; Alva, Vikram; Hartmann, Marcus D.; Lupas, Andrei N.eLife (2019), 8 (), e49853/1-e49853/15CODEN: ELIFA8; ISSN:2050-084X. (eLife Sciences Publications Ltd.)β-Propellers arise through the amplification of a supersecondary structure element called a blade. This process produces toroids of between four and twelve repeats, which are almost always arranged sequentially in a single polypeptide chain. We found that new propellers evolve continuously by amplification from single blades. We therefore investigated whether such nascent propellers can fold as homo-oligomers before they have been fully amplified within a single chain. One- to six-bladed building blocks derived from two seven-bladed WD40 propellers yielded stable homo-oligomers with six to nine blades, depending on the size of the building block. High resoln. structures for tetramers of two blades, trimers of three blades, and dimers of four and five blades, resp., show structurally diverse propellers and include a novel fold, highlighting the inherent flexibility of the WD40 blade. Our data support the hypothesis that subdomain-sized fragments can provide structural versatility in the evolution of new proteins.
- 4Stirnimann, C. U.; Petsalaki, E.; Russell, R. B.; Müller, C. W. WD40 proteins propel cellular networks. Trends Biochem. Sci. 2010, 35, 565– 574, DOI: 10.1016/j.tibs.2010.04.003Google Scholar4WD40 proteins propel cellular networksStirnimann, Christian U.; Petsalaki, Evangelia; Russell, Robert B.; Mueller, Christoph W.Trends in Biochemical Sciences (2010), 35 (10), 565-574CODEN: TBSCDB; ISSN:0968-0004. (Elsevier Ltd.)A review. Recent findings indicate that WD40 domains play central roles in biol. processes by acting as hubs in cellular networks; however, they have been studied less intensely than other common domains, such as the kinase, PDZ or SH3 domains. As suggested by various interactome studies, they are among the most promiscuous interactors. Structural studies suggest that this property stems from their ability, as scaffolds, to interact with diverse proteins, peptides or nucleic acids using multiple surfaces or modes of interaction. A general scaffolding role is supported by the fact that no WD40 domain has been found with intrinsic enzymic activity despite often being part of large mol. machines. We discuss the WD40 domain distributions in protein networks and structures of WD40-contg. assemblies to demonstrate their versatility in mediating crit. cellular functions.
- 5Santosh Kumar, H. S.; Kumar, V.; Kumar, V.; Pattar, S.; Telkar, S. Towards the construction of an interactome for Human WD40 protein family. Bioinformation 2016, 12, 54– 61, DOI: 10.6026/97320630012054Google Scholar5Towards the construction of an interactome for Human WD40 protein familySantosh Kumar Hulikal Shivashankara; Kumar Vadlapudi; Pattar Sharath; Telkar SandeepBioinformation (2016), 12 (2), 54-61 ISSN:0973-2063.WD40 proteins are involved in a variety of protein-protein interactions as part of a multi-protein assembly modulating diverse and critical cellular process. It is known that several proteins of this family have been implicated in different disorders such as developmental abnormalities and cancer. However, molecular functions of many proteins in this family are yet unknown and it is of clinical interest. Therefore, it is of interest to define, construct, understand, analyze, evaluate, redefine and refine an interactome for WD40 protein family. We used data from literature mining using Cytoscape followed by linear regression analysis between Betweenness centrality and stress scores to define a model to filter the nodes in a representative WD40 interactome construction. We identified 10 ranked nodes in this analysis and subsequent microarray data selected three of them in insulin resistance that is further demonstrated in HepG2 cell culture models. We also observed the expression of GRWD1, RBBP5 and WDR5 genes during perturbation. Thus, we report hub nodes of WD40 interactome in insulin resistance. It should be noted that the pipeline using protein interaction network help find new proteins of clinical importance.
- 6Migliori, V.; Mapelli, M.; Guccione, E. On WD40 proteins: propelling our knowledge of transcriptional control?. Epigenetics 2012, 7, 815– 822, DOI: 10.4161/epi.21140Google Scholar6On WD40 proteins: propelling our knowledge of transcriptional control?Migliori, Valentina; Mapelli, Marina; Guccione, ErnestoEpigenetics (2012), 7 (8), 815-822CODEN: EPIGB6; ISSN:1559-2294. (Landes Bioscience)A review. A direct effect of post-translational modifications (PTMs) on nucleosomes is the formation of a dynamic platform able to assemble the transcriptional machinery and to recruit chromatin modifiers. The histone code hypothesis suggests that histone PTMs can act as binding sites for chromatin readers and effector proteins, such as the bromodomains, that selectively interact with acetylated lysines, or the "royal family" and the PHD finger domains, which are able to recognize methylated arginines and lysines. In this review we will discuss recent data describing the function of WD40 proteins as a new class of histone readers, with particular emphasis on the ones able to recognize methylated arginine and lysine residues. We will discuss how WDR5, a classical seven-bladed WD40 propeller, is able to bind with similar affinities both the catalytic subunit of the Trithorax-like complexes, and the histone H3 tail either unmodified or sym. dimethylated on arginine 2 (H3R2me2s). Furthermore, we will speculate on how these mutually exclusive interactions of WDR5 may play a role in mediating different degrees of H3K4 methylations at both promoters and distal regulatory sites. Finally, we will summarize recent literature elucidating how other WD40 proteins such as NURF55, EED and LRWD1 recognize methylated histone tails, highlighting similarities and differences among them.
- 7Jain, B. P.; Pandey, S. WD40 Repeat Proteins: Signalling Scaffold with Diverse Functions. Protein J. 2018, 37, 391– 406, DOI: 10.1007/s10930-018-9785-7Google Scholar7WD40 Repeat Proteins: Signalling Scaffold with Diverse FunctionsJain, Buddhi Prakash; Pandey, ShwetaProtein Journal (2018), 37 (5), 391-406CODEN: PJROAH; ISSN:1572-3887. (Springer)A review. The WD40 domain is one of the most abundant and interacting domains in the eukaryotic genome. In proteins the WD domain folds into a β-propeller structure, providing a platform for the interaction and assembly of several proteins into a signalosome. WD40 repeats contg. proteins, in lower eukaryotes, are mainly involved in growth, cell cycle, development and virulence, while in higher organisms, they play an important role in diverse cellular functions like signal transduction, cell cycle control, intracellular transport, chromatin remodelling, cytoskeletal organization, apoptosis, development, transcriptional regulation, immune responses. To play the regulatory role in various processes, they act as a scaffold for protein-protein or protein-DNA interaction. So far, no WD40 domain has been identified with intrinsic enzymic activity. Several WD40 domain-contg. proteins have been recently characterized in prokaryotes as well. The review summarizes the vast array of functions performed by different WD40 domain contg. proteins, their domain organization and functional conservation during the course of evolution.
- 8Cosgrove, M. S.; Patel, A. Mixed lineage leukemia: a structure-function perspective of the MLL1 protein. FEBS J. 2010, 277, 1832– 1842, DOI: 10.1111/j.1742-4658.2010.07609.xGoogle Scholar8Mixed lineage leukemia: a structure-function perspective of the MLL1 proteinCosgrove, Michael S.; Patel, AnamikaFEBS Journal (2010), 277 (8), 1832-1842CODEN: FJEOAC; ISSN:1742-464X. (Wiley-Blackwell)A review. Several acute lymphoblastic and myelogenous leukemias are correlated with alterations in the human mixed lineage leukemia protein-1 (MLL1) gene. MLL1 is a member of the evolutionarily conserved SET1 family of histone H3 lysine 4 (H3K4) methyltransferases, which are required for the regulation of distinct groups of developmentally regulated genes in metazoans. Despite the important biol. role of SET1 family enzymes and their involvement in human leukemias, relatively little is understood about how these enzymes work. Here, the authors review several recent structural and biochem. studies that are beginning to shed light on the mol. mechanisms for the regulation of H3K4 methylation by the human MLL1 enzyme.
- 9Li, Y.; Han, J.; Zhang, Y.; Cao, F.; Liu, Z.; Li, S.; Wu, J.; Hu, C.; Wang, Y.; Shuai, J.; Chen, J.; Cao, L.; Li, D.; Shi, P.; Tian, C.; Zhang, J.; Dou, Y.; Li, G.; Chen, Y.; Lei, M. Structural basis for activity regulation of MLL family methyltransferases. Nature 2016, 530, 447– 452, DOI: 10.1038/nature16952Google Scholar9Structural basis for activity regulation of MLL family methyltransferasesLi, Yanjing; Han, Jianming; Zhang, Yuebin; Cao, Fang; Liu, Zhijun; Li, Shuai; Wu, Jian; Hu, Chunyi; Wang, Yan; Shuai, Jin; Chen, Juan; Cao, Liaoran; Li, Dangsheng; Shi, Pan; Tian, Changlin; Zhang, Jian; Dou, Yali; Li, Guohui; Chen, Yong; Lei, MingNature (London, United Kingdom) (2016), 530 (7591), 447-452CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)The mixed lineage leukemia (MLL) family of proteins (including MLL1-MLL4, SET1A and SET1B) specifically methylate histone 3 Lys4, and have pivotal roles in the transcriptional regulation of genes involved in haematopoiesis and development. The methyltransferase activity of MLL1, by itself severely compromised, is stimulated by the three conserved factors WDR5, RBBP5 and ASH2L, which are shared by all MLL family complexes. However, the mol. mechanism of how these factors regulate the activity of MLL proteins still remains poorly understood. Here we show that a minimized human RBBP5-ASH2L heterodimer is the structural unit that interacts with and activates all MLL family histone methyltransferases. Our structural, biochem. and computational analyses reveal a two-step activation mechanism of MLL family proteins. These findings provide unprecedented insights into the common theme and functional plasticity in complex assembly and activity regulation of MLL family methyltransferases, and also suggest a universal regulation mechanism for most histone methyltransferases.
- 10Vedadi, M.; Blazer, L.; Eram, M. S.; Barsyte-Lovejoy, D.; Arrowsmith, C. H.; Hajian, T. Targeting human SET1/MLL family of proteins. Protein Sci. 2017, 26, 662– 676, DOI: 10.1002/pro.3129Google Scholar10Targeting human SET1/MLL family of proteinsVedadi, Masoud; Blazer, Levi; Eram, Mohammad S.; Barsyte-Lovejoy, Dalia; Arrowsmith, Cheryl H.; Hajian, TaranehProtein Science (2017), 26 (4), 662-676CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)The SET1 family of proteins, and in particular MLL1, are essential regulators of transcription and key mediators of normal development and disease. Here, we summarize the detailed characterization of the methyltransferase activity of SET1 complexes and the role of the key subunits, WDR5, RbBP5, ASH2L, and DPY30. We present new data on full kinetic characterization of human MLL1, MLL3, SET1A, and SET1B trimeric, tetrameric, and pentameric complexes to elaborate on substrate specificities and compare our findings with what has been reported before. We also review exciting recent work identifying potent inhibitors of oncogenic MLL1 function through disruption of protein-protein interactions within the MLL1 complex.
- 11Xue, H.; Yao, T.; Cao, M.; Zhu, G.; Li, Y.; Yuan, G.; Chen, Y.; Lei, M.; Huang, J. Structural basis of nucleosome recognition and modification by MLL methyltransferases. Nature 2019, 573, 445– 449, DOI: 10.1038/s41586-019-1528-1Google Scholar11Structural basis of nucleosome recognition and modification by MLL methyltransferasesXue, Han; Yao, Tonghui; Cao, Mi; Zhu, Guanjun; Li, Yan; Yuan, Guiyong; Chen, Yong; Lei, Ming; Huang, JingNature (London, United Kingdom) (2019), 573 (7774), 445-449CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Methyltransferases of the mixed-lineage leukemia (MLL) family-which include MLL1, MLL2, MLL3, MLL4, SET1A and SET1B-implement methylation of histone H3 on lysine 4 (H3K4), and have crit. and distinct roles in the regulation of transcription in haematopoiesis, adipogenesis and development. The C-terminal catalytic SET (Su(var.)3-9, enhancer of zeste and trithorax) domains of MLL proteins are assocd. with a common set of regulatory factors (WDR5, RBBP5, ASH2L and DPY30) to achieve specific activities. Current knowledge of the regulation of MLL activity is limited to the catalysis of histone H3 peptides, and how H3K4 Me marks are deposited on nucleosomes is poorly understood. H3K4 methylation is stimulated by mono-ubiquitination of histone H2B on lysine 120 (H2BK120ub1), a prevalent histone H2B mark that disrupts chromatin compaction and favors open chromatin structures, but the underlying mechanism remains unknown10-12. Here we report cryo-electron microscopy structures of human MLL1 and MLL3 catalytic modules assocd. with nucleosome core particles that contain H2BK120ub1 or unmodified H2BK120. These structures demonstrate that the MLL1 and MLL3 complexes both make extensive contacts with the histone-fold and DNA regions of the nucleosome; this allows ease of access to the histone H3 tail, which is essential for the efficient methylation of H3K4. The H2B-conjugated ubiquitin binds directly to RBBP5, orienting the assocn. between MLL1 or MLL3 and the nucleosome. The MLL1 and MLL3 complexes display different structural organizations at the interface between the WDR5, RBBP5 and MLL1 (or the corresponding MLL3) subunits, which accounts for the opposite roles of WDR5 in regulating the activity of the two enzymes. These findings transform our understanding of the structural basis for the regulation of MLL activity at the nucleosome level, and highlight the pivotal role of nucleosome regulation in histone-tail modification.
- 12Jiang, H. The complex activities of the SET1/MLL complex core subunits in development and disease. Biochim. Biophys. Acta, Gene Regul. Mech. 2020, 1863, 194560, DOI: 10.1016/j.bbagrm.2020.194560Google Scholar12The complex activities of the SET1/MLL complex core subunits in development and diseaseJiang, HaoBiochimica et Biophysica Acta, Gene Regulatory Mechanisms (2020), 1863 (7), 194560CODEN: BBAGC6; ISSN:1874-9399. (Elsevier B.V.)In mammalian cells, the SET1/MLL complexes are the main writers of the H3K4 Me mark that is assocd. with active gene expression. The activities of these complexes are critically dependent on the assocn. of the catalytic subunit with their shared core subunits, WDR5, RBBP5, ASH2L, and DPY30, collectively referred as WRAD. In addn., some of these core subunits can bind to proteins other than the SET1/MLL complex components. This review starts with discussion of the mol. activities of these core subunits, with an emphasis on DPY30 in organizing the assembly of the SET1/MLL complexes with other assocd. factors. This review then focuses on the roles of the core subunits in stem cells and development, as well as in diseased cell states, mainly cancer, and ends with discussion on dissecting the responsible activities of the core subunits and how we may target them for potential disease treatment. This article is part of a Special Issue entitled: The MLL family of proteins in normal development and disease edited by Thomas A Milne.
- 13Sha, L.; Ayoub, A.; Cho, U.-S.; Dou, Y. Insights on the regulation of the MLL/SET1 family histone methyltransferases. Biochim. Biophys. Acta, Gene Regul. Mech. 2020, 1863, 194561, DOI: 10.1016/j.bbagrm.2020.194561Google Scholar13Insights on the regulation of the MLL/SET1 family histone methyltransferasesSha, Liang; Ayoub, Alex; Cho, Uhn-Soo; Dou, YaliBiochimica et Biophysica Acta, Gene Regulatory Mechanisms (2020), 1863 (7), 194561CODEN: BBAGC6; ISSN:1874-9399. (Elsevier B.V.)A review. In eukaryotes, histone H3K4 methylation by the MLL/SET1 family histone methyltransferases is enriched at transcription regulatory elements including gene promoters and enhancers. The level of H3K4 methylation is highly correlated with transcription activation and is one of the most frequently used histone post-translational modifications to predict transcriptional outcome. Recently, it has been shown that rearrangement of the cellular landscape of H3K4 mono-methylation at distal enhancers precedes cell fate transition and is used for identification of novel regulatory elements for development and disease progression. Similarly, broad H3K4 tri-methylation regions have also been used to predict intrinsic tumor suppression properties of regulator regions in a variety of cellular models. Understanding the regulation for how H3K4 methylation is deposited and regulated is of paramount importance. In this review, we will discuss new findings on how the MLL/SET1 family enzymes are regulated on chromatin and their potential functional and regulatory implications. This article is part of a Special Issue entitled: The MLL family of proteins in normal development and disease edited by Thomas A Milne.
- 14Dou, Y.; Milne, T. A.; Ruthenburg, A. J.; Lee, S.; Lee, J. W.; Verdine, G. L.; Allis, C. D.; Roeder, R. G. Regulation of MLL1 H3K4 methyltransferase activity by its core components. Nat. Struct. Mol. Biol. 2006, 13, 713– 719, DOI: 10.1038/nsmb1128Google Scholar14Regulation of MLL1 H3K4 methyltransferase activity by its core componentsDou, Yali; Milne, Thomas A.; Ruthenburg, Alexander J.; Lee, Seunghee; Lee, Jae Woon; Verdine, Gregory L.; Allis, C. David; Roeder, Robert G.Nature Structural & Molecular Biology (2006), 13 (8), 713-719CODEN: NSMBCU; ISSN:1545-9993. (Nature Publishing Group)Histone H3 Lys4 (H3K4) methylation is a prevalent mark assocd. with transcription activation. A common feature of several H3K4 methyltransferase complexes is the presence of three structural components (RbBP5, Ash2L and WDR5) and a catalytic subunit contg. a SET domain. Here we report the first biochem. reconstitution of a functional four-component mixed-lineage leukemia protein-1 (MLL1) core complex. This reconstitution, combined with in vivo assays, allows direct anal. of the contribution of each component to MLL1 enzymic activity and their roles in transcriptional regulation. Moreover, taking clues from a crystal structure anal., we demonstrate that WDR5 mediates interactions of the MLL1 catalytic unit both with the common structural platform and with the histone substrate. Mechanistic insights gained from this study can be generalized to the whole family of SET1-like histone methyltransferases in mammals.
- 15Patel, A.; Dharmarajan, V.; Vought, V. E.; Cosgrove, M. S. On the mechanism of multiple lysine methylation by the human mixed lineage leukemia protein-1 (MLL1) core complex. J. Biol. Chem. 2009, 284, 24242– 24256, DOI: 10.1074/jbc.m109.014498Google Scholar15On the Mechanism of Multiple Lysine Methylation by the Human Mixed Lineage Leukemia Protein-1 (MLL1) Core ComplexPatel, Anamika; Dharmarajan, Venkatasubramanian; Vought, Valarie E.; Cosgrove, Michael S.Journal of Biological Chemistry (2009), 284 (36), 24242-24256CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Transcription in eukaryotic genomes depends on enzymes that regulate the degree of histone H3 lysine 4 (H3K4) methylation. The mixed lineage leukemia protein-1 (MLL1) is a member of the SET1 family of H3K4 methyltransferases and is frequently rearranged in acute leukemias. Despite sequence comparisons that predict that SET1 family enzymes should only monomethylate their substrates, mono-, di-, and trimethylation of H3K4 has been attributed to SET1 family complexes in vivo and in vitro. To better understand this paradox, we have biochem. reconstituted and characterized a five-component 200-kDa MLL1 core complex contg. human MLL1, WDR5, RbBP5, Ash2L, and DPY-30. We demonstrate that the isolated MLL1 SET domain is a slow monomethyltransferase and that tyrosine 3942 of MLL1 prevents di- and trimethylation of H3K4. In contrast, a complex contg. the MLL1 SET domain, WDR5, RbBP5, Ash2L, and DPY-30, displays a marked ∼600-fold increase in enzymic activity but only to the di-Me form of H3K4. Single turnover kinetic expts. reveal that the reaction leading to H3K4 dimethylation involves the transient accumulation of a monomethylated species, suggesting that the MLL1 core complex uses a non-processive mechanism to catalyze multiple lysine methylation. We have also discovered that the non-SET domain components of the MLL1 core complex possess a previously unrecognized methyltransferase activity that catalyzes H3K4 dimethylation within the MLL1 core complex. Our results suggest that the mechanism of multiple lysine methylation by the MLL1 core complex involves the sequential addn. of two Me groups at two distinct active sites within the complex.
- 16Shinsky, S. A.; Monteith, K. E.; Viggiano, S.; Cosgrove, M. S. Biochemical reconstitution and phylogenetic comparison of human SET1 family core complexes involved in histone methylation. J. Biol. Chem. 2015, 290, 6361– 6375, DOI: 10.1074/jbc.m114.627646Google Scholar16Biochemical Reconstitution and Phylogenetic Comparison of Human SET1 Family Core Complexes Involved in Histone MethylationShinsky, Stephen A.; Monteith, Kelsey E.; Viggiano, Susan; Cosgrove, Michael S.Journal of Biological Chemistry (2015), 290 (10), 6361-6375CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Mixed lineage leukemia protein-1 (MLL1) is a member of the SET1 family of histone H3 lysine 4 (H3K4) methyltransferases that are required for metazoan development. MLL1 is the best characterized human SET1 family member, which includes MLL1-4 and SETd1A/B. MLL1 assembles with WDR5, RBBP5, ASH2L, DPY-30 (WRAD) to form the MLL1 core complex, which is required for H3K4 dimethylation and transcriptional activation. Because all SET1 family proteins interact with WRAD in vivo, it is hypothesized they are regulated by similar mechanisms. However, recent evidence suggests differences among family members that may reflect unique regulatory inputs in the cell. Missing is an understanding of the intrinsic enzymic activities of different SET1 family complexes under std. conditions. In this investigation, we reconstituted each human SET1 family core complex and compared subunit assembly and enzymic activities. We found that in the absence of WRAD, all but one SET domain catalyzes at least weak H3K4 monomethylation. In the presence of WRAD, all SET1 family members showed stimulated monomethyltransferase activity but differed in their di- and trimethylation activities. We found that these differences are correlated with evolutionary lineage, suggesting these enzyme complexes have evolved to accomplish unique tasks within metazoan genomes. To understand the structural basis for these differences, we employed a "phylogenetic scanning mutagenesis" assay and identified a cluster of amino acid substitutions that confer a WRAD-dependent gain-of-function dimethylation activity on complexes assembled with the MLL3 or Drosophila trithorax proteins. These results form the basis for understanding how WRAD differentially regulates SET1 family complexes in vivo.
- 17Thomas, L. R.; Wang, Q.; Grieb, B. C.; Phan, J.; Foshage, A. M.; Sun, Q.; Olejniczak, E. T.; Clark, T.; Dey, S.; Lorey, S.; Alicie, B.; Howard, G. C.; Cawthon, B.; Ess, K. C.; Eischen, C. M.; Zhao, Z.; Fesik, S. W.; Tansey, W. P. Interaction with WDR5 promotes target gene recognition and tumorigenesis by MYC. Mol. Cell. Biochem. 2015, 58, 440– 452, DOI: 10.1016/j.molcel.2015.02.028Google ScholarThere is no corresponding record for this reference.
- 18Thomas, L. R.; Adams, C. M.; Wang, J.; Weissmiller, A. M.; Creighton, J.; Lorey, S. L.; Liu, Q.; Fesik, S. W.; Eischen, C. M.; Tansey, W. P. Interaction of the oncoprotein transcription factor MYC with its chromatin cofactor WDR5 is essential for tumor maintenance. Proc. Natl. Acad. Sci. U.S.A. 2019, 116, 25260– 25268, DOI: 10.1073/pnas.1910391116Google Scholar18Interaction of the oncoprotein transcription factor MYC with its chromatin cofactor WDR5 is essential for tumor maintenanceThomas, Lance R.; Adams, Clare M.; Wang, Jing; Weissmiller, April M.; Creighton, Joy; Lorey, Shelly L.; Liu, Qi; Fesik, Stephen W.; Eischen, Christine M.; Tansey, William P.Proceedings of the National Academy of Sciences of the United States of America (2019), 116 (50), 25260-25268CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The oncoprotein transcription factor MYC is overexpressed in the majority of cancers. Key to its oncogenic activity is the ability of MYC to regulate gene expression patterns that drive and maintain the malignant state. MYC is also considered a validated anticancer target, but efforts to pharmacol. inhibit MYC have failed. The dependence of MYC on cofactors creates opportunities for therapeutic intervention, but for any cofactor this requires structural understanding of how the cofactor interacts with MYC, knowledge of the role it plays in MYC function, and demonstration that disrupting the cofactor interaction will cause existing cancers to regress. One cofactor for which structural information is available is WDR5, which interacts with MYC to facilitate its recruitment to chromatin. To explore whether disruption of the MY-WDR5 interaction could potentially become a viable anticancer strategy, we developed a Burkitt's lymphoma system that allows replacement of wild-type MYC for mutants that are defective for WDR5 binding or all known nuclear MYC functions. Using this system, we show that WDR5 recruits MYC to chromatin to control the expression of genes linked to biomass accumulation. We further show that disrupting the MYC-WDR5 interaction within the context of an existing cancer promotes rapid and comprehensive tumor regression in vivo. These observations connect WDR5 to a core tumorigenic function of MYC and establish that, if a therapeutic window can be established, MYC-WDR5 inhibitors could be developed as anticancer agents.
- 19Thomas, L. R.; Adams, C. M.; Fesik, S. W.; Eischen, C. M.; Tansey, W. P. Targeting MYC through WDR5. Mol. Cell. Oncol. 2020, 7, 1709388, DOI: 10.1080/23723556.2019.1709388Google Scholar19Targeting MYC through WDR5Thomas Lance R; Tansey William P; Adams Clare M; Eischen Christine M; Fesik Stephen W; Tansey William PMolecular & cellular oncology (2020), 7 (2), 1709388 ISSN:2372-3556.The oncoprotein transcription factor MYC is overexpressed in most cancers and is responsible for hundreds of thousands of cancer deaths worldwide every year. MYC is also a highly validated - but currently undruggable - anti-cancer target. We recently showed that breaking the interaction of MYC with its chromatin co-factor WD repeat-containing protein 5 (WDR5) promotes tumor regression in mouse xenografts, laying the foundation for a new strategy to inhibit MYC in the clinic.
- 20Thomas, L. R.; Foshage, A. M.; Weissmiller, A. M.; Tansey, W. P. The MYC-WDR5 Nexus and Cancer. Cancer Res. 2015, 75, 4012– 4015, DOI: 10.1158/0008-5472.can-15-1216Google Scholar20The MYC-WDR5 Nexus and CancerThomas, Lance R.; Foshage, Audra M.; Weissmiller, April M.; Tansey, William P.Cancer Research (2015), 75 (19), 4012-4015CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)The MYC oncogenes encode a family of transcription factors that feature prominently in cancer. MYC proteins are overexpressed or deregulated in a majority of malignancies and drive tumorigenesis by inducing widespread transcriptional reprogramming that promotes cell proliferation, metab., and genomic instability. The ability of MYC to regulate transcription depends on its dimerization with MAX, which creates a DNA-binding domain that recognizes specific sequences in the regulatory elements of MYC target genes. Recently, we discovered that recognition of target genes by MYC also depends on its interaction with WDR5, a WD40-repeat protein that exists as part of several chromatin-regulatory complexes. Here, we discuss how interaction of MYC with WDR5 could create an avidity-based chromatin recognition mechanism that allows MYC to select its target genes in response to both genetic and epigenetic determinants. We rationalize how the MYC-WDR5 interaction provides plasticity in target gene selection by MYC and speculate on the biochem. and genomic contexts in which this interaction occurs. Finally, we discuss how properties of the MYC-WDR5 interface make it an attractive point for discovery of small-mol. inhibitors of MYC function in cancer cells.
- 21Guarnaccia, A. D.; Rose, K. L.; Wang, J.; Zhao, B.; Popay, T. M.; Wang, C. E.; Guerrazzi, K.; Hill, S.; Woodley, C. M.; Hansen, T. J.; Lorey, S. L.; Shaw, J. G.; Payne, W. G.; Weissmiller, A. M.; Olejniczak, E. T.; Fesik, S. W.; Liu, Q.; Tansey, W. P. Impact of WIN site inhibitor on the WDR5 interactome. Cell Rep. 2021, 34, 108636, DOI: 10.1016/j.celrep.2020.108636Google Scholar21Impact of WIN site inhibitor on the WDR5 interactomeGuarnaccia, Alissa D.; Rose, Kristie L.; Wang, Jing; Zhao, Bin; Popay, Tessa M.; Wang, Christina E.; Guerrazzi, Kiana; Hill, Salisha; Woodley, Chase M.; Hansen, Tyler J.; Lorey, Shelly L.; Shaw, J. Grace; Payne, William G.; Weissmiller, April M.; Olejniczak, Edward T.; Fesik, Stephen W.; Liu, Qi; Tansey, William P.Cell Reports (2021), 34 (3), 108636CODEN: CREED8; ISSN:2211-1247. (Cell Press)The chromatin-assocd. protein WDR5 is a promising pharmacol. target in cancer, with most drug discovery efforts directed against an arginine-binding cavity in WDR5 called the WIN site. Despite a clear expectation that WIN site inhibitors will alter the repertoire of WDR5 interaction partners, their impact on the WDR5 interactome remains unknown. Here, we use quant. proteomics to delineate how the WDR5 interactome is changed by WIN site inhibition. We show that the WIN site inhibitor alters the interaction of WDR5 with dozens of proteins, including those linked to phosphatidylinositol 3-kinase (PI3K) signaling. As proof of concept, we demonstrate that the master kinase PDPK1 is a bona fide high-affinity WIN site binding protein that engages WDR5 to modulate transcription of genes expressed in the G2 phase of the cell cycle. This dataset expands our understanding of WDR5 and serves as a resource for deciphering the action of WIN site inhibitors.
- 22Patel, A.; Dharmarajan, V.; Cosgrove, M. S. Structure of WDR5 bound to mixed lineage leukemia protein-1 peptide. J. Biol. Chem. 2008, 283, 32158– 32161, DOI: 10.1074/jbc.c800164200Google Scholar22Structure of WDR5 Bound to Mixed Lineage Leukemia Protein-1 PeptidePatel, Anamika; Dharmarajan, Venkatasubramanian; Cosgrove, Michael S.Journal of Biological Chemistry (2008), 283 (47), 32158-32161CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The mixed lineage leukemia protein-1 (MLL1) catalyzes histone H3 lysine 4 methylation and is regulated by interaction with WDR5 (WD-repeat protein-5), RbBP5 (retinoblastoma-binding protein-5), and the Ash2L (absent, small, homeotic disks-2-like) oncoprotein. In the accompanying investigation, we describe the identification of a conserved arginine contg. motif, called the "Win" or WDR5 interaction motif, that is essential for the assembly and H3K4 dimethylation activity of the MLL1 core complex. Here we present a 1.7-Å crystal structure of WDR5 bound to a peptide derived from the MLL1 Win motif. Our results show that Arg-3765 of MLL1 is bound in the same arginine binding pocket on WDR5 that was previously suggested to bind histone H3. Thermodn. binding expts. show that the MLL1 Win peptide is preferentially recognized by WDR5. These results are consistent with a model in which WDR5 recognizes Arg-3765 of MLL1, which is essential for the assembly and enzymic activity of the MLL1 core complex.
- 23Patel, A.; Vought, V. E.; Dharmarajan, V.; Cosgrove, M. S. A conserved arginine-containing motif crucial for the assembly and enzymatic activity of the mixed lineage leukemia protein-1 core complex. J. Biol. Chem. 2008, 283, 32162– 32175, DOI: 10.1074/jbc.m806317200Google Scholar23A Conserved Arginine-containing Motif Crucial for the Assembly and Enzymatic Activity of the Mixed Lineage Leukemia Protein-1 Core ComplexPatel, Anamika; Vought, Valarie E.; Dharmarajan, Venkatasubramanian; Cosgrove, Michael S.Journal of Biological Chemistry (2008), 283 (47), 32162-32175CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The mixed lineage leukemia protein-1 (MLL1) belongs to the SET1 family of histone H3 lysine 4 methyltransferases. Recent studies indicate that the catalytic subunits of SET1 family members are regulated by interaction with a conserved core group of proteins that include the WD repeat protein-5 (WDR5), retinoblastoma-binding protein-5 (RbBP5), and the absent small homeotic-2-like protein (Ash2L). It has been suggested that WDR5 functions to bridge the interactions between the catalytic and regulatory subunits of SET1 family complexes. However, the mol. details of these interactions are unknown. To gain insight into the interactions among these proteins, we have detd. the biophys. basis for the interaction between the human WDR5 and MLL1. Our studies reveal that WDR5 preferentially recognizes a previously unidentified and conserved arginine-contg. motif, called the "Win" or WDR5 interaction motif, which is located in the N-SET region of MLL1 and other SET1 family members. Surprisingly, our structural and functional studies show that WDR5 recognizes arginine 3765 of the MLL1 Win motif using the same arginine-binding pocket on WDR5 that was previously shown to bind histone H3. We demonstrate that WDR5's recognition of arginine 3765 of MLL1 is essential for the assembly and enzymic activity of the MLL1 core complex in vitro.
- 24Song, J.-J.; Kingston, R. E. WDR5 interacts with mixed lineage leukemia (MLL) protein via the histone H3-binding pocket. J. Biol. Chem. 2008, 283, 35258– 35264, DOI: 10.1074/jbc.m806900200Google Scholar24WDR5 Interacts with Mixed Lineage Leukemia (MLL) Protein via the Histone H3-binding PocketSong, Ji-Joon; Kingston, Robert E.Journal of Biological Chemistry (2008), 283 (50), 35258-35264CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)WDR5 is a component of the mixed lineage leukemia (MLL) complex, which methylates lysine 4 of histone H3, and was identified as a methylated Lys-4 histone H3-binding protein. Here, we present a crystal structure of WDR5 bound to an MLL peptide. Surprisingly, we find that WDR5 utilizes the same pocket shown to bind histone H3 for this MLL interaction. Furthermore, the WDR5-MLL interaction is disrupted preferentially by mono- and di-methylated Lys-4 histone H3 over unmodified and tri-methylated Lys-4 histone H3. These data implicate a delicate interplay between the effector, WDR5, the catalytic subunit, MLL, and the substrate, histone H3, of the MLL complex. We suggest that the activity of the MLL complex might be regulated through this interplay.
- 25Odho, Z.; Southall, S. M.; Wilson, J. R. Characterization of a novel WDR5-binding site that recruits RbBP5 through a conserved motif to enhance methylation of histone H3 lysine 4 by mixed lineage leukemia protein-1. J. Biol. Chem. 2010, 285, 32967– 32976, DOI: 10.1074/jbc.m110.159921Google Scholar25Characterization of a novel WDR5-binding site that recruits RbBP5 through a conserved motif to enhance methylation of histone H3 lysine 4 by mixed lineage leukemia protein-1Odho, Zain; Southall, Stacey M.; Wilson, Jon R.Journal of Biological Chemistry (2010), 285 (43), 32967-32976CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Histone modification is well established as a fundamental mechanism driving the regulation of transcription, replication, and DNA repair through the control of chromatin structure. Likewise, it is apparent that incorrect targeting of histone modifications contributes to misregulated gene expression and hence to developmental disorders and diseases of genomic instability such as cancer. The KMT2 family of SET domain methyltransferases, typified by mixed lineage leukemia protein-1 (MLL1), is responsible for histone H3 lysine 4 methylation, a marker of active genes. To ensure that this modification is correctly targeted, a multiprotein complex assocs. with the methyltransferase and directs activity. We have identified a novel interaction site on the core complex protein WD repeat protein-5 (WDR5), and we mapped the complementary site on its partner retinoblastoma-binding protein-5 (RbBP5). We have characterized this interaction by x-ray crystallog. and show how it is fundamental to the assembly of the complex and to the regulation of methyltransferase activity. We show which region of RbBP5 contributes directly to mixed lineage leukemia activation, and we combine our structural and biochem. data to produce a model to show how WDR5 and RbBP5 act cooperatively to stimulate activity.
- 26Chacón Simon, S.; Wang, F.; Thomas, L. R.; Phan, J.; Zhao, B.; Olejniczak, E. T.; Macdonald, J. D.; Shaw, J. G.; Schlund, C.; Payne, W.; Creighton, J.; Stauffer, S. R.; Waterson, A. G.; Tansey, W. P.; Fesik, S. W. Discovery of WD Repeat-Containing Protein 5 (WDR5)-MYC Inhibitors Using Fragment-Based Methods and Structure-Based Design. J. Med. Chem. 2020, 63, 4315– 4333, DOI: 10.1021/acs.jmedchem.0c00224Google Scholar26Discovery of WD Repeat-Containing Protein 5 (WDR5)-MYC Inhibitors Using Fragment-Based Methods and Structure-Based DesignChacon Simon, Selena; Wang, Feng; Thomas, Lance R.; Phan, Jason; Zhao, Bin; Olejniczak, Edward T.; MacDonald, Jonathan D.; Shaw, J. Grace; Schlund, Caden; Payne, William; Creighton, Joy; Stauffer, Shaun R.; Waterson, Alex G.; Tansey, William P.; Fesik, Stephen W.Journal of Medicinal Chemistry (2020), 63 (8), 4315-4333CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The frequent deregulation of MYC and its elevated expression via multiple mechanisms drives cells to a tumorigenic state. Indeed, MYC is overexpressed in up to ~ 50% of human cancers and is considered a highly validated anticancer target. Recently, we discovered that WD repeat-contg. protein 5 (WDR5) binds to MYC and is a crit. cofactor required for the recruitment of MYC to its target genes and reported the first small mol. inhibitors of the WDR5-MYC interaction using structure-based design. These compds. display high binding affinity, but have poor physicochem. properties and are hence not suitable for in vivo studies. Herein, we conducted an NMR-based fragment screening to identify addnl. chem. matter and, using a structure-based approach, we merged a fragment hit with the previously reported sulfonamide series. Compds. in this series can disrupt the WDR5-MYC interaction in cells, and as a consequence, we obsd. a redn. of MYC localization to chromatin.
- 27Gao, J.; Chang, M. T.; Johnsen, H. C.; Gao, S. P.; Sylvester, B. E.; Sumer, S. O.; Zhang, H.; Solit, D. B.; Taylor, B. S.; Schultz, N.; Sander, C. 3D clusters of somatic mutations in cancer reveal numerous rare mutations as functional targets. Genome Med. 2017, 9, 4, DOI: 10.1186/s13073-016-0393-xGoogle Scholar273D clusters of somatic mutations in cancer reveal numerous rare mutations as functional targetsGao, Jianjiong; Chang, Matthew T.; Johnsen, Hannah C.; Gao, Sizhi Paul; Sylvester, Brooke E.; Sumer, Selcuk Onur; Zhang, Hongxin; Solit, David B.; Taylor, Barry S.; Schultz, Nikolaus; Sander, ChrisGenome Medicine (2017), 9 (), 4/1-4/13CODEN: GMEECG; ISSN:1756-994X. (BioMed Central Ltd.)Many mutations in cancer are of unknown functional significance. Std. methods use statistically significant recurrence of mutations in tumor samples as an indicator of functional impact. We extend such analyses into the long tail of rare mutations by considering recurrence of mutations in clusters of spatially close residues in protein structures. Analyzing 10,000 tumor exomes, we identify more than 3000 rarely mutated residues in proteins as potentially functional and exptl. validate several in RAC1 and MAP2K1. These potential driver mutations (web resources: 3dhotspots.org and cBioPortal.org) can extend the scope of genomically informed clin. trials and of personalized choice of therapy.
- 28Acuner, S. E.; Sumbul, F.; Torun, H.; Haliloglu, T. Oncogenic mutations on Rac1 affect global intrinsic dynamics underlying GTP and PAK1 binding. Biophys. J. 2021, 120, 866– 876, DOI: 10.1016/j.bpj.2021.01.016Google Scholar28Oncogenic mutations on Rac1 affect global intrinsic dynamics underlying GTP and PAK1 bindingAcuner, Saliha Ece; Sumbul, Fidan; Torun, Hamdi; Haliloglu, TurkanBiophysical Journal (2021), 120 (5), 866-876CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)Rac1 is a small member of the Rho GTPase family. One of the most important downstream effectors of Rac1 is a serine/threonine kinase, p21-activated kinase 1 (PAK1). Mutational activation of PAK1 by Rac1 has oncogenic signaling effects. Here, although we focus on Rac1-PAK1 interaction by at.-force-microscopy-based single-mol. force spectroscopy expts., we explore the effect of active mutations on the intrinsic dynamics and binding interactions of Rac1 by Gaussian network model anal. and mol. dynamics simulations. We observe that Rac1 oncogenic mutations are at the hinges of three global modes of motion, suggesting the mech. changes as potential markers of oncogenicity. Indeed, the dissocn. of wild-type Rac1-PAK1 complex shows two distinct unbinding dynamic states that are reduced to one with constitutively active Q61L and oncogenic Y72C mutant Rac1, as revealed by single-mol. force spectroscopy expts. Q61L and Y72C mutations change the mechanics of the Rac1-PAK1 complex by increasing the elasticity of the protein and slowing down the transition to the unbound state. On the other hand, Rac1's intrinsic dynamics reveal more flexible GTP and PAK1-binding residues on switches I and II with Q61L, Y72C, oncogenic P29S and Q61R, and neg. T17N mutations. The cooperativity in the fluctuations of GTP-binding sites around the p-loop and switch I decreases in all mutants, mostly in Q61L, whereas some PAK1-binding residues display enhanced coupling with GTP-binding sites in Q61L and Y72C and within each other in P29S. The predicted binding free energies of the modeled Rac1-PAK1 complexes show that the change in the dynamic behavior likely means a more favorable PAK1 interaction. Overall, these findings suggest that the active mutations affect intrinsic functional dynamic events and alter the mechanics underlying the binding of Rac1 to GTP and upstream and downstream partners including PAK1.
- 29Forbes, S. A.; Beare, D.; Bindal, N.; Bamford, S.; Ward, S.; Cole, C. G.; Jia, M.; Kok, C.; Boutselakis, H.; De, T.; Sondka, Z.; Ponting, L.; Stefancsik, R.; Harsha, B.; Tate, J.; Dawson, E.; Thompson, S.; Jubb, H.; Campbell, P. J. COSMIC: High-Resolution Cancer Genetics Using the Catalogue of Somatic Mutations in Cancer. Curr. Protoc. Hum. Genet. 2016, 91, 10– 37, DOI: 10.1002/cphg.21Google Scholar29COSMIC: High-Resolution Cancer Genetics Using the Catalogue of Somatic Mutations in CancerForbes S A; Beare D; Bindal N; Bamford S; Ward S; Cole C G; Jia M; Kok C; Boutselakis H; De T; Sondka Z; Ponting L; Stefancsik R; Harsha B; Tate J; Dawson E; Thompson S; Jubb H; Campbell P JCurrent protocols in human genetics (2016), 91 (), 10.11.1-10.11.37 ISSN:.COSMIC (http://cancer.sanger.ac.uk) is an expert-curated database of somatic mutations in human cancer. Broad and comprehensive in scope, recent releases in 2016 describe over 4 million coding mutations across all human cancer disease types. Mutations are annotated across the entire genome, but expert curation is focused on over 400 key cancer genes. Now encompassing the majority of molecular mutation mechanisms in oncogenetics, COSMIC additionally describes 10 million non-coding mutations, 1 million copy-number aberrations, 9 million gene-expression variants, and almost 8 million differentially methylated CpGs. This information combines a consistent interpretation of the data from the major cancer genome consortia and cancer genome literature with exhaustive hand curation of over 22,000 gene-specific literature publications. This unit describes the graphical Web site in detail; alternative protocols overview other ways the entire database can be accessed, analyzed, and downloaded. © 2016 by John Wiley & Sons, Inc.
- 30Tate, J. G.; Bamford, S.; Jubb, H. C.; Sondka, Z.; Beare, D. M.; Bindal, N.; Boutselakis, H.; Cole, C. G.; Creatore, C.; Dawson, E.; Fish, P.; Harsha, B.; Hathaway, C.; Jupe, S. C.; Kok, C. Y.; Noble, K.; Ponting, L.; Ramshaw, C. C.; Rye, C. E.; Speedy, H. E.; Stefancsik, R.; Thompson, S. L.; Wang, S.; Ward, S.; Campbell, P. J.; Forbes, S. A. COSMIC: the Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res. 2019, 47, D941– d947, DOI: 10.1093/nar/gky1015Google Scholar30COSMIC: the Catalogue Of Somatic Mutations In CancerTate, John G.; Bamford, Sally; Jubb, Harry C.; Sondka, Zbyslaw; Beare, David M.; Bindal, Nidhi; Boutselakis, Harry; Cole, Charlotte G.; Creatore, Celestino; Dawson, Elisabeth; Fish, Peter; Harsha, Bhavana; Hathaway, Charlie; Jupe, Steve C.; Kok, Chai Yin; Noble, Kate; Ponting, Laura; Ramshaw, Christopher C.; Rye, Claire E.; Speedy, Helen E.; Stefancsik, Ray; Thompson, Sam L.; Wang, Shicai; Ward, Sari; Campbell, Peter J.; Forbes, Simon A.Nucleic Acids Research (2019), 47 (D1), D941-D947CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)COSMIC, the Catalog Of Somatic Mutations In Cancer (https://cancer.sanger.ac.uk) is the most detailed and comprehensive resource for exploring the effect of somatic mutations in human cancer. The latest release, COSMIC v86 (August 2018), includes almost 6 million coding mutations across 1.4 million tumor samples, curated from over 26,000 publications. In addn. to coding mutations, COSMIC covers all the genetic mechanisms by which somatic mutations promote cancer, including non-coding mutations, gene fusions, copy-no. variants and drug resistance mutations. COSMIC is primarily handcurated, ensuring quality, accuracy and descriptive data capture. Building on our manual curation processes, we are introducing new initiatives that allow us to prioritize key genes and diseases, and to react more quickly and comprehensively to new findings in the literature. Alongside improvements to the public website and data-download systems, new functionality in COSMIC-3D allows exploration of mutations within three-dimensional protein structures, their protein structural and functional impacts, and implications for druggability. In parallel with COSMIC's deep and broad variant coverage, the Cancer Gene Census (CGC) describes a curated catalog of genes driving every form of human cancer. Currently describing 719 genes, the CGC has recently introduced functional descriptions of how each gene drives disease, summarized into the 10 cancer Hallmarks.
- 31Torkamani, A.; Schork, N. J. Prediction of cancer driver mutations in protein kinases. Cancer Res. 2008, 68, 1675– 1682, DOI: 10.1158/0008-5472.can-07-5283Google Scholar31Prediction of Cancer Driver Mutations in Protein KinasesTorkamani, Ali; Schork, Nicholas J.Cancer Research (2008), 68 (6), 1675-1682CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)A large no. of somatic mutations accumulate during the process of tumorigenesis. A subset of these mutations contribute to tumor progression (known as "driver" mutations) whereas the majority of these mutations are effectively neutral (known as "passenger" mutations). The ability to differentiate between drivers and passengers will be crit. to the success of upcoming large-scale cancer DNA resequencing projects. Here we show a method capable of discriminating between drivers and passengers in the most frequently cancer-assocd. protein family, protein kinases. We apply this method to multiple cancer data sets, validating its accuracy by showing that it is capable of identifying known drivers, has excellent agreement with previous statistical ests. of the frequency of drivers, and provides strong evidence that predicted drivers are under pos. selection by various sequence and structural analyses. Furthermore, we identify particular positions in protein kinases that seem to play a role in oncogenesis. Finally, we provide a ranked list of candidate driver mutations.
- 32Ryslik, G. A.; Cheng, Y.; Modis, Y.; Zhao, H. Leveraging protein quaternary structure to identify oncogenic driver mutations. BMC Bioinf. 2016, 17, 137, DOI: 10.1186/s12859-016-0963-3Google Scholar32Leveraging protein quaternary structure to identify oncogenic driver mutationsRyslik, Gregory A.; Cheng, Yuwei; Modis, Yorgo; Zhao, HongyuBMC Bioinformatics (2016), 17 (), 137/1-137/13CODEN: BBMIC4; ISSN:1471-2105. (BioMed Central Ltd.)Background: Identifying key "driver" mutations which are responsible for tumorigenesis is crit. in the development of new oncol. drugs. Due to multiple pharmacol. successes in treating cancers that are caused by such driver mutations, a large body of methods have been developed to differentiate these mutations from the benign "passenger" mutations which occur in the tumor but do not further progress the disease. Under the hypothesis that driver mutations tend to cluster in key regions of the protein, the development of algorithms that identify these clusters has become a crit. area of research. Results: We have developed a novel methodol., QuartPAC (Quaternary Protein Amino acid Clustering), that identifies non-random mutational clustering while utilizing the protein quaternary structure in 3D space. By integrating the spatial information in the Protein Data Bank (PDB) and the mutational data in the Catalog of Somatic Mutations in Cancer (COSMIC), QuartPAC is able to identify clusters which are otherwise missed in a variety of proteins. The R package is available on Bioconductor at: http://bioconductor.jp/packages/3.1/bioc/html/QuartPAC.html. Conclusion:QuartPAC provides a unique tool to identify mutational clustering while accounting for the complete folded protein quaternary structure.
- 33Kumar, S.; Clarke, D.; Gerstein, M. B. Leveraging protein dynamics to identify cancer mutational hotspots using 3D structures. Proc. Natl. Acad. Sci. U.S.A. 2019, 116, 18962– 18970, DOI: 10.1073/pnas.1901156116Google Scholar33Leveraging protein dynamics to identify cancer mutational hotspots using 3D structuresKumar, Sushant; Clarke, Declan; Gerstein, Mark B.Proceedings of the National Academy of Sciences of the United States of America (2019), 116 (38), 18962-18970CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Large-scale exome sequencing of tumors has enabled the identification of cancer drivers using recurrence-based approaches. Some of these methods also employ 3D protein structures to identify mutational hotspots in cancer-assocd. genes. In detg. such mutational clusters in structures, existing approaches overlook protein dynamics, despite its essential role in protein function. We present a framework to identify cancer driver genes using a dynamics-based search of mutational hotspot communities. Mutations are mapped to protein structures, which are partitioned into distinct residue communities. These communities are identified in a framework where residue-residue contact edges are weighted by correlated motions (as inferred by dynamics-based models). We then search for signals of pos. selection among these residue communities to identify putative driver genes, while applying our method to the TCGA (The Cancer Genome Atlas) PanCancer Atlas missense mutation catalog. Overall, we predict 1 or more mutational hotspots within the resolved structures of proteins encoded by 434 genes. These genes were enriched among biol. processes assocd. with tumor progression. Addnl., a comparison between our approach and existing cancer hotspot detection methods using structural data suggests that including protein dynamics significantly increases the sensitivity of driver detection.
- 34Kamburov, A.; Lawrence, M. S.; Polak, P.; Leshchiner, I.; Lage, K.; Golub, T. R.; Lander, E. S.; Getz, G. Comprehensive assessment of cancer missense mutation clustering in protein structures. Proc. Natl. Acad. Sci. U.S.A. 2015, 112, E5486– E5495, DOI: 10.1073/pnas.1516373112Google Scholar34Comprehensive assessment of cancer missense mutation clustering in protein structuresKamburov, Atanas; Lawrence, Michael S.; Polak, Paz; Leshchiner, Ignaty; Lage, Kasper; Golub, Todd R.; Lander, Eric S.; Getz, GadProceedings of the National Academy of Sciences of the United States of America (2015), 112 (40), E5486-E5495CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Large-scale tumor sequencing projects enabled the identification of many new cancer gene candidates through computational approaches. Here, we describe a general method to detect cancer genes based on significant 3D clustering of mutations relative to the structure of the encoded protein products. The approach can also be used to search for proteins with an enrichment of mutations at binding interfaces with a protein, nucleic acid, or small mol. partner. We applied this approach to systematically analyze the PanCancer compendium of somatic mutations from 4,742 tumors relative to all known 3D structures of human proteins in the Protein Data Bank. We detected significant 3D clustering of missense mutations in several previously known oncoproteins including HRAS, EGFR, and PIK3CA. Although clustering of missense mutations is often regarded as a hallmark of oncoproteins, we obsd. that a no. of tumor suppressors, including FBXW7, VHL, and STK11, also showed such clustering. Beside these known cases, we also identified significant 3D clustering of missense mutations in NUF2, which encodes a component of the kinetochore, that could affect chromosome segregation and lead to aneuploidy. Anal. of interaction interfaces revealed enrichment of mutations in the interfaces between FBXW7-CCNE1, HRAS-RASA1, CUL4B-CAND1, OGT-HCFC1, PPP2R1A-PPP2R5C/PPP2R2A, DICER1-Mg2+, MAX-DNA, SRSF2-RNA, and others. Together, our results indicate that systematic consideration of 3D structure can assist in the identification of cancer genes and in the understanding of the functional role of their mutations.
- 35Dharmarajan, V.; Lee, J.-H.; Patel, A.; Skalnik, D. G.; Cosgrove, M. S. Structural basis for WDR5 interaction (Win) motif recognition in human SET1 family histone methyltransferases. J. Biol. Chem. 2012, 287, 27275– 27289, DOI: 10.1074/jbc.m112.364125Google Scholar35Structural Basis for WDR5 Interaction (Win) Motif Recognition in Human SET1 Family Histone MethyltransferasesDharmarajan, Venkatasubramanian; Lee, Jeong-Heon; Patel, Anamika; Skalnik, David G.; Cosgrove, Michael S.Journal of Biological Chemistry (2012), 287 (33), 27275-27289CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Translocations and amplifications of the mixed lineage leukemia-1 (MLL1) gene are assocd. with aggressive myeloid and lymphocytic leukemias in humans. MLL1 is a member of the SET1 family of histone H3 lysine 4 (H3K4) methyltransferases, which are required for transcription of genes involved in hematopoiesis and development. MLL1 assocs. with a subcomplex contg. WDR5, RbBP5, Ash2L, and DPY-30 (WRAD), which together form the MLL1 core complex that is required for sequential mono- and dimethylation of H3K4. We previously demonstrated that WDR5 binds the conserved WDR5 interaction (Win) motif of MLL1 in vitro, an interaction that is required for the H3K4 dimethylation activity of the MLL1 core complex. In this investigation, we demonstrate that arginine 3765 of the MLL1 Win motif is required to co-immunoppt. WRAD from mammalian cells, suggesting that the WDR5-Win motif interaction is important for the assembly of the MLL1 core complex in vivo. We also demonstrate that peptides that mimic SET1 family Win motif sequences inhibit H3K4 dimethylation by the MLL1 core complex with varying degrees of efficiency. To understand the structural basis for these differences, we detd. structures of WDR5 bound to six different naturally occurring Win motif sequences at resolns. ranging from 1.9 to 1.2 Å. Our results reveal that binding energy differences result from interactions between non-conserved residues C-terminal to the Win motif and to a lesser extent from subtle variation of residues within the Win motif. These results highlight a new class of methylation inhibitors that may be useful for the treatment of MLL1-related malignancies.
- 36Zhang, P.; Lee, H.; Brunzelle, J. S.; Couture, J.-F. The plasticity of WDR5 peptide-binding cleft enables the binding of the SET1 family of histone methyltransferases. Nucleic Acids Res. 2012, 40, 4237– 4246, DOI: 10.1093/nar/gkr1235Google Scholar36The plasticity of WDR5 peptide-binding cleft enables the binding of the SET1 family of histone methyltransferasesZhang, Pamela; Lee, Hwabin; Brunzelle, Joseph S.; Couture, Jean-FrancoisNucleic Acids Research (2012), 40 (9), 4237-4246CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)In mammals, the SET1 family of lysine methyltransferases (KMTs), which includes MLL1-5, SET1A and SET1B, catalyzes the methylation of lysine-4 (Lys-4) on histone H3. Recent reports have demonstrated that a three-subunit complex composed of WD-repeat protein-5 (WDR5), retinoblastoma-binding protein-5 (RbBP5) and absent, small, homeotic disks-2-like (ASH2L) stimulates the methyltransferase activity of MLL1. On the basis of studies showing that this stimulation is in part controlled by an interaction between WDR5 and a small region located in close proximity of the MLL1 catalytic domain [referred to as the WDR5-interacting motif (Win)], it has been suggested that WDR5 might play an analogous role in scaffolding the other SET1 complexes. We herein provide biochem. and structural evidence showing that WDR5 binds the Win motifs of MLL2-4, SET1A and SET1B. Comparative anal. of WDR5-Win complexes reveals that binding of the Win motifs is achieved by the plasticity of WDR5 peptidyl-arginine-binding cleft allowing the C-terminal ends of the Win motifs to be maintained in structurally divergent conformations. Consistently, enzymic assays reveal that WDR5 plays an important role in the optimal stimulation of MLL2-4, SET1A and SET1B methyltransferase activity by the RbBP5-ASH2L heterodimer. Overall, our findings illustrate the function of WDR5 in scaffolding the SET1 family of KMTs and further emphasize on the important role of WDR5 in regulating global histone H3 Lys-4 methylation.
- 37Muntean, A. G.; Hess, J. L. The pathogenesis of mixed-lineage leukemia. Annu. Rev. Pathol.: Mech. Dis. 2012, 7, 283– 301, DOI: 10.1146/annurev-pathol-011811-132434Google Scholar37The pathogenesis of mixed-lineage leukemiaMuntean, Andrew G.; Hess, Jay L.Annual Review of Pathology: Mechanisms of Disease (2012), 7 (), 283-301CODEN: ARPMCU; ISSN:1553-4006. (Annual Reviews Inc.)A review. Aggressive leukemias arise in both children and adults as a result of rearrangements to the mixed-lineage leukemia gene (MLL) located on chromosome 11q23. MLL encodes a large histone methyltransferase that directly binds DNA and pos. regulates gene transcription, including homeobox (HOX) genes. MLL is involved in chromosomal translocations, partial tandem duplications, and amplifications, all of which result in hematopoietic malignancies due to sustained HOX expression and stalled differentiation. MLL lesions are assocd. with both acute myeloid leukemia and acute lymphoid leukemia and are usually assocd. with a relatively poor prognosis despite improved treatment options such as allogeneic hematopoietic stem cell transplantation, which underscores the need for new treatment regimens. Recent advances have begun to reveal the mol. mechanisms that drive MLL-assocd. leukemias, which, in turn, have provided opportunities for therapeutic development. Here, we discuss the etiol. of MLL leukemias and potential directions for future therapy.
- 38Chen, X.; Xie, W.; Gu, P.; Cai, Q.; Wang, B.; Xie, Y.; Dong, W.; He, W.; Zhong, G.; Lin, T.; Huang, J. Upregulated WDR5 promotes proliferation, self-renewal and chemoresistance in bladder cancer via mediating H3K4 trimethylation. Sci. Rep. 2015, 5, 8293, DOI: 10.1038/srep08293Google Scholar38Upregulated WDR5 promotes proliferation, self-renewal and chemoresistance in bladder cancer via mediating H3K4 trimethylationChen, Xu; Xie, Weibin; Gu, Peng; Cai, Qingqing; Wang, Bo; Xie, Yun; Dong, Wen; He, Wang; Zhong, Guangzheng; Lin, Tianxin; Huang, JianScientific Reports (2015), 5 (), 8293CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)WD repeat domain 5 (WDR5) plays an important role in various biol. functions through the epigenetic regulation of gene transcription; however, its role in bladder cancer remains largely unknown. Our study investigated the role of WDR5 in bladder cancer and demonstrated that WDR5 was upregulated in bladder cancer tissues, and elevated WDR5 protein levels pos. correlated with advanced tumor stage and poor survival. Through gain or loss of function, we demonstrated that WDR5 promoted proliferation, self-renewal and chemoresistance to cisplatin in bladder cancer cells in vitro, and tumor growth in vivo. Mechanistically, WDR5 regulated various functions in bladder cancer by mediating the transcription of cyclin B1, cyclin E1, cyclin E2, UHMK1, MCL1, BIRC3 and Nanog by histone H3 lysine 4 trimethylation. Therefore, we have discovered that WDR5 plays an important role in bladder cancer suggesting that WDR5 is a potential biomarker and a promising target in the treatment of bladder cancer.
- 39Chen, X.; Gu, P.; Li, K.; Xie, W.; Chen, C.; Lin, T.; Huang, J. Gene expression profiling of WDR5 regulated genes in bladder cancer. Genomics Data 2015, 5, 27– 29, DOI: 10.1016/j.gdata.2015.05.003Google Scholar39Gene expression profiling of WDR5 regulated genes in bladder cancerChen Xu; Gu Peng; Lin Tianxin; Li Kuiqing; Xie Weibin; Chen Changhao; Huang JianGenomics data (2015), 5 (), 27-9 ISSN:2213-5960.WD repeat domain 5 (WDR5) plays an important role in various biological functions through the epigenetic regulation of gene transcription (Wysocka et al., 2005 [1]; Sandstrom et al., 2014[2]; Ang et al., 2011[3]). Recently, our study found that WDR5 was upregulated in bladder cancer tissues, promoted bladder cancer cell proliferation, self-renewal and chemoresistance to cisplatin in bladder cancer cells in vitro, and tumor growth in vivo (Chen et al., 2015). To gain a molecular understanding of the role of WDR5 in promoting bladder cancer, we performed a genome-wide analysis on WDR5 knockdown by microarray gene expression profiling. Here we provide detailed experimental methods and analysis for the microarray data, which have been deposited into Gene Expression Omnibus (GEO): GSE59132.
- 40Ge, Z.; Song, E. J.; Kawasawa, Y. I.; Li, J.; Dovat, S.; Song, C. WDR5 high expression and its effect on tumorigenesis in leukemia. Oncotarget 2016, 7, 37740– 37754, DOI: 10.18632/oncotarget.9312Google Scholar40WDR5 high expression and its effect on tumorigenesis in leukemiaGe Zheng; Ge Zheng; Song Evelyn J; Dovat Sinisa; Song Chunhua; Ge Zheng; Li Jianyong; Kawasawa Yuka ImamuraOncotarget (2016), 7 (25), 37740-37754 ISSN:.WD repeat domain 5 (WDR5) plays an important role in various biological functions through the epigenetic regulation of gene transcription. However, the oncogenic effect of WDR5 in leukemia remains largely unknown. Here, we found WDR5 expression is increased in leukemia patients. High expression of WDR5 is associated with high risk leukemia; Patients with WDR5 and MLL1 high expression have poor complete remission rate. We further identified the global genomic binding of WDR5 in leukemic cells and found the genomic co-localization of WDR5 binding with H3K4me3 enrichment. Moreover, WDR5 knockdown by shRNA suppresses cell proliferation, induces apoptosis, inhibits the expression of WDR5 targets, and blocks the H3K4me3 enrichment on the promoter of its targets. We also observed the positive correlation of WDR5 expression with these targets in the cohort study of leukemia patients. Our data reveal that WDR5 may have oncogenic effect and WDR5-mediated H3K4 methylation plays an important role in leukemogenesis.
- 41Sun, W.; Guo, F.; Liu, M. Up-regulated WDR5 promotes gastric cancer formation by induced cyclin D1 expression. J. Cell. Biochem. 2018, 119, 3304– 3316, DOI: 10.1002/jcb.26491Google Scholar41Up-regulated WDR5 promotes gastric cancer formation by induced cyclin D1 expressionSun, Wei; Guo, Fuchao; Liu, MingkaiJournal of Cellular Biochemistry (2018), 119 (4), 3304-3316CODEN: JCEBD5; ISSN:0730-2312. (Wiley-Blackwell)Gastric cancer (GC) is the fourth common cancer and second leading cause of cancer-related mortality in the world. WD repeat domain 5 (WDR5) has been identified that its functions as an important role in various biol. functions through the epigenetic regulation of gene transcription. However, the oncogenic effect of WDR5 in gastric cancer remains largely unknown. In this study, we investigated the role of WDR5 in gastric cancer genesis. We found that WDR5 expression is increased in gastric cancer patients. Through survival anal., we found that high expression of WDR5 is assocd. with high risk gastric cancer; patients who with WDR5 high expression have poor survival rate compared with those who with WDR5 low expression. To make further investigation, we identified that WDR5 is targeted for cell cycle arrest by the Cyclin D1 in a process that is regulated by H3K4me3. Moreover, over-expression of WDR5 promotes cell proliferation, induces S/G2/M arrest in cell cycle, and promotes the expression of WDR5 targets, as well as that of H3K4me3 on the promoter of its targets. Inversely, WDR5 knockdown by shRNA inhibits cell proliferation, reverses S/G2/M arrest in cell cycle, and suppresses the expression of WDR5 targets, as well as that of H3K4me3. We also obsd. the pos. correlation of WDR5 expression with its target in the cohort study of gastric patients. Taken together, our data reveal that WDR5 may have oncogenic effect and WDR5-mediated H3K4 methylation plays an important role in gastric cancer.
- 42Wang, F.; Zhang, J.; Ke, X.; Peng, W.; Zhao, G.; Peng, S.; Xu, J.; Xu, B.; Cui, H. WDR5-Myc axis promotes the progression of glioblastoma and neuroblastoma by transcriptional activating CARM1. Biochem. Biophys. Res. Commun. 2020, 523, 699– 706, DOI: 10.1016/j.bbrc.2019.12.101Google Scholar42WDR5-Myc axis promotes the progression of glioblastoma and neuroblastoma by transcriptional activating CARM1Wang, Feng; Zhang, Jiayi; Ke, Xiaoxue; Peng, Wen; Zhao, Gaichao; Peng, Shihan; Xu, Jie; Xu, Bo; Cui, HongjuanBiochemical and Biophysical Research Communications (2020), 523 (3), 699-706CODEN: BBRCA9; ISSN:0006-291X. (Elsevier B.V.)The WD repeat domain 5 (WDR5), also known as SWD3 and BIG-3, is often overexpressed in cancers, however its mol. function in cancer remains to be elucidated. In this study, we found that WDR5 promoted the proliferation and self-renewal of glioblastoma and neuroblastoma cells. The data from databases and Western blot assay showed that CARM1 is a downstream gene of WDR5-Myc axis. In addn., we obsd. that WDR5 promoted the binding of Myc to CARM1 promoter by interacting with Myc and inducing histone 3 lysine 4 trimethylation (H3K4me3). Dual luciferase reporter system indicated that Myc binds to the upstream region (-520 to -515) before transcription start site (TSS) of CARM1 promoter. These findings suggest a novel regulatory model for the proliferation and tumorigenesis of glioblastoma and neuroblastoma by WDR5-Myc axis.
- 43Bryan, A. F.; Wang, J.; Howard, G. C.; Guarnaccia, A. D.; Woodley, C. M.; Aho, E. R.; Rellinger, E. J.; Matlock, B. K.; Flaherty, D. K.; Lorey, S. L.; Chung, D. H.; Fesik, S. W.; Liu, Q.; Weissmiller, A. M.; Tansey, W. P. WDR5 is a conserved regulator of protein synthesis gene expression. Nucleic Acids Res. 2020, 48, 2924– 2941, DOI: 10.1093/nar/gkaa051Google Scholar43WDR5 is a conserved regulator of protein synthesis gene expressionBryan, Audra F.; Wang, Jing; Howard, Gregory C.; Guarnaccia, Alissa D.; Woodley, Chase M.; Aho, Erin R.; Rellinger, Eric J.; Matlock, Brittany K.; Flaherty, David K.; Lorey, Shelly L.; Chung, Dai H.; Fesik, Stephen W.; Liu, Qi; Weissmiller, April M.; Tansey, William P.Nucleic Acids Research (2020), 48 (6), 2924-2941CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)WDR5 is a highly-conserved nuclear protein that performs multiple scaffolding functions in the context of chromatin. WDR5 is also a promising target for pharmacol. inhibition in cancer, with small mol. inhibitors of an arginine-binding pocket of WDR5 (the 'WIN' site) showing efficacy against a range of cancer cell lines in vitro. Efforts to understand WDR5, or establish the mechanism of action of WIN site inhibitors, however, are stymied by its many functions in the nucleus, and a lack of knowledge of the conserved gene networks--if any--that are under its control. Here, we have performed comparative genomic analyses to identify the conserved sites of WDR5 binding to chromatin, and the conserved genes regulated by WDR5, across a diverse panel of cancer cell lines. We show that a specific cohort of protein synthesis genes (PSGs) are invariantly bound by WDR5, demonstrate that the WIN site anchors WDR5 to chromatin at these sites, and establish that PSGs are bona fide, acute, and persistent targets of WIN site blockade. Together, these data reveal that WDR5 plays a predominant transcriptional role in biomass accumulation and provide further evidence that WIN site inhibitors act to repress gene networks linked to protein synthesis homeostasis. In Saccharomyces cerevisiae, ribosomal protein gene (RPG) promoters display binding sites for either Rap1 or Abf1 transcription factors. Unlike Rap1-assocd. promoters, the small cohort of Abf1-dependent RPGs (Abf1-RPGs) has not been extensively investigated. We show that RPL3, RPL4B, RPP1A, RPS22B and RPS28A/B share a common promoter architecture, with an Abf1 site upstream of a conserved element matching the sequence recognized by Fhl1, a transcription factor which together with Ifh1 orchestrates Rap1-assocd. RPG regulation. Abf1 and Fhl1 promoter assocn. was confirmed by ChIP and/or gel retardation assays. Mutational anal. revealed a more severe requirement of Abf1 than Fhl1 binding sites for RPG transcription. In the case of RPS22B an unusual Tbf1 binding site promoted both RPS22B and intron-hosted SNR44 expression. Abf1-RPG down-regulation upon TOR pathway inhibition was much attenuated at defective mutant promoters unable to bind Abf1. TORC1 inactivation caused the expected redn. of Ifh1 occupancy at RPS22B and RPL3 promoters, but unexpectedly it entailed largely increased Abf1 assocn. with Abf1-RPG promoters. We present evidence that Abf1 recruitment upon nutritional stress, also obsd. for representative ribosome biogenesis genes, favors RPG transcriptional rescue upon nutrient replenishment, thus pointing to nutrient-regulated Abf1 dynamics at promoters as a novel mechanism in ribosome biogenesis control.
- 44Imran, A.; Moyer, B. S.; Canning, A. J.; Kalina, D.; Duncan, T. M.; Moody, K. J.; Wolfe, A. J.; Cosgrove, M. S.; Movileanu, L. Kinetics of the multitasking high-affinity Win binding site of WDR5 in restricted and unrestricted conditions. Biochem. J. 2021, 478, 2145– 2161, DOI: 10.1042/bcj20210253Google Scholar44Kinetics of the multitasking high-affinity Win binding site of WDR5 in restricted and unrestricted conditionsImran, Ali; Moyer, Brandon S.; Canning, Ashley J.; Kalina, Dan; Duncan, Thomas M.; Moody, Kelsey J.; Wolfe, Aaron J.; Cosgrove, Michael S.; Movileanu, LiviuBiochemical Journal (2021), 478 (11), 2145-2161CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)Recent advances in quant. proteomics show that WD40 proteins play a pivotal role in numerous cellular networks. Yet, they have been fairly unexplored and their phys. assocns. with other proteins are ambiguous. A quant. understanding of these interactions has wide-ranging significance. WD40 repeat protein 5 (WDR5) interacts with all members of human SET1/MLL methyltransferases, which regulate methylation of the histone 3 lysine 4 (H3K4). Here, using real-time binding measurements in a high-throughput setting, we identified the kinetic fingerprint of transient assocns. between WDR5 and 14-residue WDR5 interaction (Win) motif peptides of each SET1 protein (SET1Win). Our results reveal that the high-affinity WDR5-SET1Win interactions feature slow assocn. kinetics. This finding is likely due to the requirement of SET1Win to insert into the narrow WDR5 cavity, also named the Win binding site. Furthermore, our explorations indicate fairly slow dissocn. kinetics. This conclusion is in accordance with the primary role of WDR5 in maintaining the functional integrity of a large multisubunit complex, which regulates the histone methylation. Because the Win binding site is considered a key therapeutic target, the immediate outcomes of this study could form the basis for accelerated developments in medical biotechnol.
- 45Mayse, L. A.; Imran, A.; Larimi, M. G.; Cosgrove, M. S.; Wolfe, A. J.; Movileanu, L. Disentangling the recognition complexity of a protein hub using a nanopore. Nat. Commun. 2022, 13, 978, DOI: 10.1038/s41467-022-28465-8Google Scholar45Disentangling the recognition complexity of a protein hub using a nanoporeMayse, Lauren Ashley; Imran, Ali; Larimi, Motahareh Ghahari; Cosgrove, Michael S.; Wolfe, Aaron James; Movileanu, LiviuNature Communications (2022), 13 (1), 978CODEN: NCAOBW; ISSN:2041-1723. (Nature Portfolio)WD40 repeat proteins are frequently involved in processing cell signaling and scaffolding large multi-subunit machineries. Despite their significance in physiol. and disease-like conditions, their reversible interactions with other proteins remain modestly examd. Here, we show the development and validation of a protein nanopore for the detection and quantification of WD40 repeat protein 5 (WDR5), a chromatin-assocd. hub involved in epigenetic regulation of histone methylation. Our nanopore sensor is equipped with a 14-residue Win motif of mixed lineage leukemia 4 methyltransferase (MLL4Win), a WDR5 ligand. Our approach reveals a broad dynamic range of MLL4Win-WDR5 interactions and three distant subpopulations of binding events, representing three modes of protein recognition. The three binding events are confirmed as specific interactions using a weakly binding WDR5 deriv. and various environmental contexts. These outcomes demonstrate the substantial sensitivity of our nanopore sensor, which can be utilized in protein analytics.
- 46Karatas, H.; Townsend, E. C.; Cao, F.; Chen, Y.; Bernard, D.; Liu, L.; Lei, M.; Dou, Y.; Wang, S. High-affinity, small-molecule peptidomimetic inhibitors of MLL1/WDR5 protein-protein interaction. J. Am. Chem. Soc. 2013, 135, 669– 682, DOI: 10.1021/ja306028qGoogle Scholar46High-Affinity, Small-Molecule Peptidomimetic Inhibitors of MLL1/WDR5 Protein-Protein InteractionKaratas, Hacer; Townsend, Elizabeth C.; Cao, Fang; Chen, Yong; Bernard, Denzil; Liu, Liu; Lei, Ming; Dou, Yali; Wang, ShaomengJournal of the American Chemical Society (2013), 135 (2), 669-682CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Mixed lineage leukemia 1 (MLL1) is a histone H3 lysine 4 (H3K4) methyltransferase, and targeting the MLL1 enzymic activity has been proposed as a novel therapeutic strategy for the treatment of acute leukemia harboring MLL1 fusion proteins. The MLL1/WDR5 protein-protein interaction is essential for MLL1 enzymic activity. In the present study, we designed a large no. of peptidomimetics to target the MLL1/WDR5 interaction based upon -CO-ARA-NH-, the min. binding motif derived from MLL1. Our study led to the design of high-affinity peptidomimetics, which bind to WDR5 with Ki < 1 nM and function as potent antagonists of MLL1 activity in a fully reconstituted in vitro H3K4 methyltransferase assay. Detn. of co-crystal structures of two potent peptidomimetics in complex with WDR5 establishes their structural basis for high-affinity binding to WDR5. Evaluation of one such peptidomimetic, MM-102, in bone marrow cells transduced with MLL1-AF9 fusion construct shows that the compd. effectively decreases the expression of HoxA9 and Meis-1, two crit. MLL1 target genes in MLL1 fusion protein mediated leukemogenesis. MM-102 also specifically inhibits cell growth and induces apoptosis in leukemia cells harboring MLL1 fusion proteins. Our study provides the first proof-of-concept for the design of small-mol. inhibitors of the WDR5/MLL1 protein-protein interaction as a novel therapeutic approach for acute leukemia harboring MLL1 fusion proteins.
- 47Cao, F.; Townsend, E. C.; Karatas, H.; Xu, J.; Li, L.; Lee, S.; Liu, L.; Chen, Y.; Ouillette, P.; Zhu, J.; Hess, J. L.; Atadja, P.; Lei, M.; Qin, Z. S.; Malek, S.; Wang, S.; Dou, Y. Targeting MLL1 H3K4 methyltransferase activity in mixed-lineage leukemia. Mol. Cell 2014, 53, 247– 261, DOI: 10.1016/j.molcel.2013.12.001Google Scholar47Targeting MLL1 H3K4 Methyltransferase Activity in Mixed-Lineage LeukemiaCao, Fang; Townsend, Elizabeth C.; Karatas, Hacer; Xu, Jing; Li, Li; Lee, Shirley; Liu, Liu; Chen, Yong; Ouillette, Peter; Zhu, Jidong; Hess, Jay L.; Atadja, Peter; Lei, Ming; Qin, Zhaohui S.; Malek, Sami; Wang, Shaomeng; Dou, YaliMolecular Cell (2014), 53 (2), 247-261CODEN: MOCEFL; ISSN:1097-2765. (Elsevier Inc.)Here we report a comprehensive characterization of our recently developed inhibitor MM-401 that targets the MLL1 H3K4 methyltransferase activity. MM-401 is able to specifically inhibit MLL1 activity by blocking MLL1-WDR5 interaction and thus the complex assembly. This targeting strategy does not affect other mixed-lineage leukemia (MLL) family histone methyltransferases (HMTs), revealing a unique regulatory feature for the MLL1 complex. Using MM-401 and its enantiomer control MM-NC-401, we show that inhibiting MLL1 methyltransferase activity specifically blocks proliferation of MLL cells by inducing cell-cycle arrest, apoptosis, and myeloid differentiation without general toxicity to normal bone marrow cells or non-MLL cells. More importantly, transcriptome analyses show that MM-401 induces changes in gene expression similar to those of MLL1 deletion, supporting a predominant role of MLL1 activity in regulating MLL1-dependent leukemia transcription program. We envision broad applications for MM-401 in basic and translational research.
- 48Alicea-Velázquez, N. L.; Shinsky, S. A.; Loh, D. M.; Lee, J. H.; Skalnik, D. G.; Cosgrove, M. S. Targeted Disruption of the Interaction between WD-40 Repeat Protein 5 (WDR5) and Mixed Lineage Leukemia (MLL)/SET1 Family Proteins Specifically Inhibits MLL1 and SETd1A Methyltransferase Complexes. J. Biol. Chem. 2016, 291, 22357– 22372, DOI: 10.1074/jbc.M116.752626Google Scholar48Targeted Disruption of the Interaction between WD-40 Repeat Protein 5 (WDR5) and Mixed Lineage Leukemia (MLL)/SET1 Family Proteins Specifically Inhibits MLL1 and SETd1A Methyltransferase ComplexesAlicea-Velazquez, Nilda L.; Shinsky, Stephen A.; Loh, Daniel M.; Lee, Jeong-Heon; Skalnik, David G.; Cosgrove, Michael S.Journal of Biological Chemistry (2016), 291 (43), 22357-22372CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)MLL1 belongs to the SET1 family of histone H3 lysine 4 (H3K4) methyltransferases, composed of MLL1-4 and SETd1A/B. MLL1 translocations are present in acute leukemias, and mutations in several family members are assocd. with cancer and developmental disorders. MLL1 assocs. with a subcomplex contg. WDR5, RbBP5, ASH2L, and DPY-30 (WRAD), forming the MLL1 core complex required for H3K4 mono- and dimethylation and transcriptional activation. Core complex assembly requires interaction of WDR5 with the MLL1 Win (WDR5 interaction) motif, which is conserved across the SET1 family. Agents that mimic the SET1 family Win motif inhibit the MLL1 core complex and have become an attractive approach for targeting MLL1 in cancers. Like MLL1, other SET1 family members interact with WRAD, but the roles of the Win motif in complex assembly and enzymic activity remain unexplored. Here, we show that the Win motif is necessary for interaction of WDR5 with all members of the human SET1 family. Mutation of the Win motif-WDR5 interface severely disrupts assembly and activity of MLL1 and SETd1A complexes but only modestly disrupts MLL2/4 and SETd1B complexes without significantly altering enzymic activity in vitro. Notably, in the absence of WDR5, MLL3 interacts with RAD and shows enhanced activity. To further probe the role of the Win motif-WDR5 interaction, we designed a peptidomimetic that binds WDR5 (Kd ∼3 nm) and selectively inhibits activity of MLL1 and SETd1A core complexes within the SET1 family. Our results reveal that SET1 family complexes with the weakest Win motif-WDR5 interaction are more susceptible to Win motif-based inhibitors.
- 49Karatas, H.; Li, Y.; Liu, L.; Ji, J.; Lee, S.; Chen, Y.; Yang, J.; Huang, L.; Bernard, D.; Xu, J.; Townsend, E. C.; Cao, F.; Ran, X.; Li, X.; Wen, B.; Sun, D.; Stuckey, J. A.; Lei, M.; Dou, Y.; Wang, S. Discovery of a Highly Potent, Cell-Permeable Macrocyclic Peptidomimetic (MM-589) Targeting the WD Repeat Domain 5 Protein (WDR5)-Mixed Lineage Leukemia (MLL) Protein-Protein Interaction. J. Med. Chem. 2017, 60, 4818– 4839, DOI: 10.1021/acs.jmedchem.6b01796Google Scholar49Discovery of a Highly Potent, Cell-Permeable Macrocyclic Peptidomimetic (MM-589) Targeting the WD Repeat Domain 5 Protein (WDR5)-Mixed Lineage Leukemia (MLL) Protein-Protein InteractionKaratas, Hacer; Li, Yangbing; Liu, Liu; Ji, Jiao; Lee, Shirley; Chen, Yong; Yang, Jiuling; Huang, Liyue; Bernard, Denzil; Xu, Jing; Townsend, Elizabeth C.; Cao, Fang; Ran, Xu; Li, Xiaoqin; Wen, Bo; Sun, Duxin; Stuckey, Jeanne A.; Lei, Ming; Dou, Yali; Wang, ShaomengJournal of Medicinal Chemistry (2017), 60 (12), 4818-4839CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)We report herein the design, synthesis and evaluation of macrocyclic peptidomimetics that bind to WDR5 and block the WDR5-MLL protein-protein interaction. Compd. (I) (R1 = Me; n = 1) (MM-589) binds to WDR5 with an IC50 value of 0.90 nM (Ki value <1 nM), and inhibits the MLL H3K4 methyltransferase (HMT) activity with an IC50 value of 12.7 nM. Compd. I (R1 = Me; n = 1) potently and selectively inhibits cell growth in human leukemia cell lines harboring MLL translocations, and is >40 times better than the previously reported compd. MM-401. Co-crystal structures of I (R1 = H, n = 5) and (R1 = Me; n = 1) complexed with WDR5 provide structural basis for their high affinity binding to WDR5. Addnl., we have developed and optimized a new, AlphaLISA-based MLL HMT functional assay to facilitate the functional evaluation of these designed compds. Compd. I (R1 = Me; n = 1) represents the most potent inhibitor of the WDR5-MLL interaction reported to date and further optimization of I (R1 = Me; n = 1) may yield a new therapy for acute leukemia.
- 50Schapira, M.; Tyers, M.; Torrent, M.; Arrowsmith, C. H. WD40 repeat domain proteins: a novel target class?. Nat. Rev. Drug Discovery 2017, 16, 773– 786, DOI: 10.1038/nrd.2017.179Google Scholar50WD40 repeat domain proteins: a novel target class?Schapira, Matthieu; Tyers, Mike; Torrent, Maricel; Arrowsmith, Cheryl H.Nature Reviews Drug Discovery (2017), 16 (11), 773-786CODEN: NRDDAG; ISSN:1474-1776. (Nature Research)Antagonism of protein-protein interactions (PPIs) with small mols. is becoming more feasible as a therapeutic approach. Successful PPI inhibitors tend to target proteins contg. deep peptide-binding grooves or pockets rather than the more common large, flat protein interaction surfaces. Here, we review one of the most abundant PPI domains in the human proteome, the WD40 repeat (WDR) domain, which has a central peptide-binding pocket and is a member of the β-propeller domain-contg. protein family. Recently, two WDR domain-contg. proteins, WDR5 and EED, as well as other β-propeller domains have been successfully targeted by potent, specific, cell-active, drug-like chem. probes. Could WDR domains be a novel target class for drug discovery. Although the research is at an early stage and therefore not clin. validated, cautious optimism is justified, as WDR domain-contg. proteins are involved in multiple disease-assocd. pathways. The druggability and structural diversity of WDR domain binding pockets suggest that understanding how to target this prevalent domain class will open up areas of disease biol. that have so far resisted drug discovery efforts.
- 51Aho, E. R.; Weissmiller, A. M.; Fesik, S. W.; Tansey, W. P. Targeting WDR5: A WINning Anti-Cancer Strategy?. Epigenet. Insights 2019, 12, 2516865719865282, DOI: 10.1177/2516865719865282Google ScholarThere is no corresponding record for this reference.
- 52Aho, E. R.; Wang, J.; Gogliotti, R. D.; Howard, G. C.; Phan, J.; Acharya, P.; Macdonald, J. D.; Cheng, K.; Lorey, S. L.; Lu, B.; Wenzel, S.; Foshage, A. M.; Alvarado, J.; Wang, F.; Shaw, J. G.; Zhao, B.; Weissmiller, A. M.; Thomas, L. R.; Vakoc, C. R.; Hall, M. D.; Hiebert, S. W.; Liu, Q.; Stauffer, S. R.; Fesik, S. W.; Tansey, W. P. Displacement of WDR5 from Chromatin by a WIN Site Inhibitor with Picomolar Affinity. Cell Rep. 2019, 26, 2916– 2928.e13, DOI: 10.1016/j.celrep.2019.02.047Google Scholar52Displacement of WDR5 from Chromatin by a WIN Site Inhibitor with Picomolar AffinityAho, Erin R.; Wang, Jing; Gogliotti, Rocco D.; Howard, Gregory C.; Phan, Jason; Acharya, Pankaj; Macdonald, Jonathan D.; Cheng, Ken; Lorey, Shelly L.; Lu, Bin; Wenzel, Sabine; Foshage, Audra M.; Alvarado, Joseph; Wang, Feng; Shaw, J. Grace; Zhao, Bin; Weissmiller, April M.; Thomas, Lance R.; Vakoc, Christopher R.; Hall, Matthew D.; Hiebert, Scott W.; Liu, Qi; Stauffer, Shaun R.; Fesik, Stephen W.; Tansey, William P.Cell Reports (2019), 26 (11), 2916-2928.e13CODEN: CREED8; ISSN:2211-1247. (Cell Press)The chromatin-assocd. protein WDR5 is a promising target for pharmacol. inhibition in cancer. Drug discovery efforts center on the blockade of the "WIN site" of WDR5, a well-defined pocket that is amenable to small mol. inhibition. Various cancer contexts have been proposed to be targets for WIN site inhibitors, but a lack of understanding of WDR5 target genes and of the primary effects of WIN site inhibitors hampers their utility. Here, by the discovery of potent WIN site inhibitors, we demonstrate that the WIN site links WDR5 to chromatin at a small cohort of loci, including a specific subset of ribosome protein genes. WIN site inhibitors rapidly displace WDR5 from chromatin and decrease the expression of assocd. genes, causing translational inhibition, nucleolar stress, and p53 induction. Our studies define a mode by which WDR5 engages chromatin and forecast that WIN site blockade could have utility against multiple cancer types.
- 53Dennis, M. L.; Morrow, B. J.; Dolezal, O.; Cuzzupe, A. N.; Stupple, A. E.; Newman, J.; Bentley, J.; Hattarki, M.; Nuttall, S. D.; Foitzik, R. C.; Street, I. P.; Stupple, P. A.; Monahan, B. J.; Peat, T. S. Fragment screening for a protein-protein interaction inhibitor to WDR5. Struct. Dyn. 2019, 6, 064701, DOI: 10.1063/1.5122849Google Scholar53Fragment screening for a protein-protein interaction inhibitor to WDR5Dennis, Matthew L.; Morrow, Benjamin J.; Dolezal, Olan; Cuzzupe, Anthony N.; Stupple, Alexandra E.; Newman, Janet; Bentley, John; Hattarki, Meghan; Nuttall, Stewart D.; Foitzik, Richard C.; Street, Ian P.; Stupple, Paul A.; Monahan, Brendon J.; Peat, Thomas. S.Structural Dynamics (2019), 6 (6), 064701/1-064701/20CODEN: SDTYAE; ISSN:2329-7778. (American Institute of Physics)In this study, a fragment screen with Surface Plasmon Resonance (SPR) was used to identify a highly ligand-efficient imidazole-contg. compd. that is bound in the WIN site. The subsequent medicinal chem. campaign-guided by a suite of high-resoln. cocrystal structures with WDR5-progressed the initial hit to a low micromolar binder. One outcome from this study is a moiety that substitutes well for the side chain of arginine; a tripeptide contg. one such substitution was resolved in a high resoln. structure (1.5 Å) with a binding mode analogous to the native tripeptide. SPR furthermore indicates a similar residence time (kd = ∼0.06 s-1) for these two analogs. This novel scaffold therefore represents a possible means to overcome the potential permeability issues of WDR5 ligands that possess highly basic groups like guanidine. The series reported here furthers the understanding of the WDR5 WIN site and functions as a starting point for the development of more potent WDR5 inhibitors that may serve as cancer therapeutics. (c) 2019 American Institute of Physics.
- 54Bolshan, Y.; Getlik, M.; Kuznetsova, E.; Wasney, G. A.; Hajian, T.; Poda, G.; Nguyen, K. T.; Wu, H.; Dombrovski, L.; Dong, A.; Senisterra, G.; Schapira, M.; Arrowsmith, C. H.; Brown, P. J.; Al-Awar, R.; Vedadi, M.; Smil, D. Synthesis, Optimization, and Evaluation of Novel Small Molecules as Antagonists of WDR5-MLL Interaction. ACS Med. Chem. Lett. 2013, 4, 353– 357, DOI: 10.1021/ml300467nGoogle Scholar54Synthesis, Optimization, and Evaluation of Novel Small Molecules as Antagonists of WDR5-MLL InteractionBolshan, Yuri; Getlik, Matthaus; Kuznetsova, Ekaterina; Wasney, Gregory A.; Hajian, Taraneh; Poda, Gennadiy; Nguyen, Kong T.; Wu, Hong; Dombrovski, Ludmila; Dong, Aiping; Senisterra, Guillermo; Schapira, Matthieu; Arrowsmith, Cheryl H.; Brown, Peter J.; Al-awar, Rima; Vedadi, Masoud; Smil, DavidACS Medicinal Chemistry Letters (2013), 4 (3), 353-357CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)The WD40-repeat protein WDR5 plays a crit. role in maintaining the integrity of MLL complexes and fully activating their methyltransferase function. MLL complexes, the trithorax-like family of SET1 methyltransferases, catalyze trimethylation of lysine 4 on histone 3, and they have been widely implicated in various cancers. Antagonism of WDR5 and MLL subunit interaction by small mols. has recently been presented as a practical way to inhibit activity of the MLL1 complex, and N-(2-(4-methylpiperazin-1-yl)-5-substituted-phenyl) benzamides were reported as potent and selective antagonists of such an interaction. Here, we describe the protein crystal structure guided optimization of prototypic compd. I (Kdis = 7 μM), leading to identification of more potent antagonist II (Kdis = 0.3 μM).
- 55Getlik, M.; Smil, D.; Zepeda-Velázquez, C.; Bolshan, Y.; Poda, G.; Wu, H.; Dong, A.; Kuznetsova, E.; Marcellus, R.; Senisterra, G.; Dombrovski, L.; Hajian, T.; Kiyota, T.; Schapira, M.; Arrowsmith, C. H.; Brown, P. J.; Vedadi, M.; Al-Awar, R. Structure-Based Optimization of a Small Molecule Antagonist of the Interaction Between WD Repeat-Containing Protein 5 (WDR5) and Mixed-Lineage Leukemia 1 (MLL1). J. Med. Chem. 2016, 59, 2478– 2496, DOI: 10.1021/acs.jmedchem.5b01630Google Scholar55Structure-Based Optimization of a Small Molecule Antagonist of the Interaction Between WD Repeat-Containing Protein 5 (WDR5) and Mixed-Lineage Leukemia 1 (MLL1)Getlik, Matthaus; Smil, David; Zepeda-Velazquez, Carlos; Bolshan, Yuri; Poda, Gennady; Wu, Hong; Dong, Aiping; Kuznetsova, Ekaterina; Marcellus, Richard; Senisterra, Guillermo; Dombrovski, Ludmila; Hajian, Taraneh; Kiyota, Taira; Schapira, Matthieu; Arrowsmith, Cheryl H.; Brown, Peter J.; Vedadi, Masoud; Al-awar, RimaJournal of Medicinal Chemistry (2016), 59 (6), 2478-2496CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)WD repeat-contg. protein 5 (WDR5) is an important component of the multiprotein complex essential for activating mixed-lineage leukemia 1 (MLL1). Rearrangement of the MLL1 gene is assocd. with onset and progression of acute myeloid and lymphoblastic leukemias, and targeting the WDR5-MLL1 interaction may result in new cancer therapeutics. Our previous work showed that binding of small mol. ligands to WDR5 can modulate its interaction with MLL1, suppressing MLL1 methyltransferase activity. Initial structure-activity relationship studies identified N-(2-(4-methylpiperazin-1-yl)-5-substituted-phenyl) benzamides as potent and selective antagonists of this protein-protein interaction. Guided by crystal structure data and supported by in silico library design, we optimized the scaffold by varying the C-1 benzamide and C-5 substituents. This allowed us to develop the first highly potent (Kdisp < 100 nM) small mol. antagonists of the WDR5-MLL1 interaction and demonstrate that N-(4-(4-methylpiperazin-1-yl)-3'-(morpholinomethyl)-[1,1'-biphenyl]-3-yl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyridine-3-carboxamide 16d (OICR-9429) is a potent and selective chem. probe suitable to help dissect the biol. role of WDR5.
- 56Schapira, M.; Arrowsmith, C. H. Methyltransferase inhibitors for modulation of the epigenome and beyond. Curr. Opin. Chem. Biol. 2016, 33, 81– 87, DOI: 10.1016/j.cbpa.2016.05.030Google Scholar56Methyltransferase inhibitors for modulation of the epigenome and beyondSchapira, Matthieu; Arrowsmith, Cheryl H.Current Opinion in Chemical Biology (2016), 33 (), 81-87CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)Over the past two years tremendous progress has been made in the discovery of new inhibitors of protein lysine and arginine methyltransferases, establishing this class of epigenetic enzymes, along with DNA methyltransferases, as druggable protein families. New inhibitors of protein methyltransferases have been described with a variety of mechanisms of action including cofactor competitive, substrate competitive, allosteric inhibitors and disruptors of protein-protein interactions. Inhibitors have been used extensively in oncol. studies, and inhibitors of EZH2, and DOT1L are currently in clin. trials. Finally, advances in understanding the clin. mechanism of action of 5-azacytidine and related DNA hypomethylation agents were reported, revealing a likely role for a cell autonomous innate immune response.
- 57Gupta, A.; Xu, J.; Lee, S.; Tsai, S. T.; Zhou, B.; Kurosawa, K.; Werner, M. S.; Koide, A.; Ruthenburg, A. J.; Dou, Y.; Koide, S. Facile target validation in an animal model with intracellularly expressed monobodies. Nat. Chem. Biol. 2018, 14, 895– 900, DOI: 10.1038/s41589-018-0099-zGoogle Scholar57Facile target validation in an animal model with intracellularly expressed monobodiesGupta, Ankit; Xu, Jing; Lee, Shirley; Tsai, Steven T.; Zhou, Bo; Kurosawa, Kohei; Werner, Michael S.; Koide, Akiko; Ruthenburg, Alexander J.; Dou, Yali; Koide, ShoheiNature Chemical Biology (2018), 14 (9), 895-900CODEN: NCBABT; ISSN:1552-4450. (Nature Research)Rapidly detg. the biol. effect of perturbing a site within a potential drug target could guide drug discovery efforts, but it remains challenging. Here, the authors describe a facile target validation approach that exploits monobodies, small synthetic binding proteins that can be fully functionally expressed in cells. The authors developed a potent and selective monobody to WDR5, a core component of the mixed lineage leukemia (MLL) methyltransferase complex. The monobody bound to the MLL interaction site of WDR5, the same binding site for small-mol. inhibitors whose efficacy has been demonstrated in cells but not in animals. As a genetically encoded reagent, the monobody inhibited proliferation of an MLL-AF9 cell line in vitro, suppressed its leukemogenesis and conferred a survival benefit in an in vivo mouse leukemia model. The capacity of this approach to readily bridge biochem., structural, cellular characterization and tests in animal models may accelerate discovery and validation of druggable sites.
- 58Concepcion, J.; Witte, K.; Wartchow, C.; Choo, S.; Yao, D.; Persson, H.; Wei, J.; Li, P.; Heidecker, B.; Ma, W.; Varma, R.; Zhao, L.-S.; Perillat, D.; Carricato, G.; Recknor, M.; Du, K.; Ho, H.; Ellis, T.; Gamez, J.; Howes, M.; Phi-Wilson, J.; Lockard, S.; Zuk, R.; Tan, H. Label-free detection of biomolecular interactions using BioLayer interferometry for kinetic characterization. Comb. Chem. High Throughput Screen. 2009, 12, 791– 800, DOI: 10.2174/138620709789104915Google Scholar58Label-free detection of biomolecular interactions using biolayer interferometry for kinetic characterizationConcepcion, Joy; Witte, Krista; Wartchow, Charles; Choo, Sae; Yao, Danfeng; Persson, Henrik; Wei, Jing; Li, Pu; Heidecker, Bettina; Ma, Weilei; Varma, Ram; Zhao, Lian-She; Perillat, Donald; Carricato, Greg; Recknor, Michael; Du, Kevin; Ho, Huddee; Ellis, Tim; Gamez, Juan; Howes, Michael; Phi-Wilson, Janette; Lockard, Scott; Zuk, Robert; Tan, HongCombinatorial Chemistry & High Throughput Screening (2009), 12 (8), 791-800CODEN: CCHSFU; ISSN:1386-2073. (Bentham Science Publishers Ltd.)A review. The anal. of biomol. interactions is key in the drug development process. Label-free biosensor methods provide information on binding, kinetics, concn., and the affinity of an interaction. These techniques provide real-time monitoring of interactions between an immobilized ligand (such as a receptor) to an analyte in soln. without the use of labels. Advances in biosensor design and detection using BioLayer Interferometry (BLI) provide a simple platform that enables label-free monitoring of biomol. interactions without the use of flow cells. We review the applications of BLI in a wide variety of research and development environments for quantifying antibodies and proteins and measuring kinetics parameters.
- 59Weeramange, C. J.; Fairlamb, M. S.; Singh, D.; Fenton, A. W.; Swint-Kruse, L. The strengths and limitations of using biolayer interferometry to monitor equilibrium titrations of biomolecules. Protein Sci 2020, 29, 1018– 1034, DOI: 10.1002/pro.3827Google Scholar59The strengths and limitations of using biolayer interferometry to monitor equilibrium titrations of biomoleculesWeeramange, Chamitha J.; Fairlamb, Max S.; Singh, Dipika; Fenton, Aron W.; Swint-Kruse, LiskinProtein Science (2020), 29 (4), 1018-1034CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)Every method used to quantify biomol. interactions has its own strengths and limitations. To quantify protein-DNA binding affinities, nitrocellulose filter binding assays with 32P-labeled DNA quantify Kd values from 10-12 to 10-8 M but have several tech. limitations. Here, we considered the suitability of biolayer interferometry (BLI), which monitors assocn. and dissocn. of a sol. macromol. to an immobilized species; the ratio koff/kon dets. Kd. However, for lactose repressor protein (LacI) and an engineered repressor protein ("LLhF") binding immobilized DNA, complicated kinetic curves precluded this anal. Thus, we detd. whether the amplitude of the BLI signal at equil. related linearly to the fraction of protein bound to DNA. A key question was the effective concn. of immobilized DNA. Equil. titrn. expts. with DNA concns. below Kd (equil. binding regime) must be analyzed differently than those with DNA near or above Kd (stoichiometric binding regime). For ForteBio streptavidin tips, the most frequent effective DNA concn. was ∼2 x 10-9 M. Although variation occurred among different lots of sensor tips, binding events with Kd ≥ 10-8 M should reliably be in the equil. binding regime. We also obsd. effects from multi-valent interactions: Tetrameric LacI bound two immobilized DNAs whereas dimeric LLhF did not. We next used BLI to quantify the amt. of inducer sugars required to allosterically diminish protein-DNA binding and to assess the affinity of fructose-1-kinase for the DNA-LLhF complex. Overall, when exptl. design corresponded with appropriate data interpretation, BLI was convenient and reliable for monitoring equil. titrns. and thereby quantifying a variety of binding interactions.
- 60Imran, A.; Moyer, B. S.; Wolfe, A. J.; Cosgrove, M. S.; Makarov, D. E.; Movileanu, L. Interplay of Affinity and Surface Tethering in Protein Recognition. J. Phys. Chem. Lett. 2022, 13, 4021– 4028, DOI: 10.1021/acs.jpclett.2c00621Google Scholar60Interplay of Affinity and Surface Tethering in Protein RecognitionImran, Ali; Moyer, Brandon S.; Wolfe, Aaron J.; Cosgrove, Michael S.; Makarov, Dmitrii E.; Movileanu, LiviuJournal of Physical Chemistry Letters (2022), 13 (18), 4021-4028CODEN: JPCLCD; ISSN:1948-7185. (American Chemical Society)Surface-tethered ligand-receptor complexes are key components in biol. signaling and adhesion. They also find increasing utility in single-mol. assays and biotechnol. applications. Here, we study the real-time binding kinetics between various surface-immobilized peptide ligands and their unrestrained receptors. A long peptide tether increases the assocn. of ligand-receptor complexes, exptl. proving the fly casting mechanism where the disorder accelerates protein recognition. On the other hand, a short peptide tether enhances the complex dissocn. Notably, the rate consts. measured for the same receptor, but under different spatial constraints, are strongly correlated to one another. Furthermore, this correlation can be used to predict how surface tethering on a ligand-receptor complex alters its binding kinetics. Our results have immediate implications in the broad areas of biomol. recognition, intrinsically disordered proteins, and biosensor technol.
- 61Wolfe, A. J.; Si, W.; Zhang, Z.; Blanden, A. R.; Hsueh, Y.-C.; Gugel, J. F.; Pham, B.; Chen, M.; Loh, S. N.; Rozovsky, S.; Aksimentiev, A.; Movileanu, L. Quantification of membrane protein-detergent complex interactions. J. Phys. Chem. B 2017, 121, 10228– 10241, DOI: 10.1021/acs.jpcb.7b08045Google Scholar61Quantification of Membrane Protein-Detergent Complex InteractionsWolfe, Aaron J.; Si, Wei; Zhang, Zhengqi; Blanden, Adam R.; Hsueh, Yi-Ching; Gugel, Jack F.; Pham, Bach; Chen, Min; Loh, Stewart N.; Rozovsky, Sharon; Aksimentiev, Aleksei; Movileanu, LiviuJournal of Physical Chemistry B (2017), 121 (44), 10228-10241CODEN: JPCBFK; ISSN:1520-5207. (American Chemical Society)Although fundamentally significant in structural, chem., and membrane biol., the interfacial protein-detergent complex (PDC) interactions have been modestly examd. because of the complicated behavior of both detergents and membrane proteins in aq. phase. Membrane proteins are prone to unproductive aggregation resulting from poor detergent solvation, but the participating forces in this phenomenon remain ambiguous. Here, using rational membrane protein design, targeted chem. modification, and steady-state fluorescence polarization spectroscopy, the detergent desolvation of membrane proteins can be quant. evaluated. Depleting the detergent in the sample well produced a two-state transition of membrane proteins between a fully detergent-solvated state and a detergent-desolvated state, the nature of which depended on the interfacial PDC interactions. Using a panel of six membrane proteins of varying hydrophobic topog., structural fingerprint, and charge distribution on the solvent-accessible surface, the authors provide direct exptl. evidence for the contributions of the electrostatic and hydrophobic interactions to the protein solvation properties. Moreover, all-atom mol. dynamics simulations report the major contribution of the hydrophobic forces exerted at the PDC interface. This semiquant. approach might be extended in the future to include studies of the interfacial PDC interactions of other challenging membrane protein systems of unknown structure. This would have practical importance in protein extn., solubilization, stabilization, and crystn.
- 62Wolfe, A. J.; Gugel, J. F.; Chen, M.; Movileanu, L. Kinetics of Membrane Protein-Detergent Interactions Depend on Protein Electrostatics. J. Phys. Chem. B 2018, 122, 9471– 9481, DOI: 10.1021/acs.jpcb.8b07889Google Scholar62Kinetics of Membrane Protein-Detergent Interactions Depend on Protein ElectrostaticsWolfe, Aaron J.; Gugel, Jack F.; Chen, Min; Movileanu, LiviuJournal of Physical Chemistry B (2018), 122 (41), 9471-9481CODEN: JPCBFK; ISSN:1520-5207. (American Chemical Society)Interactions of a membrane protein with a detergent micelle represent a fundamental process with practical implications in structural and chem. biol. Quant. assessment of the kinetics of protein-detergent complex (PDC) interactions has always been challenged by complicated behavior of both membrane proteins and solubilizing detergents in aq. phase. Here, we show the kinetic reads of the desorption of maltoside-contg. detergents from β-barrel membrane proteins. Using steady-state fluorescence polarization (FP) anisotropy measurements, we recorded real-time, specific signatures of the PDC interactions. The results of these measurements were used to infer the model-dependent rate consts. of assocn. and dissocn. of the proteomicelles. Remarkably, the kinetics of the PDC interactions depend on the overall protein charge despite the nonionic nature of the detergent monomers. In the future, this approach might be employed for high-throughput screening of kinetic fingerprints of different membrane proteins stabilized in micelles that contain mixts. of various detergents.
- 63Forbes, S. A.; Beare, D.; Boutselakis, H.; Bamford, S.; Bindal, N.; Tate, J.; Cole, C. G.; Ward, S.; Dawson, E.; Ponting, L.; Stefancsik, R.; Harsha, B.; Kok, C. Y.; Jia, M.; Jubb, H.; Sondka, Z.; Thompson, S.; De, T.; Campbell, P. J. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res. 2017, 45, D777– d783, DOI: 10.1093/nar/gkw1121Google Scholar63COSMIC: somatic cancer genetics at high-resolutionForbes, Simon A.; Beare, David; Boutselakis, Harry; Bamford, Sally; Bindal, Nidhi; Tate, John; Cole, Charlotte G.; Ward, Sari; Dawson, Elisabeth; Ponting, Laura; Stefancsik, Raymund; Harsha, Bhavana; Kok, Chai Yin; Jia, Mingming; Jubb, Harry; Sondka, Zbyslaw; Thompson, Sam; De, Tisham; Campbell, Peter J.Nucleic Acids Research (2017), 45 (D1), D777-D783CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)COSMIC, the Catalog of Somatic Mutations in Cancer (http://cancer.sanger.ac.uk) is a high-resoln. resource for exploring targets and trends in the genetics of human cancer. Currently the broadest database of mutations in cancer, the information in COSMIC is curated by expert scientists, primarily by scrutinizing large nos. of scientific publications. Over 4 million coding mutations are described in v78 (Sept. 2016), combining genome-wide sequencing results from 28 366 tumors with complete manual curation of 23 489 individual publications focused on 186 key genes and 286 key fusion pairs across all cancers. Mol. profiling of large tumor nos. has also allowed the annotation of more than 13 million non-coding mutations, 18 029 gene fusions, 187 429 genome rearrangements, 1 271 436 abnormal copy no. segments, 9 175 462 abnormal expression variants and 7 879 142 differentially methylated CpG dinucleotides. COSMIC now details the genetics of drug resistance, novel somatic gene mutations which allow a tumor to evade therapeutic cancer drugs. Focusing initially on highly characterized drugs and genes, COSMIC v78 contains wide resistance mutation profiles across 20 drugs, detailing the recurrence of 301 unique resistance alleles across 1934 drug-resistant tumors. All information from the COSMIC database is available freely on the COSMIC website.
- 64Kumar, S.; Nussinov, R. Relationship between ion pair geometries and electrostatic strengths in proteins. Biophys. J. 2002, 83, 1595– 1612, DOI: 10.1016/s0006-3495(02)73929-5Google Scholar64Relationship between ion pair geometries and electrostatic strengths in proteinsKumar, Sandeep; Nussinov, RuthBiophysical Journal (2002), 83 (3), 1595-1612CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)The electrostatic free energy contribution of an ion pair in a protein depends on two factors, geometrical orientation of the side-chain charged groups with respect to each other and the structural context of the ion pair in the protein. Conformers in NMR ensembles enable studies of the relationship between geometry and electrostatic strengths of ion pairs, because the protein structural contexts are highly similar across different conformers. We have studied this relationship using a dataset of 22 unique ion pairs in 14 NMR conformer ensembles for 11 nonhomologous proteins. In different NMR conformers, the ion pairs are classified as salt bridges, nitrogen-oxygen (N-O) bridges and longer-range ion pairs on the basis of geometrical criteria. In salt bridges, centroids of the side-chain charged groups and at least a pair of side-chain nitrogen and oxygen atoms of the ion-pairing residues are within a 4 Å distance. In N-O bridges, at least a pair of the side-chain nitrogen and oxygen atoms of the ion-pairing residues are within 4 Å distance, but the distance between the side-chain charged group centroids is greater than 4 Å. In the longer-range ion pairs, the side-chain charged group centroids as well as the side-chain nitrogen and oxygen atoms are more than 4 Å apart. Continuum electrostatic calcns. indicate that most of the ion pairs have stabilizing electrostatic contributions when their side-chain charged group centroids are within 5 Å distance. Hence, most (∼92%) of the salt bridges and a majority (68%) of the N-O bridges are stabilizing. Most (∼89%) of the destabilizing ion pairs are the longer-range ion pairs. In the NMR conformer ensembles, the electrostatic interaction between side-chain charged groups of the ion-pairing residues is the strongest for salt bridges, considerably weaker for N-O bridges, and the weakest for longer-range ion pairs. These results suggest empirical rules for stabilizing electrostatic interactions in proteins.
- 65Nogal, B.; Bowman, C. A.; Ward, A. B. Time-course, negative-stain electron microscopy-based analysis for investigating protein-protein interactions at the single-molecule level. J. Biol. Chem. 2017, 292, 19400– 19410, DOI: 10.1074/jbc.m117.808352Google Scholar65Time-course, negative-stain electron microscopy-based analysis for investigating protein-protein interactions at the single-molecule levelNogal, Bartek; Bowman, Charles A.; Ward, Andrew B.Journal of Biological Chemistry (2017), 292 (47), 19400-19410CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Several biophys. approaches are available to study protein-protein interactions. Most approaches are conducted in bulk soln. and are therefore limited to an av. measurement of the ensemble of mol. interactions. Here, single-particle EM can enrich the understanding of protein-protein interactions at the single-mol. level and potentially capture states that are unobservable with ensemble methods because they are below the limit of detection or not conducted on an appropriate time scale. Using the HIV-1 envelope glycoprotein (Env) and its interaction with receptor CD4-binding site neutralizing antibodies as a model system, the authors both corroborate ensemble kinetics-derived parameters and demonstrate how time-course EM can further dissect stoichiometric states of complexes that are not readily observable with other methods. Visualization of the kinetics and stoichiometry of Env-antibody complexes demonstrated the applicability of this approach to qual. and semi-quant. differentiate two highly similar neutralizing antibodies. Furthermore, implementation of machine-learning techniques for sorting class avs. of these complexes into discrete subclasses of particles helped reduce human bias. The authors' data provide proof of concept that single-particle EM can be used to generate a "visual" kinetic profile that should be amenable to studying many other protein-protein interactions, is relatively simple and complementary to well-established biophys. approaches. Moreover, the authors' method provides crit. insights into broadly neutralizing antibody recognition of Env, which may inform vaccine immunogen design and immunotherapeutic development.
- 66Chen, C.; Liu, Y.; Rappaport, A. R.; Kitzing, T.; Schultz, N.; Zhao, Z.; Shroff, A. S.; Dickins, R. A.; Vakoc, C. R.; Bradner, J. E.; Stock, W.; LeBeau, M. M.; Shannon, K. M.; Kogan, S.; Zuber, J.; Lowe, S. W. MLL3 is a haploinsufficient 7q tumor suppressor in acute myeloid leukemia. Cancer Cell 2014, 25, 652– 665, DOI: 10.1016/j.ccr.2014.03.016Google Scholar66MLL3 Is a Haploinsufficient 7q Tumor Suppressor in Acute Myeloid LeukemiaChen, Chong; Liu, Yu; Rappaport, Amy R.; Kitzing, Thomas; Schultz, Nikolaus; Zhao, Zhen; Shroff, Aditya S.; Dickins, Ross A.; Vakoc, Christopher R.; Bradner, James E.; Stock, Wendy; LeBeau, Michelle M.; Shannon, Kevin M.; Kogan, Scott; Zuber, Johannes; Lowe, Scott W.Cancer Cell (2014), 25 (5), 652-665CODEN: CCAECI; ISSN:1535-6108. (Elsevier Inc.)Recurring deletions of chromosome 7 and 7q [-7/del(7q)] occur in myelodysplastic syndromes and acute myeloid leukemia (AML) and are assocd. with poor prognosis. However, the identity of functionally relevant tumor suppressors on 7q remains unclear. Using RNAi and CRISPR/Cas9 approaches, we show that an ∼50% redn. in gene dosage of the mixed lineage leukemia 3 (MLL3) gene, located on 7q36.1, cooperates with other events occurring in -7/del(7q) AMLs to promote leukemogenesis. Mll3 suppression impairs the differentiation of HSPC. Interestingly, Mll3-suppressed leukemias, like human -7/del(7q) AMLs, are refractory to conventional chemotherapy but sensitive to the BET inhibitor JQ1. Thus, our mouse model functionally validates MLL3 as a haploinsufficient 7q tumor suppressor and suggests a therapeutic option for this aggressive disease.
- 67Wu, H.-T.; Liu, Y.-E.; Hsu, K.-W.; Wang, Y.-F.; Chan, Y.-C.; Chen, Y.; Chen, D.-R. MLL3 Induced by Luteolin Causes Apoptosis in Tamoxifen-Resistant Breast Cancer Cells through H3K4 Monomethylation and Suppression of the PI3K/AKT/mTOR Pathway. Am. J. Chin. Med. 2020, 48, 1221– 1241, DOI: 10.1142/s0192415x20500603Google Scholar67MLL3 Induced by Luteolin Causes Apoptosis in Tamoxifen-Resistant Breast Cancer Cells through H3K4 Monomethylation and Suppression of the PI3K/AKT/mTOR PathwayWu, Han-Tsang; Liu, Yi-En; Hsu, Kai-Wen; Wang, Yu-Fen; Chan, Ya-Chi; Chen, Yeh; Chen, Dar-RenAmerican Journal of Chinese Medicine (2020), 48 (5), 1221-1241CODEN: AJCMBA; ISSN:0192-415X. (World Scientific Publishing Co. Pte. Ltd.)Tamoxifen is one of the most common hormone therapy drug for estrogen receptor pos. breast cancer. Tumor cells with drug resistance often cause recurrence and metastasis in cancer patients. Luteolin is a natural compd. found from various types of vegetables and exhibit anticancer activity in different cancers. This study demonstrated that luteolin inhibits the proliferation and induces apoptosis of tamoxifen-resistant ER-pos. breast cancer cells. Luteolin also causes cell cycle arrest at the G2/M phase and decreases mitochondrial membrane potential. Besides, luteolin reduces the levels of activated PI3K/AKT/mTOR signaling pathway. The combination treatment of luteolin and PI3K, AKT, or mTOR inhibitors synergistically increases apoptosis in tamoxifen-resistant ER-pos. breast cancer cells. Ras gene family (K-Ras, H-Ras, and N-Ras), an activator of PI3K, was transcriptionally repressed by luteolin via induction of tumor suppressor mixed-lineage leukemia 3 expression. MLL3 increases the level of monomethylation of Histone 3 Lysine 4 on the enhancer and promoter region of Ras genes, thus causes repression of Ras expressions. Our finding implies that luteolin was a promising natural agent against tamoxifen resistance of breast cancer.
- 68Rampias, T.; Karagiannis, D.; Avgeris, M.; Polyzos, A.; Kokkalis, A.; Kanaki, Z.; Kousidou, E.; Tzetis, M.; Kanavakis, E.; Stravodimos, K.; Manola, K. N.; Pantelias, G. E.; Scorilas, A.; Klinakis, A. The lysine-specific methyltransferase KMT2C/MLL3 regulates DNA repair components in cancer. EMBO Rep. 2019, 20, e46821 DOI: 10.15252/embr.201846821Google ScholarThere is no corresponding record for this reference.
- 69Wong, S. H. K.; Goode, D. L.; Iwasaki, M.; Wei, M. C.; Kuo, H.-P.; Zhu, L.; Schneidawind, D.; Duque-Afonso, J.; Weng, Z.; Cleary, M. L. The H3K4-Methyl Epigenome Regulates Leukemia Stem Cell Oncogenic Potential. Cancer Cell 2015, 28, 198– 209, DOI: 10.1016/j.ccell.2015.06.003Google Scholar69The H3K4-Methyl Epigenome Regulates Leukemia Stem Cell Oncogenic PotentialWong, Stephen H. K.; Goode, David L.; Iwasaki, Masayuki; Wei, Michael C.; Kuo, Hsu-Ping; Zhu, Li; Schneidawind, Dominik; Duque-Afonso, Jesus; Weng, Ziming; Cleary, Michael L.Cancer Cell (2015), 28 (2), 198-209CODEN: CCAECI; ISSN:1535-6108. (Elsevier Inc.)The genetic programs that maintain leukemia stem cell (LSC) self-renewal and oncogenic potential have been well defined; however, the comprehensive epigenetic landscape that sustains LSC cellular identity and functionality is less well established. We report that LSCs in MLL-assocd. leukemia reside in an epigenetic state of relative genome-wide high-level H3K4me3 and low-level H3K79me2. LSC differentiation is assocd. with reversal of these broad epigenetic profiles, with concomitant downregulation of crucial MLL target genes and the LSC maintenance transcriptional program that is driven by the loss of H3K4me3, but not H3K79me2. The H3K4-specific demethylase KDM5B neg. regulates leukemogenesis in murine and human MLL-rearranged AML cells, demonstrating a crucial role for the H3K4 global methylome in detg. LSC fate.
- 70Shinsky, S. A.; Cosgrove, M. S. Unique Role of the WD-40 Repeat Protein 5 (WDR5) Subunit within the Mixed Lineage Leukemia 3 (MLL3) Histone Methyltransferase Complex. J. Biol. Chem. 2015, 290, 25819– 25833, DOI: 10.1074/jbc.m115.684142Google Scholar70Unique role of the WD-40 repeat protein 5 (WDR5) subunit within the mixed lineage leukemia 3 (MLL3) histone methyltransferase complexShinsky, Stephen A.; Cosgrove, Michael S.Journal of Biological Chemistry (2015), 290 (43), 25819-25833CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The MLL3 (mixed lineage leukemia 3) protein is a member of the human SET1 family of histone H3 lysine 4 methyltransferases and contains the conserved WDR5 interaction (Win) motif and the catalytic suppressor of variegation, enhancer of zeste, trithorax (SET) domain. The human SET1 family includes MLL1-4 and SETd1A/B, which all interact with a conserved subcomplex contg. WDR5, RbBP5, Ash2L, and DPY-30 (WRAD) to form the minimal core complex required for full methyltransferase activity. However, recent evidence suggests that the WDR5 subunit may not be utilized in an identical manner within all SET1 family core complexes. Although the roles of WDR5 within the MLL1 core complex have been extensively studied, not much is known about the roles of WDR5 in other SET1 family core complexes. In this investigation, we set out to characterize the roles of the WDR5 subunit in the MLL3 core complex. We found that unlike MLL1, the MLL3 SET domain assembles with the RbBP5/Ash2L heterodimer independently of the Win motif-WDR5 interaction. Furthermore, we obsd. that WDR5 inhibits the monomethylation activity of the MLL3 core complex, which is dependent on the Win motif. We also found evidence suggesting that the WRAD subcomplex catalyzes weak H3K4 monomethylation within the context of the MLL3 core complex. Furthermore, soln. structures of the MLL3 core complex assembled with and without WDR5 by small angle x-ray scattering show similar overall topologies. Together, this work demonstrates a unique role for WDR5 in modulating the enzymic activity of the MLL3 core complex.
- 71Wysocka, J.; Swigut, T.; Milne, T. A.; Dou, Y.; Zhang, X.; Burlingame, A. L.; Roeder, R. G.; Brivanlou, A. H.; Allis, C. D. WDR5 associates with histone H3 methylated at K4 and is essential for H3 K4 methylation and vertebrate development. Cell 2005, 121, 859– 872, DOI: 10.1016/j.cell.2005.03.036Google Scholar71WDR5 associates with histone H3 methylated at K4 and is essential for H3 K4 methylation and vertebrate developmentWysocka, Joanna; Swigut, Tomek; Milne, Thomas A.; Dou, Yali; Zhang, Xin; Burlingame, Alma L.; Roeder, Robert G.; Brivanlou, Ali H.; Allis, C. DavidCell (Cambridge, MA, United States) (2005), 121 (6), 859-872CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Histone H3 lysine 4 (K4) methylation has been linked to the transcriptional activation in a variety of eukaryotic species. Here authors show that a common component of MLL1, MLL2, and hSet1 H3 K4 methyltransferase complexes, the WD40-repeat protein WDR5, directly assocs. with histone H3 di- and trimethylated at K4 and with H3-K4-dimethylated nucleosomes. WDR5 is required for binding of the methyltransferase complex to the K4-dimethylated H3 tail as well as for global H3 K4 trimethylation and HOX gene activation in human cells. WDR5 is essential for vertebrate development, in that WDR5-depleted X. laevis tadpoles exhibit a variety of developmental defects and abnormal spatial Hox gene expression. These results are the first demonstration that a WD40-repeat protein acts as a module for recognition of a specific histone modification and suggest a mechanism for reading and writing an epigenetic mark for gene activation.
- 72Itan, Y.; Casanova, J.-L. Can the impact of human genetic variations be predicted?. Proc. Natl. Acad. Sci. U.S.A. 2015, 112, 11426– 11427, DOI: 10.1073/pnas.1515057112Google Scholar72Can the impact of human genetic variations be predicted?Itan, Yuval; Casanova, Jean-LaurentProceedings of the National Academy of Sciences of the United States of America (2015), 112 (37), 11426-11427CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)There is no expanded citation for this reference.
- 73Ali, A.; Veeranki, S. N.; Tyagi, S. A SET-domain-independent role of WRAD complex in cell-cycle regulatory function of mixed lineage leukemia. Nucleic Acids Res. 2014, 42, 7611– 7624, DOI: 10.1093/nar/gku458Google Scholar73A SET-domain-independent role of WRAD complex in cell-cycle regulatory function of mixed lineage leukemiaAli, Aamir; Veeranki, Sailaja Naga; Tyagi, ShwetaNucleic Acids Research (2014), 42 (12), 7611-7624CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)MLL, the trithorax ortholog, is a well-characterized histone 3 lysine 4 methyltransferase that is crucial for proper regulation of the Hox genes during embryonic development. Chromosomal translocations, disrupting the Mll gene, lead to aggressive leukemia with poor prognosis. However, the functions of MLL in cellular processes like cell-cycle regulation are not well studied. Here we show that the MLL has a regulatory role during multiple phases of the cell cycle. RNAi-mediated knockdown reveals that MLL regulates S-phase progression and, proper segregation and cytokinesis during M phase. Using deletions and mutations, we narrow the cell-cycle regulatory role to the C subunit of MLL. Our anal. reveals that the transactivation domain and not the SET domain is important for the S-phase function of MLL. Surprisingly, disruption of MLL-WRAD interaction is sufficient to disrupt proper mitotic progression. These mitotic functions of WRAD are independent of SET domain of MLL and, therefore, define a new role of WRAD in subset of MLL functions. Finally, we address the overlapping and unique roles of the different SET family members in the cell cycle.
- 74Movileanu, L.; Cheley, S.; Howorka, S.; Braha, O.; Bayley, H. Location of a Constriction in the Lumen of a Transmembrane Pore by Targeted Covalent Attachment of Polymer Molecules. J. Gen. Physiol. 2001, 117, 239– 252, DOI: 10.1085/jgp.117.3.239Google Scholar74Location of a constriction in the lumen of a transmembrane pore by targeted covalent attachment of polymer moleculesMovileanu, Liviu; Cheley, Stephen; Howorka, Stefan; Braha, Orit; Bayley, HaganJournal of General Physiology (2001), 117 (3), 239-251CODEN: JGPLAD; ISSN:0022-1295. (Rockefeller University Press)Few methods exist for obtaining the internal dimensions of transmembrane pores for which 3-D structures are lacking or for showing that structures detd. by crystallog. reflect the internal dimensions of pores in lipid bilayers. Several approaches, involving polymer penetration and transport, have revealed limiting diams. for various pores. But, in general, these approaches do not indicate the locations of constrictions in the channel lumen. Here, the authors combine cysteine mutagenesis and chem. modification with sulfhydryl-reactive polymers to locate the constriction in the lumen of the staphylococcal α-hemolysin pore, a model protein of known structure. The rates of reaction of each of four polymeric regents (MePEG-OPSS) of different masses towards individual single cysteine mutants, comprising a set with cysteines distributed over the length of the lumen of the pore, were detd. by macroscopic current recording. The rates for the three larger polymers (1.8, 2.5, and 5.0 kDa) were normalized with respect to the rates of reaction with a 1.0 kDa polymer for each of the seven positions in the lumen. The rate of reaction of the 5.0 kDa polymer dropped dramatically at the centrally located Cys-111 residue and positions distal to Cys-111, whether the reagent was applied from the trans or the cis side of the bilayer. This semi-quant. anal. sufficed to demonstrate that a constriction is located at the midpoint of the pore lumen, as predicted by the crystal structure, and although the constriction allows a 2.5 kDa polymer to pass, transport of a 5.0 kDa mol. is greatly restricted. In addn., PEG chains gave greater redns. in pore conductance when covalently attached to the narrower regions of the lumen, permitting further definition of the interior of the pore. The procedures described here should be applicable to other pores and to related structures such as the vestibules of ion channels.
- 75Jarmoskaite, I.; AlSadhan, I.; Vaidyanathan, P. P.; Herschlag, D. How to measure and evaluate binding affinities. eLife 2020, 9, e57264 DOI: 10.7554/eLife.57264Google Scholar75How to measure and evaluate binding affinitiesJarmoskaite, Inga; Alsadhan, Ishraq; Vaidyanathan, Pavanapuresan P.; Herschlag, DanieleLife (2020), 9 (), e57264CODEN: ELIFA8; ISSN:2050-084X. (eLife Sciences Publications Ltd.)Quant. measurements of biomol. assocns. are central to biol. understanding and are needed to build and test predictive and mechanistic models. Given the advances in high-throughput technologies and the projected increase in the availability of binding data, we found it esp. timely to evaluate the current stds. for performing and reporting binding measurements. A review of 100 studies revealed that in most cases essential controls for establishing the appropriate incubation time and concn. regime were not documented, making it impossible to det. measurement reliability. Moreover, several reported affinities could be concluded to be incorrect, thereby impacting biol. interpretations. Given these challenges, we provide a framework for a broad range of researchers to evaluate, teach about, perform, and clearly document high-quality equil. binding measurements. We apply this framework and explain underlying fundamental concepts through exptl. examples with the RNA-binding protein Puf4.
- 76Wolfe, A. J.; Hsueh, Y.-C.; Blanden, A. R.; Mohammad, M. M.; Pham, B.; Thakur, A. K.; Loh, S. N.; Chen, M.; Movileanu, L. Interrogating Detergent Desolvation of Nanopore-Forming Proteins by Fluorescence Polarization Spectroscopy. Anal. Chem. 2017, 89, 8013– 8020, DOI: 10.1021/acs.analchem.7b01339Google Scholar76Interrogating Detergent Desolvation of Nanopore-Forming Proteins by Fluorescence Polarization SpectroscopyWolfe, Aaron J.; Hsueh, Yi-Ching; Blanden, Adam R.; Mohammad, Mohammad M.; Pham, Bach; Thakur, Avinash K.; Loh, Stewart N.; Chen, Min; Movileanu, LiviuAnalytical Chemistry (Washington, DC, United States) (2017), 89 (15), 8013-8020CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Understanding how membrane proteins interact with detergents is of fundamental and practical significance in structural and chem. biol. as well as in nanobiotechnol. Current methods for inspecting protein-detergent complex (PDC) interfaces require high concns. of protein and are of low throughput. Here, the authors describe a scalable, spectroscopic approach that uses nanomolar protein concns. in native solns. This approach, which is based on steady-state fluorescence polarization (FP) spectroscopy, kinetically resolves the dissocn. of detergents from membrane proteins and protein unfolding. For satisfactorily solubilizing detergents, at concns. much greater than the crit. micelle concn. (CMC), the fluorescence anisotropy was independent of detergent concn. In contrast, at detergent concns. comparable with or below the CMC, the anisotropy readout underwent a time-dependent decrease, showing a specific and sensitive protein unfolding signature. Functionally reconstituted membrane proteins into a bilayer membrane confirmed predictions made by these FP-based detns. with respect to varying refolding conditions. From a practical point of view, this 96-well anal. approach will facilitate a massively parallel assessment of the PDC interfacial interactions under a fairly broad range of micellar and environmental conditions. The authors expect that these studies will potentially accelerate research in membrane proteins pertaining to their extn., solubilization, stabilization, and crystn., as well as reconstitution into bilayer membranes.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 5 publications.
- Lauren A. Mayse, Yazheng Wang, Mohammad Ahmad, Liviu Movileanu. Real-Time Measurement of a Weak Interaction of a Transcription Factor Motif with a Protein Hub at Single-Molecule Precision. ACS Nano 2024, 18
(31)
, 20468-20481. https://doi.org/10.1021/acsnano.4c04857
- Lauren A. Mayse, Ali Imran, Yazheng Wang, Mohammad Ahmad, Rebecca A. Oot, Stephan Wilkens, Liviu Movileanu. Evaluation of Nanopore Sensor Design Using Electrical and Optical Analyses. ACS Nano 2023, 17
(11)
, 10857-10871. https://doi.org/10.1021/acsnano.3c02532
- Mohammad Ahmad, Ali Imran, Liviu Movileanu. Overlapping characteristics of weak interactions of two transcriptional regulators with WDR5. International Journal of Biological Macromolecules 2024, 258 , 128969. https://doi.org/10.1016/j.ijbiomac.2023.128969
- I. V. Astratenkova, N. D. Golberg, V. A. Rogozkin. Regulation of Gene Expression by the MYC Transcription Factor Network During Exercise. Human Physiology 2023, 49
(4)
, 444-452. https://doi.org/10.1134/S036211972360011X
- I. V. Astratenkova, N. D. Golberg, V. A. Rogozkin. Regulation of Gene Expression by the MYC Transcription Factor Network during Exercise. Физиология человека 2023, 49
(4)
, 124-132. https://doi.org/10.31857/S0131164622601014
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Figure 1
Figure 1. Two binding sites of WDR5 and the structure of the WDR5-MLL3Win complex. (a) Representations of the Win and WBM binding sites of WDR5. Orientations of WDR5 in the two cartoons are 180° with respect to each other. (b) Top view of the WDR5-MLL3Win complex. (c) Side view of the WDR5-MLL3Win complex. (d) Side view of the interaction sites between MLL3Win (green) and WDR5 (gray). All presented residues are within 5 Å of the other binding partner. The residues corresponding to the WDR5 mutations explored in this study are marked in dark gray. MLL3Win residues are labeled as well. Potential hydrogen bonds between the two binding partners are shown as yellow dotted lines. (e) Key residues of the WDR5 binding cavity involved in hydrogen bonding with the evolutionarily conserved Arg residue (R4710) of MLL3Win at position P0 (Table S1). The hydrogen bonds are indicated by thick dashed lines marked in yellow. The cutoff distance for identifying these hydrogen bonds was 4.0 Å. WDR5 was represented using pdb entry 4ERY. (35)
Figure 2
Figure 2. Label-free optical BLI sensorgrams of the WDR5 mutant-MLL3Win interactions. (a) Locations of the surface and cavity WDR5 mutations are shown in blue using surface and cross-sectional views of WDR5, respectively. (b) BLI sensorgrams showing the association and dissociation phases. For each WDR5 mutation, sensors with immobilized MLL3Win ligand were immersed in buffers containing different WDR5 concentrations (listed on sensorgrams) to monitor association kinetics. Sensors were then transferred to buffer alone to monitor dissociation kinetics.
Figure 3
Figure 3. Normalized dissociation rate constants of the WDR5 mutant-SET1Win interactions using BLI sensorgrams. The koff values for each SET1Win ligand’s interaction with mutants have been divided by the koff of that SET1Win ligand’s interaction with the native WDR5 protein. (a) MLL2Win, (b) MLL3Win, (c) MLL4Win, (d) SETd1AWin, and (e) SETd1BWin. ND stands for “Not Determined”. Using a BLI measurement, the interaction between F133L and MLL4Win was detectable, but not quantifiable.
Figure 4
Figure 4. Normalized KD of the WDR5 mutant-SET1Win interactions using BLI sensorgrams. The KD values for each SET1Win ligand’s interaction with WDR5 mutants have been divided by the KD of that SET1Win ligand’s interaction with the native WDR5 protein. (a) MLL2Win, (b) MLL3Win, (c) MLL4Win, (d) SETd1AWin, and (e) SETd1BWin. ND stands for “Not Determined”. Using a BLI measurement, the interaction between F133L and MLL4Win was detectable, but not quantifiable.
Figure 5
Figure 5. Normalized KD of the WDR5 mutant-SET1Win interactions using steady-state FP spectroscopy. The KD values for each SET1Win ligand’s interaction with WDR5 mutants have been divided by the KD of that SET1Win ligand’s interaction with the native WDR5 protein. (a) MLL1Win, (b) MLL2Win, (c) MLL3Win, (d) MLL4Win, (e) SETd1AWin, and (f) SETd1BWin. For vertical bars marked by “*”, the KD of those interactions could not be determined. Those values represent the lower-limit of the KD based on the highest WDR5 mutant concentrations used in this study.
Figure 6
Figure 6. Quantitative comparison between affinity data resulting from BLI and FP measurements. (a) 3D graph of the ratio of the normalized KD-BLI to the normalized KD-FP. (b) Two-dimensional heat map of the ratio of the normalized KD-BLI to the normalized KD-FP. Normalized KD values are the KD measured for a specific WDR5 mutant-SET1Win interaction pair divided by the KD value corresponding to the native WDR5 protein.
References
This article references 76 other publications.
- 1Xu, C.; Min, J. Structure and function of WD40 domain proteins. Protein Cell 2011, 2, 202– 214, DOI: 10.1007/s13238-011-1018-11Structure and function of WD40 domain proteinsXu, Chao; Min, JinrongProtein & Cell (2011), 2 (3), 202-214CODEN: PCREFB; ISSN:1674-800X. (Higher Education Press)A review. The WD40 domain exhibits a β-propeller architecture, often comprising seven blades. The WD40 domain is one of the most abundant domains and also among the top interacting domains in eukaryotic genomes. In this review, we will discuss the identification, definition and architecture of the WD40 domains. WD40 domain proteins are involved in a large variety of cellular processes, in which WD40 domains function as a protein-protein or protein-DNA interaction platform. WD40 domain mediates mol. recognition events mainly through the smaller top surface, but also through the bottom surface and sides. So far, no WD40 domain has been found to display enzymic activity. We will also discuss the different binding modes exhibited by the large versatile family of WD40 domain proteins. In the last part of this review, we will discuss how post-translational modifications are recognized by WD40 domain proteins.
- 2Zhang, C.; Zhang, F. The Multifunctions of WD40 Proteins in Genome Integrity and Cell Cycle Progression. J. Genomics 2015, 3, 40– 50, DOI: 10.7150/jgen.110152The Multifunctions of WD40 Proteins in Genome Integrity and Cell Cycle ProgressionZhang Caiguo; Zhang FanJournal of genomics (2015), 3 (), 40-50 ISSN:1839-9940.Eukaryotic genome encodes numerous WD40 repeat proteins, which generally function as platforms of protein-protein interactions and are involved in numerous biological process, such as signal transduction, gene transcriptional regulation, protein modifications, cytoskeleton assembly, vesicular trafficking, DNA damage and repair, cell death and cell cycle progression. Among these diverse functions, genome integrity maintenance and cell cycle progression are extremely important as deregulation of them is clinically linked to uncontrolled proliferative diseases such as cancer. Thus, we mainly summarize and discuss the recent understanding of WD40 proteins and their molecular mechanisms linked to genome stability and cell cycle progression in this review, thereby demonstrating their pervasiveness and importance in cellular networks.
- 3Afanasieva, E.; Chaudhuri, I.; Martin, J.; Hertle, E.; Ursinus, A.; Alva, V.; Hartmann, M. D.; Lupas, A. N. Structural diversity of oligomeric β-propellers with different numbers of identical blades. eLife 2019, 8, e49853 DOI: 10.7554/eLife.498533Structural diversity of oligomeric β-propellers with different numbers of identical bladesAfanasieva, Evgenia; Chaudhuri, Indronil; Martin, Joerg; Hertle, Eva; Ursinus, Astrid; Alva, Vikram; Hartmann, Marcus D.; Lupas, Andrei N.eLife (2019), 8 (), e49853/1-e49853/15CODEN: ELIFA8; ISSN:2050-084X. (eLife Sciences Publications Ltd.)β-Propellers arise through the amplification of a supersecondary structure element called a blade. This process produces toroids of between four and twelve repeats, which are almost always arranged sequentially in a single polypeptide chain. We found that new propellers evolve continuously by amplification from single blades. We therefore investigated whether such nascent propellers can fold as homo-oligomers before they have been fully amplified within a single chain. One- to six-bladed building blocks derived from two seven-bladed WD40 propellers yielded stable homo-oligomers with six to nine blades, depending on the size of the building block. High resoln. structures for tetramers of two blades, trimers of three blades, and dimers of four and five blades, resp., show structurally diverse propellers and include a novel fold, highlighting the inherent flexibility of the WD40 blade. Our data support the hypothesis that subdomain-sized fragments can provide structural versatility in the evolution of new proteins.
- 4Stirnimann, C. U.; Petsalaki, E.; Russell, R. B.; Müller, C. W. WD40 proteins propel cellular networks. Trends Biochem. Sci. 2010, 35, 565– 574, DOI: 10.1016/j.tibs.2010.04.0034WD40 proteins propel cellular networksStirnimann, Christian U.; Petsalaki, Evangelia; Russell, Robert B.; Mueller, Christoph W.Trends in Biochemical Sciences (2010), 35 (10), 565-574CODEN: TBSCDB; ISSN:0968-0004. (Elsevier Ltd.)A review. Recent findings indicate that WD40 domains play central roles in biol. processes by acting as hubs in cellular networks; however, they have been studied less intensely than other common domains, such as the kinase, PDZ or SH3 domains. As suggested by various interactome studies, they are among the most promiscuous interactors. Structural studies suggest that this property stems from their ability, as scaffolds, to interact with diverse proteins, peptides or nucleic acids using multiple surfaces or modes of interaction. A general scaffolding role is supported by the fact that no WD40 domain has been found with intrinsic enzymic activity despite often being part of large mol. machines. We discuss the WD40 domain distributions in protein networks and structures of WD40-contg. assemblies to demonstrate their versatility in mediating crit. cellular functions.
- 5Santosh Kumar, H. S.; Kumar, V.; Kumar, V.; Pattar, S.; Telkar, S. Towards the construction of an interactome for Human WD40 protein family. Bioinformation 2016, 12, 54– 61, DOI: 10.6026/973206300120545Towards the construction of an interactome for Human WD40 protein familySantosh Kumar Hulikal Shivashankara; Kumar Vadlapudi; Pattar Sharath; Telkar SandeepBioinformation (2016), 12 (2), 54-61 ISSN:0973-2063.WD40 proteins are involved in a variety of protein-protein interactions as part of a multi-protein assembly modulating diverse and critical cellular process. It is known that several proteins of this family have been implicated in different disorders such as developmental abnormalities and cancer. However, molecular functions of many proteins in this family are yet unknown and it is of clinical interest. Therefore, it is of interest to define, construct, understand, analyze, evaluate, redefine and refine an interactome for WD40 protein family. We used data from literature mining using Cytoscape followed by linear regression analysis between Betweenness centrality and stress scores to define a model to filter the nodes in a representative WD40 interactome construction. We identified 10 ranked nodes in this analysis and subsequent microarray data selected three of them in insulin resistance that is further demonstrated in HepG2 cell culture models. We also observed the expression of GRWD1, RBBP5 and WDR5 genes during perturbation. Thus, we report hub nodes of WD40 interactome in insulin resistance. It should be noted that the pipeline using protein interaction network help find new proteins of clinical importance.
- 6Migliori, V.; Mapelli, M.; Guccione, E. On WD40 proteins: propelling our knowledge of transcriptional control?. Epigenetics 2012, 7, 815– 822, DOI: 10.4161/epi.211406On WD40 proteins: propelling our knowledge of transcriptional control?Migliori, Valentina; Mapelli, Marina; Guccione, ErnestoEpigenetics (2012), 7 (8), 815-822CODEN: EPIGB6; ISSN:1559-2294. (Landes Bioscience)A review. A direct effect of post-translational modifications (PTMs) on nucleosomes is the formation of a dynamic platform able to assemble the transcriptional machinery and to recruit chromatin modifiers. The histone code hypothesis suggests that histone PTMs can act as binding sites for chromatin readers and effector proteins, such as the bromodomains, that selectively interact with acetylated lysines, or the "royal family" and the PHD finger domains, which are able to recognize methylated arginines and lysines. In this review we will discuss recent data describing the function of WD40 proteins as a new class of histone readers, with particular emphasis on the ones able to recognize methylated arginine and lysine residues. We will discuss how WDR5, a classical seven-bladed WD40 propeller, is able to bind with similar affinities both the catalytic subunit of the Trithorax-like complexes, and the histone H3 tail either unmodified or sym. dimethylated on arginine 2 (H3R2me2s). Furthermore, we will speculate on how these mutually exclusive interactions of WDR5 may play a role in mediating different degrees of H3K4 methylations at both promoters and distal regulatory sites. Finally, we will summarize recent literature elucidating how other WD40 proteins such as NURF55, EED and LRWD1 recognize methylated histone tails, highlighting similarities and differences among them.
- 7Jain, B. P.; Pandey, S. WD40 Repeat Proteins: Signalling Scaffold with Diverse Functions. Protein J. 2018, 37, 391– 406, DOI: 10.1007/s10930-018-9785-77WD40 Repeat Proteins: Signalling Scaffold with Diverse FunctionsJain, Buddhi Prakash; Pandey, ShwetaProtein Journal (2018), 37 (5), 391-406CODEN: PJROAH; ISSN:1572-3887. (Springer)A review. The WD40 domain is one of the most abundant and interacting domains in the eukaryotic genome. In proteins the WD domain folds into a β-propeller structure, providing a platform for the interaction and assembly of several proteins into a signalosome. WD40 repeats contg. proteins, in lower eukaryotes, are mainly involved in growth, cell cycle, development and virulence, while in higher organisms, they play an important role in diverse cellular functions like signal transduction, cell cycle control, intracellular transport, chromatin remodelling, cytoskeletal organization, apoptosis, development, transcriptional regulation, immune responses. To play the regulatory role in various processes, they act as a scaffold for protein-protein or protein-DNA interaction. So far, no WD40 domain has been identified with intrinsic enzymic activity. Several WD40 domain-contg. proteins have been recently characterized in prokaryotes as well. The review summarizes the vast array of functions performed by different WD40 domain contg. proteins, their domain organization and functional conservation during the course of evolution.
- 8Cosgrove, M. S.; Patel, A. Mixed lineage leukemia: a structure-function perspective of the MLL1 protein. FEBS J. 2010, 277, 1832– 1842, DOI: 10.1111/j.1742-4658.2010.07609.x8Mixed lineage leukemia: a structure-function perspective of the MLL1 proteinCosgrove, Michael S.; Patel, AnamikaFEBS Journal (2010), 277 (8), 1832-1842CODEN: FJEOAC; ISSN:1742-464X. (Wiley-Blackwell)A review. Several acute lymphoblastic and myelogenous leukemias are correlated with alterations in the human mixed lineage leukemia protein-1 (MLL1) gene. MLL1 is a member of the evolutionarily conserved SET1 family of histone H3 lysine 4 (H3K4) methyltransferases, which are required for the regulation of distinct groups of developmentally regulated genes in metazoans. Despite the important biol. role of SET1 family enzymes and their involvement in human leukemias, relatively little is understood about how these enzymes work. Here, the authors review several recent structural and biochem. studies that are beginning to shed light on the mol. mechanisms for the regulation of H3K4 methylation by the human MLL1 enzyme.
- 9Li, Y.; Han, J.; Zhang, Y.; Cao, F.; Liu, Z.; Li, S.; Wu, J.; Hu, C.; Wang, Y.; Shuai, J.; Chen, J.; Cao, L.; Li, D.; Shi, P.; Tian, C.; Zhang, J.; Dou, Y.; Li, G.; Chen, Y.; Lei, M. Structural basis for activity regulation of MLL family methyltransferases. Nature 2016, 530, 447– 452, DOI: 10.1038/nature169529Structural basis for activity regulation of MLL family methyltransferasesLi, Yanjing; Han, Jianming; Zhang, Yuebin; Cao, Fang; Liu, Zhijun; Li, Shuai; Wu, Jian; Hu, Chunyi; Wang, Yan; Shuai, Jin; Chen, Juan; Cao, Liaoran; Li, Dangsheng; Shi, Pan; Tian, Changlin; Zhang, Jian; Dou, Yali; Li, Guohui; Chen, Yong; Lei, MingNature (London, United Kingdom) (2016), 530 (7591), 447-452CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)The mixed lineage leukemia (MLL) family of proteins (including MLL1-MLL4, SET1A and SET1B) specifically methylate histone 3 Lys4, and have pivotal roles in the transcriptional regulation of genes involved in haematopoiesis and development. The methyltransferase activity of MLL1, by itself severely compromised, is stimulated by the three conserved factors WDR5, RBBP5 and ASH2L, which are shared by all MLL family complexes. However, the mol. mechanism of how these factors regulate the activity of MLL proteins still remains poorly understood. Here we show that a minimized human RBBP5-ASH2L heterodimer is the structural unit that interacts with and activates all MLL family histone methyltransferases. Our structural, biochem. and computational analyses reveal a two-step activation mechanism of MLL family proteins. These findings provide unprecedented insights into the common theme and functional plasticity in complex assembly and activity regulation of MLL family methyltransferases, and also suggest a universal regulation mechanism for most histone methyltransferases.
- 10Vedadi, M.; Blazer, L.; Eram, M. S.; Barsyte-Lovejoy, D.; Arrowsmith, C. H.; Hajian, T. Targeting human SET1/MLL family of proteins. Protein Sci. 2017, 26, 662– 676, DOI: 10.1002/pro.312910Targeting human SET1/MLL family of proteinsVedadi, Masoud; Blazer, Levi; Eram, Mohammad S.; Barsyte-Lovejoy, Dalia; Arrowsmith, Cheryl H.; Hajian, TaranehProtein Science (2017), 26 (4), 662-676CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)The SET1 family of proteins, and in particular MLL1, are essential regulators of transcription and key mediators of normal development and disease. Here, we summarize the detailed characterization of the methyltransferase activity of SET1 complexes and the role of the key subunits, WDR5, RbBP5, ASH2L, and DPY30. We present new data on full kinetic characterization of human MLL1, MLL3, SET1A, and SET1B trimeric, tetrameric, and pentameric complexes to elaborate on substrate specificities and compare our findings with what has been reported before. We also review exciting recent work identifying potent inhibitors of oncogenic MLL1 function through disruption of protein-protein interactions within the MLL1 complex.
- 11Xue, H.; Yao, T.; Cao, M.; Zhu, G.; Li, Y.; Yuan, G.; Chen, Y.; Lei, M.; Huang, J. Structural basis of nucleosome recognition and modification by MLL methyltransferases. Nature 2019, 573, 445– 449, DOI: 10.1038/s41586-019-1528-111Structural basis of nucleosome recognition and modification by MLL methyltransferasesXue, Han; Yao, Tonghui; Cao, Mi; Zhu, Guanjun; Li, Yan; Yuan, Guiyong; Chen, Yong; Lei, Ming; Huang, JingNature (London, United Kingdom) (2019), 573 (7774), 445-449CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Methyltransferases of the mixed-lineage leukemia (MLL) family-which include MLL1, MLL2, MLL3, MLL4, SET1A and SET1B-implement methylation of histone H3 on lysine 4 (H3K4), and have crit. and distinct roles in the regulation of transcription in haematopoiesis, adipogenesis and development. The C-terminal catalytic SET (Su(var.)3-9, enhancer of zeste and trithorax) domains of MLL proteins are assocd. with a common set of regulatory factors (WDR5, RBBP5, ASH2L and DPY30) to achieve specific activities. Current knowledge of the regulation of MLL activity is limited to the catalysis of histone H3 peptides, and how H3K4 Me marks are deposited on nucleosomes is poorly understood. H3K4 methylation is stimulated by mono-ubiquitination of histone H2B on lysine 120 (H2BK120ub1), a prevalent histone H2B mark that disrupts chromatin compaction and favors open chromatin structures, but the underlying mechanism remains unknown10-12. Here we report cryo-electron microscopy structures of human MLL1 and MLL3 catalytic modules assocd. with nucleosome core particles that contain H2BK120ub1 or unmodified H2BK120. These structures demonstrate that the MLL1 and MLL3 complexes both make extensive contacts with the histone-fold and DNA regions of the nucleosome; this allows ease of access to the histone H3 tail, which is essential for the efficient methylation of H3K4. The H2B-conjugated ubiquitin binds directly to RBBP5, orienting the assocn. between MLL1 or MLL3 and the nucleosome. The MLL1 and MLL3 complexes display different structural organizations at the interface between the WDR5, RBBP5 and MLL1 (or the corresponding MLL3) subunits, which accounts for the opposite roles of WDR5 in regulating the activity of the two enzymes. These findings transform our understanding of the structural basis for the regulation of MLL activity at the nucleosome level, and highlight the pivotal role of nucleosome regulation in histone-tail modification.
- 12Jiang, H. The complex activities of the SET1/MLL complex core subunits in development and disease. Biochim. Biophys. Acta, Gene Regul. Mech. 2020, 1863, 194560, DOI: 10.1016/j.bbagrm.2020.19456012The complex activities of the SET1/MLL complex core subunits in development and diseaseJiang, HaoBiochimica et Biophysica Acta, Gene Regulatory Mechanisms (2020), 1863 (7), 194560CODEN: BBAGC6; ISSN:1874-9399. (Elsevier B.V.)In mammalian cells, the SET1/MLL complexes are the main writers of the H3K4 Me mark that is assocd. with active gene expression. The activities of these complexes are critically dependent on the assocn. of the catalytic subunit with their shared core subunits, WDR5, RBBP5, ASH2L, and DPY30, collectively referred as WRAD. In addn., some of these core subunits can bind to proteins other than the SET1/MLL complex components. This review starts with discussion of the mol. activities of these core subunits, with an emphasis on DPY30 in organizing the assembly of the SET1/MLL complexes with other assocd. factors. This review then focuses on the roles of the core subunits in stem cells and development, as well as in diseased cell states, mainly cancer, and ends with discussion on dissecting the responsible activities of the core subunits and how we may target them for potential disease treatment. This article is part of a Special Issue entitled: The MLL family of proteins in normal development and disease edited by Thomas A Milne.
- 13Sha, L.; Ayoub, A.; Cho, U.-S.; Dou, Y. Insights on the regulation of the MLL/SET1 family histone methyltransferases. Biochim. Biophys. Acta, Gene Regul. Mech. 2020, 1863, 194561, DOI: 10.1016/j.bbagrm.2020.19456113Insights on the regulation of the MLL/SET1 family histone methyltransferasesSha, Liang; Ayoub, Alex; Cho, Uhn-Soo; Dou, YaliBiochimica et Biophysica Acta, Gene Regulatory Mechanisms (2020), 1863 (7), 194561CODEN: BBAGC6; ISSN:1874-9399. (Elsevier B.V.)A review. In eukaryotes, histone H3K4 methylation by the MLL/SET1 family histone methyltransferases is enriched at transcription regulatory elements including gene promoters and enhancers. The level of H3K4 methylation is highly correlated with transcription activation and is one of the most frequently used histone post-translational modifications to predict transcriptional outcome. Recently, it has been shown that rearrangement of the cellular landscape of H3K4 mono-methylation at distal enhancers precedes cell fate transition and is used for identification of novel regulatory elements for development and disease progression. Similarly, broad H3K4 tri-methylation regions have also been used to predict intrinsic tumor suppression properties of regulator regions in a variety of cellular models. Understanding the regulation for how H3K4 methylation is deposited and regulated is of paramount importance. In this review, we will discuss new findings on how the MLL/SET1 family enzymes are regulated on chromatin and their potential functional and regulatory implications. This article is part of a Special Issue entitled: The MLL family of proteins in normal development and disease edited by Thomas A Milne.
- 14Dou, Y.; Milne, T. A.; Ruthenburg, A. J.; Lee, S.; Lee, J. W.; Verdine, G. L.; Allis, C. D.; Roeder, R. G. Regulation of MLL1 H3K4 methyltransferase activity by its core components. Nat. Struct. Mol. Biol. 2006, 13, 713– 719, DOI: 10.1038/nsmb112814Regulation of MLL1 H3K4 methyltransferase activity by its core componentsDou, Yali; Milne, Thomas A.; Ruthenburg, Alexander J.; Lee, Seunghee; Lee, Jae Woon; Verdine, Gregory L.; Allis, C. David; Roeder, Robert G.Nature Structural & Molecular Biology (2006), 13 (8), 713-719CODEN: NSMBCU; ISSN:1545-9993. (Nature Publishing Group)Histone H3 Lys4 (H3K4) methylation is a prevalent mark assocd. with transcription activation. A common feature of several H3K4 methyltransferase complexes is the presence of three structural components (RbBP5, Ash2L and WDR5) and a catalytic subunit contg. a SET domain. Here we report the first biochem. reconstitution of a functional four-component mixed-lineage leukemia protein-1 (MLL1) core complex. This reconstitution, combined with in vivo assays, allows direct anal. of the contribution of each component to MLL1 enzymic activity and their roles in transcriptional regulation. Moreover, taking clues from a crystal structure anal., we demonstrate that WDR5 mediates interactions of the MLL1 catalytic unit both with the common structural platform and with the histone substrate. Mechanistic insights gained from this study can be generalized to the whole family of SET1-like histone methyltransferases in mammals.
- 15Patel, A.; Dharmarajan, V.; Vought, V. E.; Cosgrove, M. S. On the mechanism of multiple lysine methylation by the human mixed lineage leukemia protein-1 (MLL1) core complex. J. Biol. Chem. 2009, 284, 24242– 24256, DOI: 10.1074/jbc.m109.01449815On the Mechanism of Multiple Lysine Methylation by the Human Mixed Lineage Leukemia Protein-1 (MLL1) Core ComplexPatel, Anamika; Dharmarajan, Venkatasubramanian; Vought, Valarie E.; Cosgrove, Michael S.Journal of Biological Chemistry (2009), 284 (36), 24242-24256CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Transcription in eukaryotic genomes depends on enzymes that regulate the degree of histone H3 lysine 4 (H3K4) methylation. The mixed lineage leukemia protein-1 (MLL1) is a member of the SET1 family of H3K4 methyltransferases and is frequently rearranged in acute leukemias. Despite sequence comparisons that predict that SET1 family enzymes should only monomethylate their substrates, mono-, di-, and trimethylation of H3K4 has been attributed to SET1 family complexes in vivo and in vitro. To better understand this paradox, we have biochem. reconstituted and characterized a five-component 200-kDa MLL1 core complex contg. human MLL1, WDR5, RbBP5, Ash2L, and DPY-30. We demonstrate that the isolated MLL1 SET domain is a slow monomethyltransferase and that tyrosine 3942 of MLL1 prevents di- and trimethylation of H3K4. In contrast, a complex contg. the MLL1 SET domain, WDR5, RbBP5, Ash2L, and DPY-30, displays a marked ∼600-fold increase in enzymic activity but only to the di-Me form of H3K4. Single turnover kinetic expts. reveal that the reaction leading to H3K4 dimethylation involves the transient accumulation of a monomethylated species, suggesting that the MLL1 core complex uses a non-processive mechanism to catalyze multiple lysine methylation. We have also discovered that the non-SET domain components of the MLL1 core complex possess a previously unrecognized methyltransferase activity that catalyzes H3K4 dimethylation within the MLL1 core complex. Our results suggest that the mechanism of multiple lysine methylation by the MLL1 core complex involves the sequential addn. of two Me groups at two distinct active sites within the complex.
- 16Shinsky, S. A.; Monteith, K. E.; Viggiano, S.; Cosgrove, M. S. Biochemical reconstitution and phylogenetic comparison of human SET1 family core complexes involved in histone methylation. J. Biol. Chem. 2015, 290, 6361– 6375, DOI: 10.1074/jbc.m114.62764616Biochemical Reconstitution and Phylogenetic Comparison of Human SET1 Family Core Complexes Involved in Histone MethylationShinsky, Stephen A.; Monteith, Kelsey E.; Viggiano, Susan; Cosgrove, Michael S.Journal of Biological Chemistry (2015), 290 (10), 6361-6375CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Mixed lineage leukemia protein-1 (MLL1) is a member of the SET1 family of histone H3 lysine 4 (H3K4) methyltransferases that are required for metazoan development. MLL1 is the best characterized human SET1 family member, which includes MLL1-4 and SETd1A/B. MLL1 assembles with WDR5, RBBP5, ASH2L, DPY-30 (WRAD) to form the MLL1 core complex, which is required for H3K4 dimethylation and transcriptional activation. Because all SET1 family proteins interact with WRAD in vivo, it is hypothesized they are regulated by similar mechanisms. However, recent evidence suggests differences among family members that may reflect unique regulatory inputs in the cell. Missing is an understanding of the intrinsic enzymic activities of different SET1 family complexes under std. conditions. In this investigation, we reconstituted each human SET1 family core complex and compared subunit assembly and enzymic activities. We found that in the absence of WRAD, all but one SET domain catalyzes at least weak H3K4 monomethylation. In the presence of WRAD, all SET1 family members showed stimulated monomethyltransferase activity but differed in their di- and trimethylation activities. We found that these differences are correlated with evolutionary lineage, suggesting these enzyme complexes have evolved to accomplish unique tasks within metazoan genomes. To understand the structural basis for these differences, we employed a "phylogenetic scanning mutagenesis" assay and identified a cluster of amino acid substitutions that confer a WRAD-dependent gain-of-function dimethylation activity on complexes assembled with the MLL3 or Drosophila trithorax proteins. These results form the basis for understanding how WRAD differentially regulates SET1 family complexes in vivo.
- 17Thomas, L. R.; Wang, Q.; Grieb, B. C.; Phan, J.; Foshage, A. M.; Sun, Q.; Olejniczak, E. T.; Clark, T.; Dey, S.; Lorey, S.; Alicie, B.; Howard, G. C.; Cawthon, B.; Ess, K. C.; Eischen, C. M.; Zhao, Z.; Fesik, S. W.; Tansey, W. P. Interaction with WDR5 promotes target gene recognition and tumorigenesis by MYC. Mol. Cell. Biochem. 2015, 58, 440– 452, DOI: 10.1016/j.molcel.2015.02.028There is no corresponding record for this reference.
- 18Thomas, L. R.; Adams, C. M.; Wang, J.; Weissmiller, A. M.; Creighton, J.; Lorey, S. L.; Liu, Q.; Fesik, S. W.; Eischen, C. M.; Tansey, W. P. Interaction of the oncoprotein transcription factor MYC with its chromatin cofactor WDR5 is essential for tumor maintenance. Proc. Natl. Acad. Sci. U.S.A. 2019, 116, 25260– 25268, DOI: 10.1073/pnas.191039111618Interaction of the oncoprotein transcription factor MYC with its chromatin cofactor WDR5 is essential for tumor maintenanceThomas, Lance R.; Adams, Clare M.; Wang, Jing; Weissmiller, April M.; Creighton, Joy; Lorey, Shelly L.; Liu, Qi; Fesik, Stephen W.; Eischen, Christine M.; Tansey, William P.Proceedings of the National Academy of Sciences of the United States of America (2019), 116 (50), 25260-25268CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The oncoprotein transcription factor MYC is overexpressed in the majority of cancers. Key to its oncogenic activity is the ability of MYC to regulate gene expression patterns that drive and maintain the malignant state. MYC is also considered a validated anticancer target, but efforts to pharmacol. inhibit MYC have failed. The dependence of MYC on cofactors creates opportunities for therapeutic intervention, but for any cofactor this requires structural understanding of how the cofactor interacts with MYC, knowledge of the role it plays in MYC function, and demonstration that disrupting the cofactor interaction will cause existing cancers to regress. One cofactor for which structural information is available is WDR5, which interacts with MYC to facilitate its recruitment to chromatin. To explore whether disruption of the MY-WDR5 interaction could potentially become a viable anticancer strategy, we developed a Burkitt's lymphoma system that allows replacement of wild-type MYC for mutants that are defective for WDR5 binding or all known nuclear MYC functions. Using this system, we show that WDR5 recruits MYC to chromatin to control the expression of genes linked to biomass accumulation. We further show that disrupting the MYC-WDR5 interaction within the context of an existing cancer promotes rapid and comprehensive tumor regression in vivo. These observations connect WDR5 to a core tumorigenic function of MYC and establish that, if a therapeutic window can be established, MYC-WDR5 inhibitors could be developed as anticancer agents.
- 19Thomas, L. R.; Adams, C. M.; Fesik, S. W.; Eischen, C. M.; Tansey, W. P. Targeting MYC through WDR5. Mol. Cell. Oncol. 2020, 7, 1709388, DOI: 10.1080/23723556.2019.170938819Targeting MYC through WDR5Thomas Lance R; Tansey William P; Adams Clare M; Eischen Christine M; Fesik Stephen W; Tansey William PMolecular & cellular oncology (2020), 7 (2), 1709388 ISSN:2372-3556.The oncoprotein transcription factor MYC is overexpressed in most cancers and is responsible for hundreds of thousands of cancer deaths worldwide every year. MYC is also a highly validated - but currently undruggable - anti-cancer target. We recently showed that breaking the interaction of MYC with its chromatin co-factor WD repeat-containing protein 5 (WDR5) promotes tumor regression in mouse xenografts, laying the foundation for a new strategy to inhibit MYC in the clinic.
- 20Thomas, L. R.; Foshage, A. M.; Weissmiller, A. M.; Tansey, W. P. The MYC-WDR5 Nexus and Cancer. Cancer Res. 2015, 75, 4012– 4015, DOI: 10.1158/0008-5472.can-15-121620The MYC-WDR5 Nexus and CancerThomas, Lance R.; Foshage, Audra M.; Weissmiller, April M.; Tansey, William P.Cancer Research (2015), 75 (19), 4012-4015CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)The MYC oncogenes encode a family of transcription factors that feature prominently in cancer. MYC proteins are overexpressed or deregulated in a majority of malignancies and drive tumorigenesis by inducing widespread transcriptional reprogramming that promotes cell proliferation, metab., and genomic instability. The ability of MYC to regulate transcription depends on its dimerization with MAX, which creates a DNA-binding domain that recognizes specific sequences in the regulatory elements of MYC target genes. Recently, we discovered that recognition of target genes by MYC also depends on its interaction with WDR5, a WD40-repeat protein that exists as part of several chromatin-regulatory complexes. Here, we discuss how interaction of MYC with WDR5 could create an avidity-based chromatin recognition mechanism that allows MYC to select its target genes in response to both genetic and epigenetic determinants. We rationalize how the MYC-WDR5 interaction provides plasticity in target gene selection by MYC and speculate on the biochem. and genomic contexts in which this interaction occurs. Finally, we discuss how properties of the MYC-WDR5 interface make it an attractive point for discovery of small-mol. inhibitors of MYC function in cancer cells.
- 21Guarnaccia, A. D.; Rose, K. L.; Wang, J.; Zhao, B.; Popay, T. M.; Wang, C. E.; Guerrazzi, K.; Hill, S.; Woodley, C. M.; Hansen, T. J.; Lorey, S. L.; Shaw, J. G.; Payne, W. G.; Weissmiller, A. M.; Olejniczak, E. T.; Fesik, S. W.; Liu, Q.; Tansey, W. P. Impact of WIN site inhibitor on the WDR5 interactome. Cell Rep. 2021, 34, 108636, DOI: 10.1016/j.celrep.2020.10863621Impact of WIN site inhibitor on the WDR5 interactomeGuarnaccia, Alissa D.; Rose, Kristie L.; Wang, Jing; Zhao, Bin; Popay, Tessa M.; Wang, Christina E.; Guerrazzi, Kiana; Hill, Salisha; Woodley, Chase M.; Hansen, Tyler J.; Lorey, Shelly L.; Shaw, J. Grace; Payne, William G.; Weissmiller, April M.; Olejniczak, Edward T.; Fesik, Stephen W.; Liu, Qi; Tansey, William P.Cell Reports (2021), 34 (3), 108636CODEN: CREED8; ISSN:2211-1247. (Cell Press)The chromatin-assocd. protein WDR5 is a promising pharmacol. target in cancer, with most drug discovery efforts directed against an arginine-binding cavity in WDR5 called the WIN site. Despite a clear expectation that WIN site inhibitors will alter the repertoire of WDR5 interaction partners, their impact on the WDR5 interactome remains unknown. Here, we use quant. proteomics to delineate how the WDR5 interactome is changed by WIN site inhibition. We show that the WIN site inhibitor alters the interaction of WDR5 with dozens of proteins, including those linked to phosphatidylinositol 3-kinase (PI3K) signaling. As proof of concept, we demonstrate that the master kinase PDPK1 is a bona fide high-affinity WIN site binding protein that engages WDR5 to modulate transcription of genes expressed in the G2 phase of the cell cycle. This dataset expands our understanding of WDR5 and serves as a resource for deciphering the action of WIN site inhibitors.
- 22Patel, A.; Dharmarajan, V.; Cosgrove, M. S. Structure of WDR5 bound to mixed lineage leukemia protein-1 peptide. J. Biol. Chem. 2008, 283, 32158– 32161, DOI: 10.1074/jbc.c80016420022Structure of WDR5 Bound to Mixed Lineage Leukemia Protein-1 PeptidePatel, Anamika; Dharmarajan, Venkatasubramanian; Cosgrove, Michael S.Journal of Biological Chemistry (2008), 283 (47), 32158-32161CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The mixed lineage leukemia protein-1 (MLL1) catalyzes histone H3 lysine 4 methylation and is regulated by interaction with WDR5 (WD-repeat protein-5), RbBP5 (retinoblastoma-binding protein-5), and the Ash2L (absent, small, homeotic disks-2-like) oncoprotein. In the accompanying investigation, we describe the identification of a conserved arginine contg. motif, called the "Win" or WDR5 interaction motif, that is essential for the assembly and H3K4 dimethylation activity of the MLL1 core complex. Here we present a 1.7-Å crystal structure of WDR5 bound to a peptide derived from the MLL1 Win motif. Our results show that Arg-3765 of MLL1 is bound in the same arginine binding pocket on WDR5 that was previously suggested to bind histone H3. Thermodn. binding expts. show that the MLL1 Win peptide is preferentially recognized by WDR5. These results are consistent with a model in which WDR5 recognizes Arg-3765 of MLL1, which is essential for the assembly and enzymic activity of the MLL1 core complex.
- 23Patel, A.; Vought, V. E.; Dharmarajan, V.; Cosgrove, M. S. A conserved arginine-containing motif crucial for the assembly and enzymatic activity of the mixed lineage leukemia protein-1 core complex. J. Biol. Chem. 2008, 283, 32162– 32175, DOI: 10.1074/jbc.m80631720023A Conserved Arginine-containing Motif Crucial for the Assembly and Enzymatic Activity of the Mixed Lineage Leukemia Protein-1 Core ComplexPatel, Anamika; Vought, Valarie E.; Dharmarajan, Venkatasubramanian; Cosgrove, Michael S.Journal of Biological Chemistry (2008), 283 (47), 32162-32175CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The mixed lineage leukemia protein-1 (MLL1) belongs to the SET1 family of histone H3 lysine 4 methyltransferases. Recent studies indicate that the catalytic subunits of SET1 family members are regulated by interaction with a conserved core group of proteins that include the WD repeat protein-5 (WDR5), retinoblastoma-binding protein-5 (RbBP5), and the absent small homeotic-2-like protein (Ash2L). It has been suggested that WDR5 functions to bridge the interactions between the catalytic and regulatory subunits of SET1 family complexes. However, the mol. details of these interactions are unknown. To gain insight into the interactions among these proteins, we have detd. the biophys. basis for the interaction between the human WDR5 and MLL1. Our studies reveal that WDR5 preferentially recognizes a previously unidentified and conserved arginine-contg. motif, called the "Win" or WDR5 interaction motif, which is located in the N-SET region of MLL1 and other SET1 family members. Surprisingly, our structural and functional studies show that WDR5 recognizes arginine 3765 of the MLL1 Win motif using the same arginine-binding pocket on WDR5 that was previously shown to bind histone H3. We demonstrate that WDR5's recognition of arginine 3765 of MLL1 is essential for the assembly and enzymic activity of the MLL1 core complex in vitro.
- 24Song, J.-J.; Kingston, R. E. WDR5 interacts with mixed lineage leukemia (MLL) protein via the histone H3-binding pocket. J. Biol. Chem. 2008, 283, 35258– 35264, DOI: 10.1074/jbc.m80690020024WDR5 Interacts with Mixed Lineage Leukemia (MLL) Protein via the Histone H3-binding PocketSong, Ji-Joon; Kingston, Robert E.Journal of Biological Chemistry (2008), 283 (50), 35258-35264CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)WDR5 is a component of the mixed lineage leukemia (MLL) complex, which methylates lysine 4 of histone H3, and was identified as a methylated Lys-4 histone H3-binding protein. Here, we present a crystal structure of WDR5 bound to an MLL peptide. Surprisingly, we find that WDR5 utilizes the same pocket shown to bind histone H3 for this MLL interaction. Furthermore, the WDR5-MLL interaction is disrupted preferentially by mono- and di-methylated Lys-4 histone H3 over unmodified and tri-methylated Lys-4 histone H3. These data implicate a delicate interplay between the effector, WDR5, the catalytic subunit, MLL, and the substrate, histone H3, of the MLL complex. We suggest that the activity of the MLL complex might be regulated through this interplay.
- 25Odho, Z.; Southall, S. M.; Wilson, J. R. Characterization of a novel WDR5-binding site that recruits RbBP5 through a conserved motif to enhance methylation of histone H3 lysine 4 by mixed lineage leukemia protein-1. J. Biol. Chem. 2010, 285, 32967– 32976, DOI: 10.1074/jbc.m110.15992125Characterization of a novel WDR5-binding site that recruits RbBP5 through a conserved motif to enhance methylation of histone H3 lysine 4 by mixed lineage leukemia protein-1Odho, Zain; Southall, Stacey M.; Wilson, Jon R.Journal of Biological Chemistry (2010), 285 (43), 32967-32976CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Histone modification is well established as a fundamental mechanism driving the regulation of transcription, replication, and DNA repair through the control of chromatin structure. Likewise, it is apparent that incorrect targeting of histone modifications contributes to misregulated gene expression and hence to developmental disorders and diseases of genomic instability such as cancer. The KMT2 family of SET domain methyltransferases, typified by mixed lineage leukemia protein-1 (MLL1), is responsible for histone H3 lysine 4 methylation, a marker of active genes. To ensure that this modification is correctly targeted, a multiprotein complex assocs. with the methyltransferase and directs activity. We have identified a novel interaction site on the core complex protein WD repeat protein-5 (WDR5), and we mapped the complementary site on its partner retinoblastoma-binding protein-5 (RbBP5). We have characterized this interaction by x-ray crystallog. and show how it is fundamental to the assembly of the complex and to the regulation of methyltransferase activity. We show which region of RbBP5 contributes directly to mixed lineage leukemia activation, and we combine our structural and biochem. data to produce a model to show how WDR5 and RbBP5 act cooperatively to stimulate activity.
- 26Chacón Simon, S.; Wang, F.; Thomas, L. R.; Phan, J.; Zhao, B.; Olejniczak, E. T.; Macdonald, J. D.; Shaw, J. G.; Schlund, C.; Payne, W.; Creighton, J.; Stauffer, S. R.; Waterson, A. G.; Tansey, W. P.; Fesik, S. W. Discovery of WD Repeat-Containing Protein 5 (WDR5)-MYC Inhibitors Using Fragment-Based Methods and Structure-Based Design. J. Med. Chem. 2020, 63, 4315– 4333, DOI: 10.1021/acs.jmedchem.0c0022426Discovery of WD Repeat-Containing Protein 5 (WDR5)-MYC Inhibitors Using Fragment-Based Methods and Structure-Based DesignChacon Simon, Selena; Wang, Feng; Thomas, Lance R.; Phan, Jason; Zhao, Bin; Olejniczak, Edward T.; MacDonald, Jonathan D.; Shaw, J. Grace; Schlund, Caden; Payne, William; Creighton, Joy; Stauffer, Shaun R.; Waterson, Alex G.; Tansey, William P.; Fesik, Stephen W.Journal of Medicinal Chemistry (2020), 63 (8), 4315-4333CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The frequent deregulation of MYC and its elevated expression via multiple mechanisms drives cells to a tumorigenic state. Indeed, MYC is overexpressed in up to ~ 50% of human cancers and is considered a highly validated anticancer target. Recently, we discovered that WD repeat-contg. protein 5 (WDR5) binds to MYC and is a crit. cofactor required for the recruitment of MYC to its target genes and reported the first small mol. inhibitors of the WDR5-MYC interaction using structure-based design. These compds. display high binding affinity, but have poor physicochem. properties and are hence not suitable for in vivo studies. Herein, we conducted an NMR-based fragment screening to identify addnl. chem. matter and, using a structure-based approach, we merged a fragment hit with the previously reported sulfonamide series. Compds. in this series can disrupt the WDR5-MYC interaction in cells, and as a consequence, we obsd. a redn. of MYC localization to chromatin.
- 27Gao, J.; Chang, M. T.; Johnsen, H. C.; Gao, S. P.; Sylvester, B. E.; Sumer, S. O.; Zhang, H.; Solit, D. B.; Taylor, B. S.; Schultz, N.; Sander, C. 3D clusters of somatic mutations in cancer reveal numerous rare mutations as functional targets. Genome Med. 2017, 9, 4, DOI: 10.1186/s13073-016-0393-x273D clusters of somatic mutations in cancer reveal numerous rare mutations as functional targetsGao, Jianjiong; Chang, Matthew T.; Johnsen, Hannah C.; Gao, Sizhi Paul; Sylvester, Brooke E.; Sumer, Selcuk Onur; Zhang, Hongxin; Solit, David B.; Taylor, Barry S.; Schultz, Nikolaus; Sander, ChrisGenome Medicine (2017), 9 (), 4/1-4/13CODEN: GMEECG; ISSN:1756-994X. (BioMed Central Ltd.)Many mutations in cancer are of unknown functional significance. Std. methods use statistically significant recurrence of mutations in tumor samples as an indicator of functional impact. We extend such analyses into the long tail of rare mutations by considering recurrence of mutations in clusters of spatially close residues in protein structures. Analyzing 10,000 tumor exomes, we identify more than 3000 rarely mutated residues in proteins as potentially functional and exptl. validate several in RAC1 and MAP2K1. These potential driver mutations (web resources: 3dhotspots.org and cBioPortal.org) can extend the scope of genomically informed clin. trials and of personalized choice of therapy.
- 28Acuner, S. E.; Sumbul, F.; Torun, H.; Haliloglu, T. Oncogenic mutations on Rac1 affect global intrinsic dynamics underlying GTP and PAK1 binding. Biophys. J. 2021, 120, 866– 876, DOI: 10.1016/j.bpj.2021.01.01628Oncogenic mutations on Rac1 affect global intrinsic dynamics underlying GTP and PAK1 bindingAcuner, Saliha Ece; Sumbul, Fidan; Torun, Hamdi; Haliloglu, TurkanBiophysical Journal (2021), 120 (5), 866-876CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)Rac1 is a small member of the Rho GTPase family. One of the most important downstream effectors of Rac1 is a serine/threonine kinase, p21-activated kinase 1 (PAK1). Mutational activation of PAK1 by Rac1 has oncogenic signaling effects. Here, although we focus on Rac1-PAK1 interaction by at.-force-microscopy-based single-mol. force spectroscopy expts., we explore the effect of active mutations on the intrinsic dynamics and binding interactions of Rac1 by Gaussian network model anal. and mol. dynamics simulations. We observe that Rac1 oncogenic mutations are at the hinges of three global modes of motion, suggesting the mech. changes as potential markers of oncogenicity. Indeed, the dissocn. of wild-type Rac1-PAK1 complex shows two distinct unbinding dynamic states that are reduced to one with constitutively active Q61L and oncogenic Y72C mutant Rac1, as revealed by single-mol. force spectroscopy expts. Q61L and Y72C mutations change the mechanics of the Rac1-PAK1 complex by increasing the elasticity of the protein and slowing down the transition to the unbound state. On the other hand, Rac1's intrinsic dynamics reveal more flexible GTP and PAK1-binding residues on switches I and II with Q61L, Y72C, oncogenic P29S and Q61R, and neg. T17N mutations. The cooperativity in the fluctuations of GTP-binding sites around the p-loop and switch I decreases in all mutants, mostly in Q61L, whereas some PAK1-binding residues display enhanced coupling with GTP-binding sites in Q61L and Y72C and within each other in P29S. The predicted binding free energies of the modeled Rac1-PAK1 complexes show that the change in the dynamic behavior likely means a more favorable PAK1 interaction. Overall, these findings suggest that the active mutations affect intrinsic functional dynamic events and alter the mechanics underlying the binding of Rac1 to GTP and upstream and downstream partners including PAK1.
- 29Forbes, S. A.; Beare, D.; Bindal, N.; Bamford, S.; Ward, S.; Cole, C. G.; Jia, M.; Kok, C.; Boutselakis, H.; De, T.; Sondka, Z.; Ponting, L.; Stefancsik, R.; Harsha, B.; Tate, J.; Dawson, E.; Thompson, S.; Jubb, H.; Campbell, P. J. COSMIC: High-Resolution Cancer Genetics Using the Catalogue of Somatic Mutations in Cancer. Curr. Protoc. Hum. Genet. 2016, 91, 10– 37, DOI: 10.1002/cphg.2129COSMIC: High-Resolution Cancer Genetics Using the Catalogue of Somatic Mutations in CancerForbes S A; Beare D; Bindal N; Bamford S; Ward S; Cole C G; Jia M; Kok C; Boutselakis H; De T; Sondka Z; Ponting L; Stefancsik R; Harsha B; Tate J; Dawson E; Thompson S; Jubb H; Campbell P JCurrent protocols in human genetics (2016), 91 (), 10.11.1-10.11.37 ISSN:.COSMIC (http://cancer.sanger.ac.uk) is an expert-curated database of somatic mutations in human cancer. Broad and comprehensive in scope, recent releases in 2016 describe over 4 million coding mutations across all human cancer disease types. Mutations are annotated across the entire genome, but expert curation is focused on over 400 key cancer genes. Now encompassing the majority of molecular mutation mechanisms in oncogenetics, COSMIC additionally describes 10 million non-coding mutations, 1 million copy-number aberrations, 9 million gene-expression variants, and almost 8 million differentially methylated CpGs. This information combines a consistent interpretation of the data from the major cancer genome consortia and cancer genome literature with exhaustive hand curation of over 22,000 gene-specific literature publications. This unit describes the graphical Web site in detail; alternative protocols overview other ways the entire database can be accessed, analyzed, and downloaded. © 2016 by John Wiley & Sons, Inc.
- 30Tate, J. G.; Bamford, S.; Jubb, H. C.; Sondka, Z.; Beare, D. M.; Bindal, N.; Boutselakis, H.; Cole, C. G.; Creatore, C.; Dawson, E.; Fish, P.; Harsha, B.; Hathaway, C.; Jupe, S. C.; Kok, C. Y.; Noble, K.; Ponting, L.; Ramshaw, C. C.; Rye, C. E.; Speedy, H. E.; Stefancsik, R.; Thompson, S. L.; Wang, S.; Ward, S.; Campbell, P. J.; Forbes, S. A. COSMIC: the Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res. 2019, 47, D941– d947, DOI: 10.1093/nar/gky101530COSMIC: the Catalogue Of Somatic Mutations In CancerTate, John G.; Bamford, Sally; Jubb, Harry C.; Sondka, Zbyslaw; Beare, David M.; Bindal, Nidhi; Boutselakis, Harry; Cole, Charlotte G.; Creatore, Celestino; Dawson, Elisabeth; Fish, Peter; Harsha, Bhavana; Hathaway, Charlie; Jupe, Steve C.; Kok, Chai Yin; Noble, Kate; Ponting, Laura; Ramshaw, Christopher C.; Rye, Claire E.; Speedy, Helen E.; Stefancsik, Ray; Thompson, Sam L.; Wang, Shicai; Ward, Sari; Campbell, Peter J.; Forbes, Simon A.Nucleic Acids Research (2019), 47 (D1), D941-D947CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)COSMIC, the Catalog Of Somatic Mutations In Cancer (https://cancer.sanger.ac.uk) is the most detailed and comprehensive resource for exploring the effect of somatic mutations in human cancer. The latest release, COSMIC v86 (August 2018), includes almost 6 million coding mutations across 1.4 million tumor samples, curated from over 26,000 publications. In addn. to coding mutations, COSMIC covers all the genetic mechanisms by which somatic mutations promote cancer, including non-coding mutations, gene fusions, copy-no. variants and drug resistance mutations. COSMIC is primarily handcurated, ensuring quality, accuracy and descriptive data capture. Building on our manual curation processes, we are introducing new initiatives that allow us to prioritize key genes and diseases, and to react more quickly and comprehensively to new findings in the literature. Alongside improvements to the public website and data-download systems, new functionality in COSMIC-3D allows exploration of mutations within three-dimensional protein structures, their protein structural and functional impacts, and implications for druggability. In parallel with COSMIC's deep and broad variant coverage, the Cancer Gene Census (CGC) describes a curated catalog of genes driving every form of human cancer. Currently describing 719 genes, the CGC has recently introduced functional descriptions of how each gene drives disease, summarized into the 10 cancer Hallmarks.
- 31Torkamani, A.; Schork, N. J. Prediction of cancer driver mutations in protein kinases. Cancer Res. 2008, 68, 1675– 1682, DOI: 10.1158/0008-5472.can-07-528331Prediction of Cancer Driver Mutations in Protein KinasesTorkamani, Ali; Schork, Nicholas J.Cancer Research (2008), 68 (6), 1675-1682CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)A large no. of somatic mutations accumulate during the process of tumorigenesis. A subset of these mutations contribute to tumor progression (known as "driver" mutations) whereas the majority of these mutations are effectively neutral (known as "passenger" mutations). The ability to differentiate between drivers and passengers will be crit. to the success of upcoming large-scale cancer DNA resequencing projects. Here we show a method capable of discriminating between drivers and passengers in the most frequently cancer-assocd. protein family, protein kinases. We apply this method to multiple cancer data sets, validating its accuracy by showing that it is capable of identifying known drivers, has excellent agreement with previous statistical ests. of the frequency of drivers, and provides strong evidence that predicted drivers are under pos. selection by various sequence and structural analyses. Furthermore, we identify particular positions in protein kinases that seem to play a role in oncogenesis. Finally, we provide a ranked list of candidate driver mutations.
- 32Ryslik, G. A.; Cheng, Y.; Modis, Y.; Zhao, H. Leveraging protein quaternary structure to identify oncogenic driver mutations. BMC Bioinf. 2016, 17, 137, DOI: 10.1186/s12859-016-0963-332Leveraging protein quaternary structure to identify oncogenic driver mutationsRyslik, Gregory A.; Cheng, Yuwei; Modis, Yorgo; Zhao, HongyuBMC Bioinformatics (2016), 17 (), 137/1-137/13CODEN: BBMIC4; ISSN:1471-2105. (BioMed Central Ltd.)Background: Identifying key "driver" mutations which are responsible for tumorigenesis is crit. in the development of new oncol. drugs. Due to multiple pharmacol. successes in treating cancers that are caused by such driver mutations, a large body of methods have been developed to differentiate these mutations from the benign "passenger" mutations which occur in the tumor but do not further progress the disease. Under the hypothesis that driver mutations tend to cluster in key regions of the protein, the development of algorithms that identify these clusters has become a crit. area of research. Results: We have developed a novel methodol., QuartPAC (Quaternary Protein Amino acid Clustering), that identifies non-random mutational clustering while utilizing the protein quaternary structure in 3D space. By integrating the spatial information in the Protein Data Bank (PDB) and the mutational data in the Catalog of Somatic Mutations in Cancer (COSMIC), QuartPAC is able to identify clusters which are otherwise missed in a variety of proteins. The R package is available on Bioconductor at: http://bioconductor.jp/packages/3.1/bioc/html/QuartPAC.html. Conclusion:QuartPAC provides a unique tool to identify mutational clustering while accounting for the complete folded protein quaternary structure.
- 33Kumar, S.; Clarke, D.; Gerstein, M. B. Leveraging protein dynamics to identify cancer mutational hotspots using 3D structures. Proc. Natl. Acad. Sci. U.S.A. 2019, 116, 18962– 18970, DOI: 10.1073/pnas.190115611633Leveraging protein dynamics to identify cancer mutational hotspots using 3D structuresKumar, Sushant; Clarke, Declan; Gerstein, Mark B.Proceedings of the National Academy of Sciences of the United States of America (2019), 116 (38), 18962-18970CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Large-scale exome sequencing of tumors has enabled the identification of cancer drivers using recurrence-based approaches. Some of these methods also employ 3D protein structures to identify mutational hotspots in cancer-assocd. genes. In detg. such mutational clusters in structures, existing approaches overlook protein dynamics, despite its essential role in protein function. We present a framework to identify cancer driver genes using a dynamics-based search of mutational hotspot communities. Mutations are mapped to protein structures, which are partitioned into distinct residue communities. These communities are identified in a framework where residue-residue contact edges are weighted by correlated motions (as inferred by dynamics-based models). We then search for signals of pos. selection among these residue communities to identify putative driver genes, while applying our method to the TCGA (The Cancer Genome Atlas) PanCancer Atlas missense mutation catalog. Overall, we predict 1 or more mutational hotspots within the resolved structures of proteins encoded by 434 genes. These genes were enriched among biol. processes assocd. with tumor progression. Addnl., a comparison between our approach and existing cancer hotspot detection methods using structural data suggests that including protein dynamics significantly increases the sensitivity of driver detection.
- 34Kamburov, A.; Lawrence, M. S.; Polak, P.; Leshchiner, I.; Lage, K.; Golub, T. R.; Lander, E. S.; Getz, G. Comprehensive assessment of cancer missense mutation clustering in protein structures. Proc. Natl. Acad. Sci. U.S.A. 2015, 112, E5486– E5495, DOI: 10.1073/pnas.151637311234Comprehensive assessment of cancer missense mutation clustering in protein structuresKamburov, Atanas; Lawrence, Michael S.; Polak, Paz; Leshchiner, Ignaty; Lage, Kasper; Golub, Todd R.; Lander, Eric S.; Getz, GadProceedings of the National Academy of Sciences of the United States of America (2015), 112 (40), E5486-E5495CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Large-scale tumor sequencing projects enabled the identification of many new cancer gene candidates through computational approaches. Here, we describe a general method to detect cancer genes based on significant 3D clustering of mutations relative to the structure of the encoded protein products. The approach can also be used to search for proteins with an enrichment of mutations at binding interfaces with a protein, nucleic acid, or small mol. partner. We applied this approach to systematically analyze the PanCancer compendium of somatic mutations from 4,742 tumors relative to all known 3D structures of human proteins in the Protein Data Bank. We detected significant 3D clustering of missense mutations in several previously known oncoproteins including HRAS, EGFR, and PIK3CA. Although clustering of missense mutations is often regarded as a hallmark of oncoproteins, we obsd. that a no. of tumor suppressors, including FBXW7, VHL, and STK11, also showed such clustering. Beside these known cases, we also identified significant 3D clustering of missense mutations in NUF2, which encodes a component of the kinetochore, that could affect chromosome segregation and lead to aneuploidy. Anal. of interaction interfaces revealed enrichment of mutations in the interfaces between FBXW7-CCNE1, HRAS-RASA1, CUL4B-CAND1, OGT-HCFC1, PPP2R1A-PPP2R5C/PPP2R2A, DICER1-Mg2+, MAX-DNA, SRSF2-RNA, and others. Together, our results indicate that systematic consideration of 3D structure can assist in the identification of cancer genes and in the understanding of the functional role of their mutations.
- 35Dharmarajan, V.; Lee, J.-H.; Patel, A.; Skalnik, D. G.; Cosgrove, M. S. Structural basis for WDR5 interaction (Win) motif recognition in human SET1 family histone methyltransferases. J. Biol. Chem. 2012, 287, 27275– 27289, DOI: 10.1074/jbc.m112.36412535Structural Basis for WDR5 Interaction (Win) Motif Recognition in Human SET1 Family Histone MethyltransferasesDharmarajan, Venkatasubramanian; Lee, Jeong-Heon; Patel, Anamika; Skalnik, David G.; Cosgrove, Michael S.Journal of Biological Chemistry (2012), 287 (33), 27275-27289CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Translocations and amplifications of the mixed lineage leukemia-1 (MLL1) gene are assocd. with aggressive myeloid and lymphocytic leukemias in humans. MLL1 is a member of the SET1 family of histone H3 lysine 4 (H3K4) methyltransferases, which are required for transcription of genes involved in hematopoiesis and development. MLL1 assocs. with a subcomplex contg. WDR5, RbBP5, Ash2L, and DPY-30 (WRAD), which together form the MLL1 core complex that is required for sequential mono- and dimethylation of H3K4. We previously demonstrated that WDR5 binds the conserved WDR5 interaction (Win) motif of MLL1 in vitro, an interaction that is required for the H3K4 dimethylation activity of the MLL1 core complex. In this investigation, we demonstrate that arginine 3765 of the MLL1 Win motif is required to co-immunoppt. WRAD from mammalian cells, suggesting that the WDR5-Win motif interaction is important for the assembly of the MLL1 core complex in vivo. We also demonstrate that peptides that mimic SET1 family Win motif sequences inhibit H3K4 dimethylation by the MLL1 core complex with varying degrees of efficiency. To understand the structural basis for these differences, we detd. structures of WDR5 bound to six different naturally occurring Win motif sequences at resolns. ranging from 1.9 to 1.2 Å. Our results reveal that binding energy differences result from interactions between non-conserved residues C-terminal to the Win motif and to a lesser extent from subtle variation of residues within the Win motif. These results highlight a new class of methylation inhibitors that may be useful for the treatment of MLL1-related malignancies.
- 36Zhang, P.; Lee, H.; Brunzelle, J. S.; Couture, J.-F. The plasticity of WDR5 peptide-binding cleft enables the binding of the SET1 family of histone methyltransferases. Nucleic Acids Res. 2012, 40, 4237– 4246, DOI: 10.1093/nar/gkr123536The plasticity of WDR5 peptide-binding cleft enables the binding of the SET1 family of histone methyltransferasesZhang, Pamela; Lee, Hwabin; Brunzelle, Joseph S.; Couture, Jean-FrancoisNucleic Acids Research (2012), 40 (9), 4237-4246CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)In mammals, the SET1 family of lysine methyltransferases (KMTs), which includes MLL1-5, SET1A and SET1B, catalyzes the methylation of lysine-4 (Lys-4) on histone H3. Recent reports have demonstrated that a three-subunit complex composed of WD-repeat protein-5 (WDR5), retinoblastoma-binding protein-5 (RbBP5) and absent, small, homeotic disks-2-like (ASH2L) stimulates the methyltransferase activity of MLL1. On the basis of studies showing that this stimulation is in part controlled by an interaction between WDR5 and a small region located in close proximity of the MLL1 catalytic domain [referred to as the WDR5-interacting motif (Win)], it has been suggested that WDR5 might play an analogous role in scaffolding the other SET1 complexes. We herein provide biochem. and structural evidence showing that WDR5 binds the Win motifs of MLL2-4, SET1A and SET1B. Comparative anal. of WDR5-Win complexes reveals that binding of the Win motifs is achieved by the plasticity of WDR5 peptidyl-arginine-binding cleft allowing the C-terminal ends of the Win motifs to be maintained in structurally divergent conformations. Consistently, enzymic assays reveal that WDR5 plays an important role in the optimal stimulation of MLL2-4, SET1A and SET1B methyltransferase activity by the RbBP5-ASH2L heterodimer. Overall, our findings illustrate the function of WDR5 in scaffolding the SET1 family of KMTs and further emphasize on the important role of WDR5 in regulating global histone H3 Lys-4 methylation.
- 37Muntean, A. G.; Hess, J. L. The pathogenesis of mixed-lineage leukemia. Annu. Rev. Pathol.: Mech. Dis. 2012, 7, 283– 301, DOI: 10.1146/annurev-pathol-011811-13243437The pathogenesis of mixed-lineage leukemiaMuntean, Andrew G.; Hess, Jay L.Annual Review of Pathology: Mechanisms of Disease (2012), 7 (), 283-301CODEN: ARPMCU; ISSN:1553-4006. (Annual Reviews Inc.)A review. Aggressive leukemias arise in both children and adults as a result of rearrangements to the mixed-lineage leukemia gene (MLL) located on chromosome 11q23. MLL encodes a large histone methyltransferase that directly binds DNA and pos. regulates gene transcription, including homeobox (HOX) genes. MLL is involved in chromosomal translocations, partial tandem duplications, and amplifications, all of which result in hematopoietic malignancies due to sustained HOX expression and stalled differentiation. MLL lesions are assocd. with both acute myeloid leukemia and acute lymphoid leukemia and are usually assocd. with a relatively poor prognosis despite improved treatment options such as allogeneic hematopoietic stem cell transplantation, which underscores the need for new treatment regimens. Recent advances have begun to reveal the mol. mechanisms that drive MLL-assocd. leukemias, which, in turn, have provided opportunities for therapeutic development. Here, we discuss the etiol. of MLL leukemias and potential directions for future therapy.
- 38Chen, X.; Xie, W.; Gu, P.; Cai, Q.; Wang, B.; Xie, Y.; Dong, W.; He, W.; Zhong, G.; Lin, T.; Huang, J. Upregulated WDR5 promotes proliferation, self-renewal and chemoresistance in bladder cancer via mediating H3K4 trimethylation. Sci. Rep. 2015, 5, 8293, DOI: 10.1038/srep0829338Upregulated WDR5 promotes proliferation, self-renewal and chemoresistance in bladder cancer via mediating H3K4 trimethylationChen, Xu; Xie, Weibin; Gu, Peng; Cai, Qingqing; Wang, Bo; Xie, Yun; Dong, Wen; He, Wang; Zhong, Guangzheng; Lin, Tianxin; Huang, JianScientific Reports (2015), 5 (), 8293CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)WD repeat domain 5 (WDR5) plays an important role in various biol. functions through the epigenetic regulation of gene transcription; however, its role in bladder cancer remains largely unknown. Our study investigated the role of WDR5 in bladder cancer and demonstrated that WDR5 was upregulated in bladder cancer tissues, and elevated WDR5 protein levels pos. correlated with advanced tumor stage and poor survival. Through gain or loss of function, we demonstrated that WDR5 promoted proliferation, self-renewal and chemoresistance to cisplatin in bladder cancer cells in vitro, and tumor growth in vivo. Mechanistically, WDR5 regulated various functions in bladder cancer by mediating the transcription of cyclin B1, cyclin E1, cyclin E2, UHMK1, MCL1, BIRC3 and Nanog by histone H3 lysine 4 trimethylation. Therefore, we have discovered that WDR5 plays an important role in bladder cancer suggesting that WDR5 is a potential biomarker and a promising target in the treatment of bladder cancer.
- 39Chen, X.; Gu, P.; Li, K.; Xie, W.; Chen, C.; Lin, T.; Huang, J. Gene expression profiling of WDR5 regulated genes in bladder cancer. Genomics Data 2015, 5, 27– 29, DOI: 10.1016/j.gdata.2015.05.00339Gene expression profiling of WDR5 regulated genes in bladder cancerChen Xu; Gu Peng; Lin Tianxin; Li Kuiqing; Xie Weibin; Chen Changhao; Huang JianGenomics data (2015), 5 (), 27-9 ISSN:2213-5960.WD repeat domain 5 (WDR5) plays an important role in various biological functions through the epigenetic regulation of gene transcription (Wysocka et al., 2005 [1]; Sandstrom et al., 2014[2]; Ang et al., 2011[3]). Recently, our study found that WDR5 was upregulated in bladder cancer tissues, promoted bladder cancer cell proliferation, self-renewal and chemoresistance to cisplatin in bladder cancer cells in vitro, and tumor growth in vivo (Chen et al., 2015). To gain a molecular understanding of the role of WDR5 in promoting bladder cancer, we performed a genome-wide analysis on WDR5 knockdown by microarray gene expression profiling. Here we provide detailed experimental methods and analysis for the microarray data, which have been deposited into Gene Expression Omnibus (GEO): GSE59132.
- 40Ge, Z.; Song, E. J.; Kawasawa, Y. I.; Li, J.; Dovat, S.; Song, C. WDR5 high expression and its effect on tumorigenesis in leukemia. Oncotarget 2016, 7, 37740– 37754, DOI: 10.18632/oncotarget.931240WDR5 high expression and its effect on tumorigenesis in leukemiaGe Zheng; Ge Zheng; Song Evelyn J; Dovat Sinisa; Song Chunhua; Ge Zheng; Li Jianyong; Kawasawa Yuka ImamuraOncotarget (2016), 7 (25), 37740-37754 ISSN:.WD repeat domain 5 (WDR5) plays an important role in various biological functions through the epigenetic regulation of gene transcription. However, the oncogenic effect of WDR5 in leukemia remains largely unknown. Here, we found WDR5 expression is increased in leukemia patients. High expression of WDR5 is associated with high risk leukemia; Patients with WDR5 and MLL1 high expression have poor complete remission rate. We further identified the global genomic binding of WDR5 in leukemic cells and found the genomic co-localization of WDR5 binding with H3K4me3 enrichment. Moreover, WDR5 knockdown by shRNA suppresses cell proliferation, induces apoptosis, inhibits the expression of WDR5 targets, and blocks the H3K4me3 enrichment on the promoter of its targets. We also observed the positive correlation of WDR5 expression with these targets in the cohort study of leukemia patients. Our data reveal that WDR5 may have oncogenic effect and WDR5-mediated H3K4 methylation plays an important role in leukemogenesis.
- 41Sun, W.; Guo, F.; Liu, M. Up-regulated WDR5 promotes gastric cancer formation by induced cyclin D1 expression. J. Cell. Biochem. 2018, 119, 3304– 3316, DOI: 10.1002/jcb.2649141Up-regulated WDR5 promotes gastric cancer formation by induced cyclin D1 expressionSun, Wei; Guo, Fuchao; Liu, MingkaiJournal of Cellular Biochemistry (2018), 119 (4), 3304-3316CODEN: JCEBD5; ISSN:0730-2312. (Wiley-Blackwell)Gastric cancer (GC) is the fourth common cancer and second leading cause of cancer-related mortality in the world. WD repeat domain 5 (WDR5) has been identified that its functions as an important role in various biol. functions through the epigenetic regulation of gene transcription. However, the oncogenic effect of WDR5 in gastric cancer remains largely unknown. In this study, we investigated the role of WDR5 in gastric cancer genesis. We found that WDR5 expression is increased in gastric cancer patients. Through survival anal., we found that high expression of WDR5 is assocd. with high risk gastric cancer; patients who with WDR5 high expression have poor survival rate compared with those who with WDR5 low expression. To make further investigation, we identified that WDR5 is targeted for cell cycle arrest by the Cyclin D1 in a process that is regulated by H3K4me3. Moreover, over-expression of WDR5 promotes cell proliferation, induces S/G2/M arrest in cell cycle, and promotes the expression of WDR5 targets, as well as that of H3K4me3 on the promoter of its targets. Inversely, WDR5 knockdown by shRNA inhibits cell proliferation, reverses S/G2/M arrest in cell cycle, and suppresses the expression of WDR5 targets, as well as that of H3K4me3. We also obsd. the pos. correlation of WDR5 expression with its target in the cohort study of gastric patients. Taken together, our data reveal that WDR5 may have oncogenic effect and WDR5-mediated H3K4 methylation plays an important role in gastric cancer.
- 42Wang, F.; Zhang, J.; Ke, X.; Peng, W.; Zhao, G.; Peng, S.; Xu, J.; Xu, B.; Cui, H. WDR5-Myc axis promotes the progression of glioblastoma and neuroblastoma by transcriptional activating CARM1. Biochem. Biophys. Res. Commun. 2020, 523, 699– 706, DOI: 10.1016/j.bbrc.2019.12.10142WDR5-Myc axis promotes the progression of glioblastoma and neuroblastoma by transcriptional activating CARM1Wang, Feng; Zhang, Jiayi; Ke, Xiaoxue; Peng, Wen; Zhao, Gaichao; Peng, Shihan; Xu, Jie; Xu, Bo; Cui, HongjuanBiochemical and Biophysical Research Communications (2020), 523 (3), 699-706CODEN: BBRCA9; ISSN:0006-291X. (Elsevier B.V.)The WD repeat domain 5 (WDR5), also known as SWD3 and BIG-3, is often overexpressed in cancers, however its mol. function in cancer remains to be elucidated. In this study, we found that WDR5 promoted the proliferation and self-renewal of glioblastoma and neuroblastoma cells. The data from databases and Western blot assay showed that CARM1 is a downstream gene of WDR5-Myc axis. In addn., we obsd. that WDR5 promoted the binding of Myc to CARM1 promoter by interacting with Myc and inducing histone 3 lysine 4 trimethylation (H3K4me3). Dual luciferase reporter system indicated that Myc binds to the upstream region (-520 to -515) before transcription start site (TSS) of CARM1 promoter. These findings suggest a novel regulatory model for the proliferation and tumorigenesis of glioblastoma and neuroblastoma by WDR5-Myc axis.
- 43Bryan, A. F.; Wang, J.; Howard, G. C.; Guarnaccia, A. D.; Woodley, C. M.; Aho, E. R.; Rellinger, E. J.; Matlock, B. K.; Flaherty, D. K.; Lorey, S. L.; Chung, D. H.; Fesik, S. W.; Liu, Q.; Weissmiller, A. M.; Tansey, W. P. WDR5 is a conserved regulator of protein synthesis gene expression. Nucleic Acids Res. 2020, 48, 2924– 2941, DOI: 10.1093/nar/gkaa05143WDR5 is a conserved regulator of protein synthesis gene expressionBryan, Audra F.; Wang, Jing; Howard, Gregory C.; Guarnaccia, Alissa D.; Woodley, Chase M.; Aho, Erin R.; Rellinger, Eric J.; Matlock, Brittany K.; Flaherty, David K.; Lorey, Shelly L.; Chung, Dai H.; Fesik, Stephen W.; Liu, Qi; Weissmiller, April M.; Tansey, William P.Nucleic Acids Research (2020), 48 (6), 2924-2941CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)WDR5 is a highly-conserved nuclear protein that performs multiple scaffolding functions in the context of chromatin. WDR5 is also a promising target for pharmacol. inhibition in cancer, with small mol. inhibitors of an arginine-binding pocket of WDR5 (the 'WIN' site) showing efficacy against a range of cancer cell lines in vitro. Efforts to understand WDR5, or establish the mechanism of action of WIN site inhibitors, however, are stymied by its many functions in the nucleus, and a lack of knowledge of the conserved gene networks--if any--that are under its control. Here, we have performed comparative genomic analyses to identify the conserved sites of WDR5 binding to chromatin, and the conserved genes regulated by WDR5, across a diverse panel of cancer cell lines. We show that a specific cohort of protein synthesis genes (PSGs) are invariantly bound by WDR5, demonstrate that the WIN site anchors WDR5 to chromatin at these sites, and establish that PSGs are bona fide, acute, and persistent targets of WIN site blockade. Together, these data reveal that WDR5 plays a predominant transcriptional role in biomass accumulation and provide further evidence that WIN site inhibitors act to repress gene networks linked to protein synthesis homeostasis. In Saccharomyces cerevisiae, ribosomal protein gene (RPG) promoters display binding sites for either Rap1 or Abf1 transcription factors. Unlike Rap1-assocd. promoters, the small cohort of Abf1-dependent RPGs (Abf1-RPGs) has not been extensively investigated. We show that RPL3, RPL4B, RPP1A, RPS22B and RPS28A/B share a common promoter architecture, with an Abf1 site upstream of a conserved element matching the sequence recognized by Fhl1, a transcription factor which together with Ifh1 orchestrates Rap1-assocd. RPG regulation. Abf1 and Fhl1 promoter assocn. was confirmed by ChIP and/or gel retardation assays. Mutational anal. revealed a more severe requirement of Abf1 than Fhl1 binding sites for RPG transcription. In the case of RPS22B an unusual Tbf1 binding site promoted both RPS22B and intron-hosted SNR44 expression. Abf1-RPG down-regulation upon TOR pathway inhibition was much attenuated at defective mutant promoters unable to bind Abf1. TORC1 inactivation caused the expected redn. of Ifh1 occupancy at RPS22B and RPL3 promoters, but unexpectedly it entailed largely increased Abf1 assocn. with Abf1-RPG promoters. We present evidence that Abf1 recruitment upon nutritional stress, also obsd. for representative ribosome biogenesis genes, favors RPG transcriptional rescue upon nutrient replenishment, thus pointing to nutrient-regulated Abf1 dynamics at promoters as a novel mechanism in ribosome biogenesis control.
- 44Imran, A.; Moyer, B. S.; Canning, A. J.; Kalina, D.; Duncan, T. M.; Moody, K. J.; Wolfe, A. J.; Cosgrove, M. S.; Movileanu, L. Kinetics of the multitasking high-affinity Win binding site of WDR5 in restricted and unrestricted conditions. Biochem. J. 2021, 478, 2145– 2161, DOI: 10.1042/bcj2021025344Kinetics of the multitasking high-affinity Win binding site of WDR5 in restricted and unrestricted conditionsImran, Ali; Moyer, Brandon S.; Canning, Ashley J.; Kalina, Dan; Duncan, Thomas M.; Moody, Kelsey J.; Wolfe, Aaron J.; Cosgrove, Michael S.; Movileanu, LiviuBiochemical Journal (2021), 478 (11), 2145-2161CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)Recent advances in quant. proteomics show that WD40 proteins play a pivotal role in numerous cellular networks. Yet, they have been fairly unexplored and their phys. assocns. with other proteins are ambiguous. A quant. understanding of these interactions has wide-ranging significance. WD40 repeat protein 5 (WDR5) interacts with all members of human SET1/MLL methyltransferases, which regulate methylation of the histone 3 lysine 4 (H3K4). Here, using real-time binding measurements in a high-throughput setting, we identified the kinetic fingerprint of transient assocns. between WDR5 and 14-residue WDR5 interaction (Win) motif peptides of each SET1 protein (SET1Win). Our results reveal that the high-affinity WDR5-SET1Win interactions feature slow assocn. kinetics. This finding is likely due to the requirement of SET1Win to insert into the narrow WDR5 cavity, also named the Win binding site. Furthermore, our explorations indicate fairly slow dissocn. kinetics. This conclusion is in accordance with the primary role of WDR5 in maintaining the functional integrity of a large multisubunit complex, which regulates the histone methylation. Because the Win binding site is considered a key therapeutic target, the immediate outcomes of this study could form the basis for accelerated developments in medical biotechnol.
- 45Mayse, L. A.; Imran, A.; Larimi, M. G.; Cosgrove, M. S.; Wolfe, A. J.; Movileanu, L. Disentangling the recognition complexity of a protein hub using a nanopore. Nat. Commun. 2022, 13, 978, DOI: 10.1038/s41467-022-28465-845Disentangling the recognition complexity of a protein hub using a nanoporeMayse, Lauren Ashley; Imran, Ali; Larimi, Motahareh Ghahari; Cosgrove, Michael S.; Wolfe, Aaron James; Movileanu, LiviuNature Communications (2022), 13 (1), 978CODEN: NCAOBW; ISSN:2041-1723. (Nature Portfolio)WD40 repeat proteins are frequently involved in processing cell signaling and scaffolding large multi-subunit machineries. Despite their significance in physiol. and disease-like conditions, their reversible interactions with other proteins remain modestly examd. Here, we show the development and validation of a protein nanopore for the detection and quantification of WD40 repeat protein 5 (WDR5), a chromatin-assocd. hub involved in epigenetic regulation of histone methylation. Our nanopore sensor is equipped with a 14-residue Win motif of mixed lineage leukemia 4 methyltransferase (MLL4Win), a WDR5 ligand. Our approach reveals a broad dynamic range of MLL4Win-WDR5 interactions and three distant subpopulations of binding events, representing three modes of protein recognition. The three binding events are confirmed as specific interactions using a weakly binding WDR5 deriv. and various environmental contexts. These outcomes demonstrate the substantial sensitivity of our nanopore sensor, which can be utilized in protein analytics.
- 46Karatas, H.; Townsend, E. C.; Cao, F.; Chen, Y.; Bernard, D.; Liu, L.; Lei, M.; Dou, Y.; Wang, S. High-affinity, small-molecule peptidomimetic inhibitors of MLL1/WDR5 protein-protein interaction. J. Am. Chem. Soc. 2013, 135, 669– 682, DOI: 10.1021/ja306028q46High-Affinity, Small-Molecule Peptidomimetic Inhibitors of MLL1/WDR5 Protein-Protein InteractionKaratas, Hacer; Townsend, Elizabeth C.; Cao, Fang; Chen, Yong; Bernard, Denzil; Liu, Liu; Lei, Ming; Dou, Yali; Wang, ShaomengJournal of the American Chemical Society (2013), 135 (2), 669-682CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Mixed lineage leukemia 1 (MLL1) is a histone H3 lysine 4 (H3K4) methyltransferase, and targeting the MLL1 enzymic activity has been proposed as a novel therapeutic strategy for the treatment of acute leukemia harboring MLL1 fusion proteins. The MLL1/WDR5 protein-protein interaction is essential for MLL1 enzymic activity. In the present study, we designed a large no. of peptidomimetics to target the MLL1/WDR5 interaction based upon -CO-ARA-NH-, the min. binding motif derived from MLL1. Our study led to the design of high-affinity peptidomimetics, which bind to WDR5 with Ki < 1 nM and function as potent antagonists of MLL1 activity in a fully reconstituted in vitro H3K4 methyltransferase assay. Detn. of co-crystal structures of two potent peptidomimetics in complex with WDR5 establishes their structural basis for high-affinity binding to WDR5. Evaluation of one such peptidomimetic, MM-102, in bone marrow cells transduced with MLL1-AF9 fusion construct shows that the compd. effectively decreases the expression of HoxA9 and Meis-1, two crit. MLL1 target genes in MLL1 fusion protein mediated leukemogenesis. MM-102 also specifically inhibits cell growth and induces apoptosis in leukemia cells harboring MLL1 fusion proteins. Our study provides the first proof-of-concept for the design of small-mol. inhibitors of the WDR5/MLL1 protein-protein interaction as a novel therapeutic approach for acute leukemia harboring MLL1 fusion proteins.
- 47Cao, F.; Townsend, E. C.; Karatas, H.; Xu, J.; Li, L.; Lee, S.; Liu, L.; Chen, Y.; Ouillette, P.; Zhu, J.; Hess, J. L.; Atadja, P.; Lei, M.; Qin, Z. S.; Malek, S.; Wang, S.; Dou, Y. Targeting MLL1 H3K4 methyltransferase activity in mixed-lineage leukemia. Mol. Cell 2014, 53, 247– 261, DOI: 10.1016/j.molcel.2013.12.00147Targeting MLL1 H3K4 Methyltransferase Activity in Mixed-Lineage LeukemiaCao, Fang; Townsend, Elizabeth C.; Karatas, Hacer; Xu, Jing; Li, Li; Lee, Shirley; Liu, Liu; Chen, Yong; Ouillette, Peter; Zhu, Jidong; Hess, Jay L.; Atadja, Peter; Lei, Ming; Qin, Zhaohui S.; Malek, Sami; Wang, Shaomeng; Dou, YaliMolecular Cell (2014), 53 (2), 247-261CODEN: MOCEFL; ISSN:1097-2765. (Elsevier Inc.)Here we report a comprehensive characterization of our recently developed inhibitor MM-401 that targets the MLL1 H3K4 methyltransferase activity. MM-401 is able to specifically inhibit MLL1 activity by blocking MLL1-WDR5 interaction and thus the complex assembly. This targeting strategy does not affect other mixed-lineage leukemia (MLL) family histone methyltransferases (HMTs), revealing a unique regulatory feature for the MLL1 complex. Using MM-401 and its enantiomer control MM-NC-401, we show that inhibiting MLL1 methyltransferase activity specifically blocks proliferation of MLL cells by inducing cell-cycle arrest, apoptosis, and myeloid differentiation without general toxicity to normal bone marrow cells or non-MLL cells. More importantly, transcriptome analyses show that MM-401 induces changes in gene expression similar to those of MLL1 deletion, supporting a predominant role of MLL1 activity in regulating MLL1-dependent leukemia transcription program. We envision broad applications for MM-401 in basic and translational research.
- 48Alicea-Velázquez, N. L.; Shinsky, S. A.; Loh, D. M.; Lee, J. H.; Skalnik, D. G.; Cosgrove, M. S. Targeted Disruption of the Interaction between WD-40 Repeat Protein 5 (WDR5) and Mixed Lineage Leukemia (MLL)/SET1 Family Proteins Specifically Inhibits MLL1 and SETd1A Methyltransferase Complexes. J. Biol. Chem. 2016, 291, 22357– 22372, DOI: 10.1074/jbc.M116.75262648Targeted Disruption of the Interaction between WD-40 Repeat Protein 5 (WDR5) and Mixed Lineage Leukemia (MLL)/SET1 Family Proteins Specifically Inhibits MLL1 and SETd1A Methyltransferase ComplexesAlicea-Velazquez, Nilda L.; Shinsky, Stephen A.; Loh, Daniel M.; Lee, Jeong-Heon; Skalnik, David G.; Cosgrove, Michael S.Journal of Biological Chemistry (2016), 291 (43), 22357-22372CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)MLL1 belongs to the SET1 family of histone H3 lysine 4 (H3K4) methyltransferases, composed of MLL1-4 and SETd1A/B. MLL1 translocations are present in acute leukemias, and mutations in several family members are assocd. with cancer and developmental disorders. MLL1 assocs. with a subcomplex contg. WDR5, RbBP5, ASH2L, and DPY-30 (WRAD), forming the MLL1 core complex required for H3K4 mono- and dimethylation and transcriptional activation. Core complex assembly requires interaction of WDR5 with the MLL1 Win (WDR5 interaction) motif, which is conserved across the SET1 family. Agents that mimic the SET1 family Win motif inhibit the MLL1 core complex and have become an attractive approach for targeting MLL1 in cancers. Like MLL1, other SET1 family members interact with WRAD, but the roles of the Win motif in complex assembly and enzymic activity remain unexplored. Here, we show that the Win motif is necessary for interaction of WDR5 with all members of the human SET1 family. Mutation of the Win motif-WDR5 interface severely disrupts assembly and activity of MLL1 and SETd1A complexes but only modestly disrupts MLL2/4 and SETd1B complexes without significantly altering enzymic activity in vitro. Notably, in the absence of WDR5, MLL3 interacts with RAD and shows enhanced activity. To further probe the role of the Win motif-WDR5 interaction, we designed a peptidomimetic that binds WDR5 (Kd ∼3 nm) and selectively inhibits activity of MLL1 and SETd1A core complexes within the SET1 family. Our results reveal that SET1 family complexes with the weakest Win motif-WDR5 interaction are more susceptible to Win motif-based inhibitors.
- 49Karatas, H.; Li, Y.; Liu, L.; Ji, J.; Lee, S.; Chen, Y.; Yang, J.; Huang, L.; Bernard, D.; Xu, J.; Townsend, E. C.; Cao, F.; Ran, X.; Li, X.; Wen, B.; Sun, D.; Stuckey, J. A.; Lei, M.; Dou, Y.; Wang, S. Discovery of a Highly Potent, Cell-Permeable Macrocyclic Peptidomimetic (MM-589) Targeting the WD Repeat Domain 5 Protein (WDR5)-Mixed Lineage Leukemia (MLL) Protein-Protein Interaction. J. Med. Chem. 2017, 60, 4818– 4839, DOI: 10.1021/acs.jmedchem.6b0179649Discovery of a Highly Potent, Cell-Permeable Macrocyclic Peptidomimetic (MM-589) Targeting the WD Repeat Domain 5 Protein (WDR5)-Mixed Lineage Leukemia (MLL) Protein-Protein InteractionKaratas, Hacer; Li, Yangbing; Liu, Liu; Ji, Jiao; Lee, Shirley; Chen, Yong; Yang, Jiuling; Huang, Liyue; Bernard, Denzil; Xu, Jing; Townsend, Elizabeth C.; Cao, Fang; Ran, Xu; Li, Xiaoqin; Wen, Bo; Sun, Duxin; Stuckey, Jeanne A.; Lei, Ming; Dou, Yali; Wang, ShaomengJournal of Medicinal Chemistry (2017), 60 (12), 4818-4839CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)We report herein the design, synthesis and evaluation of macrocyclic peptidomimetics that bind to WDR5 and block the WDR5-MLL protein-protein interaction. Compd. (I) (R1 = Me; n = 1) (MM-589) binds to WDR5 with an IC50 value of 0.90 nM (Ki value <1 nM), and inhibits the MLL H3K4 methyltransferase (HMT) activity with an IC50 value of 12.7 nM. Compd. I (R1 = Me; n = 1) potently and selectively inhibits cell growth in human leukemia cell lines harboring MLL translocations, and is >40 times better than the previously reported compd. MM-401. Co-crystal structures of I (R1 = H, n = 5) and (R1 = Me; n = 1) complexed with WDR5 provide structural basis for their high affinity binding to WDR5. Addnl., we have developed and optimized a new, AlphaLISA-based MLL HMT functional assay to facilitate the functional evaluation of these designed compds. Compd. I (R1 = Me; n = 1) represents the most potent inhibitor of the WDR5-MLL interaction reported to date and further optimization of I (R1 = Me; n = 1) may yield a new therapy for acute leukemia.
- 50Schapira, M.; Tyers, M.; Torrent, M.; Arrowsmith, C. H. WD40 repeat domain proteins: a novel target class?. Nat. Rev. Drug Discovery 2017, 16, 773– 786, DOI: 10.1038/nrd.2017.17950WD40 repeat domain proteins: a novel target class?Schapira, Matthieu; Tyers, Mike; Torrent, Maricel; Arrowsmith, Cheryl H.Nature Reviews Drug Discovery (2017), 16 (11), 773-786CODEN: NRDDAG; ISSN:1474-1776. (Nature Research)Antagonism of protein-protein interactions (PPIs) with small mols. is becoming more feasible as a therapeutic approach. Successful PPI inhibitors tend to target proteins contg. deep peptide-binding grooves or pockets rather than the more common large, flat protein interaction surfaces. Here, we review one of the most abundant PPI domains in the human proteome, the WD40 repeat (WDR) domain, which has a central peptide-binding pocket and is a member of the β-propeller domain-contg. protein family. Recently, two WDR domain-contg. proteins, WDR5 and EED, as well as other β-propeller domains have been successfully targeted by potent, specific, cell-active, drug-like chem. probes. Could WDR domains be a novel target class for drug discovery. Although the research is at an early stage and therefore not clin. validated, cautious optimism is justified, as WDR domain-contg. proteins are involved in multiple disease-assocd. pathways. The druggability and structural diversity of WDR domain binding pockets suggest that understanding how to target this prevalent domain class will open up areas of disease biol. that have so far resisted drug discovery efforts.
- 51Aho, E. R.; Weissmiller, A. M.; Fesik, S. W.; Tansey, W. P. Targeting WDR5: A WINning Anti-Cancer Strategy?. Epigenet. Insights 2019, 12, 2516865719865282, DOI: 10.1177/2516865719865282There is no corresponding record for this reference.
- 52Aho, E. R.; Wang, J.; Gogliotti, R. D.; Howard, G. C.; Phan, J.; Acharya, P.; Macdonald, J. D.; Cheng, K.; Lorey, S. L.; Lu, B.; Wenzel, S.; Foshage, A. M.; Alvarado, J.; Wang, F.; Shaw, J. G.; Zhao, B.; Weissmiller, A. M.; Thomas, L. R.; Vakoc, C. R.; Hall, M. D.; Hiebert, S. W.; Liu, Q.; Stauffer, S. R.; Fesik, S. W.; Tansey, W. P. Displacement of WDR5 from Chromatin by a WIN Site Inhibitor with Picomolar Affinity. Cell Rep. 2019, 26, 2916– 2928.e13, DOI: 10.1016/j.celrep.2019.02.04752Displacement of WDR5 from Chromatin by a WIN Site Inhibitor with Picomolar AffinityAho, Erin R.; Wang, Jing; Gogliotti, Rocco D.; Howard, Gregory C.; Phan, Jason; Acharya, Pankaj; Macdonald, Jonathan D.; Cheng, Ken; Lorey, Shelly L.; Lu, Bin; Wenzel, Sabine; Foshage, Audra M.; Alvarado, Joseph; Wang, Feng; Shaw, J. Grace; Zhao, Bin; Weissmiller, April M.; Thomas, Lance R.; Vakoc, Christopher R.; Hall, Matthew D.; Hiebert, Scott W.; Liu, Qi; Stauffer, Shaun R.; Fesik, Stephen W.; Tansey, William P.Cell Reports (2019), 26 (11), 2916-2928.e13CODEN: CREED8; ISSN:2211-1247. (Cell Press)The chromatin-assocd. protein WDR5 is a promising target for pharmacol. inhibition in cancer. Drug discovery efforts center on the blockade of the "WIN site" of WDR5, a well-defined pocket that is amenable to small mol. inhibition. Various cancer contexts have been proposed to be targets for WIN site inhibitors, but a lack of understanding of WDR5 target genes and of the primary effects of WIN site inhibitors hampers their utility. Here, by the discovery of potent WIN site inhibitors, we demonstrate that the WIN site links WDR5 to chromatin at a small cohort of loci, including a specific subset of ribosome protein genes. WIN site inhibitors rapidly displace WDR5 from chromatin and decrease the expression of assocd. genes, causing translational inhibition, nucleolar stress, and p53 induction. Our studies define a mode by which WDR5 engages chromatin and forecast that WIN site blockade could have utility against multiple cancer types.
- 53Dennis, M. L.; Morrow, B. J.; Dolezal, O.; Cuzzupe, A. N.; Stupple, A. E.; Newman, J.; Bentley, J.; Hattarki, M.; Nuttall, S. D.; Foitzik, R. C.; Street, I. P.; Stupple, P. A.; Monahan, B. J.; Peat, T. S. Fragment screening for a protein-protein interaction inhibitor to WDR5. Struct. Dyn. 2019, 6, 064701, DOI: 10.1063/1.512284953Fragment screening for a protein-protein interaction inhibitor to WDR5Dennis, Matthew L.; Morrow, Benjamin J.; Dolezal, Olan; Cuzzupe, Anthony N.; Stupple, Alexandra E.; Newman, Janet; Bentley, John; Hattarki, Meghan; Nuttall, Stewart D.; Foitzik, Richard C.; Street, Ian P.; Stupple, Paul A.; Monahan, Brendon J.; Peat, Thomas. S.Structural Dynamics (2019), 6 (6), 064701/1-064701/20CODEN: SDTYAE; ISSN:2329-7778. (American Institute of Physics)In this study, a fragment screen with Surface Plasmon Resonance (SPR) was used to identify a highly ligand-efficient imidazole-contg. compd. that is bound in the WIN site. The subsequent medicinal chem. campaign-guided by a suite of high-resoln. cocrystal structures with WDR5-progressed the initial hit to a low micromolar binder. One outcome from this study is a moiety that substitutes well for the side chain of arginine; a tripeptide contg. one such substitution was resolved in a high resoln. structure (1.5 Å) with a binding mode analogous to the native tripeptide. SPR furthermore indicates a similar residence time (kd = ∼0.06 s-1) for these two analogs. This novel scaffold therefore represents a possible means to overcome the potential permeability issues of WDR5 ligands that possess highly basic groups like guanidine. The series reported here furthers the understanding of the WDR5 WIN site and functions as a starting point for the development of more potent WDR5 inhibitors that may serve as cancer therapeutics. (c) 2019 American Institute of Physics.
- 54Bolshan, Y.; Getlik, M.; Kuznetsova, E.; Wasney, G. A.; Hajian, T.; Poda, G.; Nguyen, K. T.; Wu, H.; Dombrovski, L.; Dong, A.; Senisterra, G.; Schapira, M.; Arrowsmith, C. H.; Brown, P. J.; Al-Awar, R.; Vedadi, M.; Smil, D. Synthesis, Optimization, and Evaluation of Novel Small Molecules as Antagonists of WDR5-MLL Interaction. ACS Med. Chem. Lett. 2013, 4, 353– 357, DOI: 10.1021/ml300467n54Synthesis, Optimization, and Evaluation of Novel Small Molecules as Antagonists of WDR5-MLL InteractionBolshan, Yuri; Getlik, Matthaus; Kuznetsova, Ekaterina; Wasney, Gregory A.; Hajian, Taraneh; Poda, Gennadiy; Nguyen, Kong T.; Wu, Hong; Dombrovski, Ludmila; Dong, Aiping; Senisterra, Guillermo; Schapira, Matthieu; Arrowsmith, Cheryl H.; Brown, Peter J.; Al-awar, Rima; Vedadi, Masoud; Smil, DavidACS Medicinal Chemistry Letters (2013), 4 (3), 353-357CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)The WD40-repeat protein WDR5 plays a crit. role in maintaining the integrity of MLL complexes and fully activating their methyltransferase function. MLL complexes, the trithorax-like family of SET1 methyltransferases, catalyze trimethylation of lysine 4 on histone 3, and they have been widely implicated in various cancers. Antagonism of WDR5 and MLL subunit interaction by small mols. has recently been presented as a practical way to inhibit activity of the MLL1 complex, and N-(2-(4-methylpiperazin-1-yl)-5-substituted-phenyl) benzamides were reported as potent and selective antagonists of such an interaction. Here, we describe the protein crystal structure guided optimization of prototypic compd. I (Kdis = 7 μM), leading to identification of more potent antagonist II (Kdis = 0.3 μM).
- 55Getlik, M.; Smil, D.; Zepeda-Velázquez, C.; Bolshan, Y.; Poda, G.; Wu, H.; Dong, A.; Kuznetsova, E.; Marcellus, R.; Senisterra, G.; Dombrovski, L.; Hajian, T.; Kiyota, T.; Schapira, M.; Arrowsmith, C. H.; Brown, P. J.; Vedadi, M.; Al-Awar, R. Structure-Based Optimization of a Small Molecule Antagonist of the Interaction Between WD Repeat-Containing Protein 5 (WDR5) and Mixed-Lineage Leukemia 1 (MLL1). J. Med. Chem. 2016, 59, 2478– 2496, DOI: 10.1021/acs.jmedchem.5b0163055Structure-Based Optimization of a Small Molecule Antagonist of the Interaction Between WD Repeat-Containing Protein 5 (WDR5) and Mixed-Lineage Leukemia 1 (MLL1)Getlik, Matthaus; Smil, David; Zepeda-Velazquez, Carlos; Bolshan, Yuri; Poda, Gennady; Wu, Hong; Dong, Aiping; Kuznetsova, Ekaterina; Marcellus, Richard; Senisterra, Guillermo; Dombrovski, Ludmila; Hajian, Taraneh; Kiyota, Taira; Schapira, Matthieu; Arrowsmith, Cheryl H.; Brown, Peter J.; Vedadi, Masoud; Al-awar, RimaJournal of Medicinal Chemistry (2016), 59 (6), 2478-2496CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)WD repeat-contg. protein 5 (WDR5) is an important component of the multiprotein complex essential for activating mixed-lineage leukemia 1 (MLL1). Rearrangement of the MLL1 gene is assocd. with onset and progression of acute myeloid and lymphoblastic leukemias, and targeting the WDR5-MLL1 interaction may result in new cancer therapeutics. Our previous work showed that binding of small mol. ligands to WDR5 can modulate its interaction with MLL1, suppressing MLL1 methyltransferase activity. Initial structure-activity relationship studies identified N-(2-(4-methylpiperazin-1-yl)-5-substituted-phenyl) benzamides as potent and selective antagonists of this protein-protein interaction. Guided by crystal structure data and supported by in silico library design, we optimized the scaffold by varying the C-1 benzamide and C-5 substituents. This allowed us to develop the first highly potent (Kdisp < 100 nM) small mol. antagonists of the WDR5-MLL1 interaction and demonstrate that N-(4-(4-methylpiperazin-1-yl)-3'-(morpholinomethyl)-[1,1'-biphenyl]-3-yl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyridine-3-carboxamide 16d (OICR-9429) is a potent and selective chem. probe suitable to help dissect the biol. role of WDR5.
- 56Schapira, M.; Arrowsmith, C. H. Methyltransferase inhibitors for modulation of the epigenome and beyond. Curr. Opin. Chem. Biol. 2016, 33, 81– 87, DOI: 10.1016/j.cbpa.2016.05.03056Methyltransferase inhibitors for modulation of the epigenome and beyondSchapira, Matthieu; Arrowsmith, Cheryl H.Current Opinion in Chemical Biology (2016), 33 (), 81-87CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)Over the past two years tremendous progress has been made in the discovery of new inhibitors of protein lysine and arginine methyltransferases, establishing this class of epigenetic enzymes, along with DNA methyltransferases, as druggable protein families. New inhibitors of protein methyltransferases have been described with a variety of mechanisms of action including cofactor competitive, substrate competitive, allosteric inhibitors and disruptors of protein-protein interactions. Inhibitors have been used extensively in oncol. studies, and inhibitors of EZH2, and DOT1L are currently in clin. trials. Finally, advances in understanding the clin. mechanism of action of 5-azacytidine and related DNA hypomethylation agents were reported, revealing a likely role for a cell autonomous innate immune response.
- 57Gupta, A.; Xu, J.; Lee, S.; Tsai, S. T.; Zhou, B.; Kurosawa, K.; Werner, M. S.; Koide, A.; Ruthenburg, A. J.; Dou, Y.; Koide, S. Facile target validation in an animal model with intracellularly expressed monobodies. Nat. Chem. Biol. 2018, 14, 895– 900, DOI: 10.1038/s41589-018-0099-z57Facile target validation in an animal model with intracellularly expressed monobodiesGupta, Ankit; Xu, Jing; Lee, Shirley; Tsai, Steven T.; Zhou, Bo; Kurosawa, Kohei; Werner, Michael S.; Koide, Akiko; Ruthenburg, Alexander J.; Dou, Yali; Koide, ShoheiNature Chemical Biology (2018), 14 (9), 895-900CODEN: NCBABT; ISSN:1552-4450. (Nature Research)Rapidly detg. the biol. effect of perturbing a site within a potential drug target could guide drug discovery efforts, but it remains challenging. Here, the authors describe a facile target validation approach that exploits monobodies, small synthetic binding proteins that can be fully functionally expressed in cells. The authors developed a potent and selective monobody to WDR5, a core component of the mixed lineage leukemia (MLL) methyltransferase complex. The monobody bound to the MLL interaction site of WDR5, the same binding site for small-mol. inhibitors whose efficacy has been demonstrated in cells but not in animals. As a genetically encoded reagent, the monobody inhibited proliferation of an MLL-AF9 cell line in vitro, suppressed its leukemogenesis and conferred a survival benefit in an in vivo mouse leukemia model. The capacity of this approach to readily bridge biochem., structural, cellular characterization and tests in animal models may accelerate discovery and validation of druggable sites.
- 58Concepcion, J.; Witte, K.; Wartchow, C.; Choo, S.; Yao, D.; Persson, H.; Wei, J.; Li, P.; Heidecker, B.; Ma, W.; Varma, R.; Zhao, L.-S.; Perillat, D.; Carricato, G.; Recknor, M.; Du, K.; Ho, H.; Ellis, T.; Gamez, J.; Howes, M.; Phi-Wilson, J.; Lockard, S.; Zuk, R.; Tan, H. Label-free detection of biomolecular interactions using BioLayer interferometry for kinetic characterization. Comb. Chem. High Throughput Screen. 2009, 12, 791– 800, DOI: 10.2174/13862070978910491558Label-free detection of biomolecular interactions using biolayer interferometry for kinetic characterizationConcepcion, Joy; Witte, Krista; Wartchow, Charles; Choo, Sae; Yao, Danfeng; Persson, Henrik; Wei, Jing; Li, Pu; Heidecker, Bettina; Ma, Weilei; Varma, Ram; Zhao, Lian-She; Perillat, Donald; Carricato, Greg; Recknor, Michael; Du, Kevin; Ho, Huddee; Ellis, Tim; Gamez, Juan; Howes, Michael; Phi-Wilson, Janette; Lockard, Scott; Zuk, Robert; Tan, HongCombinatorial Chemistry & High Throughput Screening (2009), 12 (8), 791-800CODEN: CCHSFU; ISSN:1386-2073. (Bentham Science Publishers Ltd.)A review. The anal. of biomol. interactions is key in the drug development process. Label-free biosensor methods provide information on binding, kinetics, concn., and the affinity of an interaction. These techniques provide real-time monitoring of interactions between an immobilized ligand (such as a receptor) to an analyte in soln. without the use of labels. Advances in biosensor design and detection using BioLayer Interferometry (BLI) provide a simple platform that enables label-free monitoring of biomol. interactions without the use of flow cells. We review the applications of BLI in a wide variety of research and development environments for quantifying antibodies and proteins and measuring kinetics parameters.
- 59Weeramange, C. J.; Fairlamb, M. S.; Singh, D.; Fenton, A. W.; Swint-Kruse, L. The strengths and limitations of using biolayer interferometry to monitor equilibrium titrations of biomolecules. Protein Sci 2020, 29, 1018– 1034, DOI: 10.1002/pro.382759The strengths and limitations of using biolayer interferometry to monitor equilibrium titrations of biomoleculesWeeramange, Chamitha J.; Fairlamb, Max S.; Singh, Dipika; Fenton, Aron W.; Swint-Kruse, LiskinProtein Science (2020), 29 (4), 1018-1034CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)Every method used to quantify biomol. interactions has its own strengths and limitations. To quantify protein-DNA binding affinities, nitrocellulose filter binding assays with 32P-labeled DNA quantify Kd values from 10-12 to 10-8 M but have several tech. limitations. Here, we considered the suitability of biolayer interferometry (BLI), which monitors assocn. and dissocn. of a sol. macromol. to an immobilized species; the ratio koff/kon dets. Kd. However, for lactose repressor protein (LacI) and an engineered repressor protein ("LLhF") binding immobilized DNA, complicated kinetic curves precluded this anal. Thus, we detd. whether the amplitude of the BLI signal at equil. related linearly to the fraction of protein bound to DNA. A key question was the effective concn. of immobilized DNA. Equil. titrn. expts. with DNA concns. below Kd (equil. binding regime) must be analyzed differently than those with DNA near or above Kd (stoichiometric binding regime). For ForteBio streptavidin tips, the most frequent effective DNA concn. was ∼2 x 10-9 M. Although variation occurred among different lots of sensor tips, binding events with Kd ≥ 10-8 M should reliably be in the equil. binding regime. We also obsd. effects from multi-valent interactions: Tetrameric LacI bound two immobilized DNAs whereas dimeric LLhF did not. We next used BLI to quantify the amt. of inducer sugars required to allosterically diminish protein-DNA binding and to assess the affinity of fructose-1-kinase for the DNA-LLhF complex. Overall, when exptl. design corresponded with appropriate data interpretation, BLI was convenient and reliable for monitoring equil. titrns. and thereby quantifying a variety of binding interactions.
- 60Imran, A.; Moyer, B. S.; Wolfe, A. J.; Cosgrove, M. S.; Makarov, D. E.; Movileanu, L. Interplay of Affinity and Surface Tethering in Protein Recognition. J. Phys. Chem. Lett. 2022, 13, 4021– 4028, DOI: 10.1021/acs.jpclett.2c0062160Interplay of Affinity and Surface Tethering in Protein RecognitionImran, Ali; Moyer, Brandon S.; Wolfe, Aaron J.; Cosgrove, Michael S.; Makarov, Dmitrii E.; Movileanu, LiviuJournal of Physical Chemistry Letters (2022), 13 (18), 4021-4028CODEN: JPCLCD; ISSN:1948-7185. (American Chemical Society)Surface-tethered ligand-receptor complexes are key components in biol. signaling and adhesion. They also find increasing utility in single-mol. assays and biotechnol. applications. Here, we study the real-time binding kinetics between various surface-immobilized peptide ligands and their unrestrained receptors. A long peptide tether increases the assocn. of ligand-receptor complexes, exptl. proving the fly casting mechanism where the disorder accelerates protein recognition. On the other hand, a short peptide tether enhances the complex dissocn. Notably, the rate consts. measured for the same receptor, but under different spatial constraints, are strongly correlated to one another. Furthermore, this correlation can be used to predict how surface tethering on a ligand-receptor complex alters its binding kinetics. Our results have immediate implications in the broad areas of biomol. recognition, intrinsically disordered proteins, and biosensor technol.
- 61Wolfe, A. J.; Si, W.; Zhang, Z.; Blanden, A. R.; Hsueh, Y.-C.; Gugel, J. F.; Pham, B.; Chen, M.; Loh, S. N.; Rozovsky, S.; Aksimentiev, A.; Movileanu, L. Quantification of membrane protein-detergent complex interactions. J. Phys. Chem. B 2017, 121, 10228– 10241, DOI: 10.1021/acs.jpcb.7b0804561Quantification of Membrane Protein-Detergent Complex InteractionsWolfe, Aaron J.; Si, Wei; Zhang, Zhengqi; Blanden, Adam R.; Hsueh, Yi-Ching; Gugel, Jack F.; Pham, Bach; Chen, Min; Loh, Stewart N.; Rozovsky, Sharon; Aksimentiev, Aleksei; Movileanu, LiviuJournal of Physical Chemistry B (2017), 121 (44), 10228-10241CODEN: JPCBFK; ISSN:1520-5207. (American Chemical Society)Although fundamentally significant in structural, chem., and membrane biol., the interfacial protein-detergent complex (PDC) interactions have been modestly examd. because of the complicated behavior of both detergents and membrane proteins in aq. phase. Membrane proteins are prone to unproductive aggregation resulting from poor detergent solvation, but the participating forces in this phenomenon remain ambiguous. Here, using rational membrane protein design, targeted chem. modification, and steady-state fluorescence polarization spectroscopy, the detergent desolvation of membrane proteins can be quant. evaluated. Depleting the detergent in the sample well produced a two-state transition of membrane proteins between a fully detergent-solvated state and a detergent-desolvated state, the nature of which depended on the interfacial PDC interactions. Using a panel of six membrane proteins of varying hydrophobic topog., structural fingerprint, and charge distribution on the solvent-accessible surface, the authors provide direct exptl. evidence for the contributions of the electrostatic and hydrophobic interactions to the protein solvation properties. Moreover, all-atom mol. dynamics simulations report the major contribution of the hydrophobic forces exerted at the PDC interface. This semiquant. approach might be extended in the future to include studies of the interfacial PDC interactions of other challenging membrane protein systems of unknown structure. This would have practical importance in protein extn., solubilization, stabilization, and crystn.
- 62Wolfe, A. J.; Gugel, J. F.; Chen, M.; Movileanu, L. Kinetics of Membrane Protein-Detergent Interactions Depend on Protein Electrostatics. J. Phys. Chem. B 2018, 122, 9471– 9481, DOI: 10.1021/acs.jpcb.8b0788962Kinetics of Membrane Protein-Detergent Interactions Depend on Protein ElectrostaticsWolfe, Aaron J.; Gugel, Jack F.; Chen, Min; Movileanu, LiviuJournal of Physical Chemistry B (2018), 122 (41), 9471-9481CODEN: JPCBFK; ISSN:1520-5207. (American Chemical Society)Interactions of a membrane protein with a detergent micelle represent a fundamental process with practical implications in structural and chem. biol. Quant. assessment of the kinetics of protein-detergent complex (PDC) interactions has always been challenged by complicated behavior of both membrane proteins and solubilizing detergents in aq. phase. Here, we show the kinetic reads of the desorption of maltoside-contg. detergents from β-barrel membrane proteins. Using steady-state fluorescence polarization (FP) anisotropy measurements, we recorded real-time, specific signatures of the PDC interactions. The results of these measurements were used to infer the model-dependent rate consts. of assocn. and dissocn. of the proteomicelles. Remarkably, the kinetics of the PDC interactions depend on the overall protein charge despite the nonionic nature of the detergent monomers. In the future, this approach might be employed for high-throughput screening of kinetic fingerprints of different membrane proteins stabilized in micelles that contain mixts. of various detergents.
- 63Forbes, S. A.; Beare, D.; Boutselakis, H.; Bamford, S.; Bindal, N.; Tate, J.; Cole, C. G.; Ward, S.; Dawson, E.; Ponting, L.; Stefancsik, R.; Harsha, B.; Kok, C. Y.; Jia, M.; Jubb, H.; Sondka, Z.; Thompson, S.; De, T.; Campbell, P. J. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res. 2017, 45, D777– d783, DOI: 10.1093/nar/gkw112163COSMIC: somatic cancer genetics at high-resolutionForbes, Simon A.; Beare, David; Boutselakis, Harry; Bamford, Sally; Bindal, Nidhi; Tate, John; Cole, Charlotte G.; Ward, Sari; Dawson, Elisabeth; Ponting, Laura; Stefancsik, Raymund; Harsha, Bhavana; Kok, Chai Yin; Jia, Mingming; Jubb, Harry; Sondka, Zbyslaw; Thompson, Sam; De, Tisham; Campbell, Peter J.Nucleic Acids Research (2017), 45 (D1), D777-D783CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)COSMIC, the Catalog of Somatic Mutations in Cancer (http://cancer.sanger.ac.uk) is a high-resoln. resource for exploring targets and trends in the genetics of human cancer. Currently the broadest database of mutations in cancer, the information in COSMIC is curated by expert scientists, primarily by scrutinizing large nos. of scientific publications. Over 4 million coding mutations are described in v78 (Sept. 2016), combining genome-wide sequencing results from 28 366 tumors with complete manual curation of 23 489 individual publications focused on 186 key genes and 286 key fusion pairs across all cancers. Mol. profiling of large tumor nos. has also allowed the annotation of more than 13 million non-coding mutations, 18 029 gene fusions, 187 429 genome rearrangements, 1 271 436 abnormal copy no. segments, 9 175 462 abnormal expression variants and 7 879 142 differentially methylated CpG dinucleotides. COSMIC now details the genetics of drug resistance, novel somatic gene mutations which allow a tumor to evade therapeutic cancer drugs. Focusing initially on highly characterized drugs and genes, COSMIC v78 contains wide resistance mutation profiles across 20 drugs, detailing the recurrence of 301 unique resistance alleles across 1934 drug-resistant tumors. All information from the COSMIC database is available freely on the COSMIC website.
- 64Kumar, S.; Nussinov, R. Relationship between ion pair geometries and electrostatic strengths in proteins. Biophys. J. 2002, 83, 1595– 1612, DOI: 10.1016/s0006-3495(02)73929-564Relationship between ion pair geometries and electrostatic strengths in proteinsKumar, Sandeep; Nussinov, RuthBiophysical Journal (2002), 83 (3), 1595-1612CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)The electrostatic free energy contribution of an ion pair in a protein depends on two factors, geometrical orientation of the side-chain charged groups with respect to each other and the structural context of the ion pair in the protein. Conformers in NMR ensembles enable studies of the relationship between geometry and electrostatic strengths of ion pairs, because the protein structural contexts are highly similar across different conformers. We have studied this relationship using a dataset of 22 unique ion pairs in 14 NMR conformer ensembles for 11 nonhomologous proteins. In different NMR conformers, the ion pairs are classified as salt bridges, nitrogen-oxygen (N-O) bridges and longer-range ion pairs on the basis of geometrical criteria. In salt bridges, centroids of the side-chain charged groups and at least a pair of side-chain nitrogen and oxygen atoms of the ion-pairing residues are within a 4 Å distance. In N-O bridges, at least a pair of the side-chain nitrogen and oxygen atoms of the ion-pairing residues are within 4 Å distance, but the distance between the side-chain charged group centroids is greater than 4 Å. In the longer-range ion pairs, the side-chain charged group centroids as well as the side-chain nitrogen and oxygen atoms are more than 4 Å apart. Continuum electrostatic calcns. indicate that most of the ion pairs have stabilizing electrostatic contributions when their side-chain charged group centroids are within 5 Å distance. Hence, most (∼92%) of the salt bridges and a majority (68%) of the N-O bridges are stabilizing. Most (∼89%) of the destabilizing ion pairs are the longer-range ion pairs. In the NMR conformer ensembles, the electrostatic interaction between side-chain charged groups of the ion-pairing residues is the strongest for salt bridges, considerably weaker for N-O bridges, and the weakest for longer-range ion pairs. These results suggest empirical rules for stabilizing electrostatic interactions in proteins.
- 65Nogal, B.; Bowman, C. A.; Ward, A. B. Time-course, negative-stain electron microscopy-based analysis for investigating protein-protein interactions at the single-molecule level. J. Biol. Chem. 2017, 292, 19400– 19410, DOI: 10.1074/jbc.m117.80835265Time-course, negative-stain electron microscopy-based analysis for investigating protein-protein interactions at the single-molecule levelNogal, Bartek; Bowman, Charles A.; Ward, Andrew B.Journal of Biological Chemistry (2017), 292 (47), 19400-19410CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Several biophys. approaches are available to study protein-protein interactions. Most approaches are conducted in bulk soln. and are therefore limited to an av. measurement of the ensemble of mol. interactions. Here, single-particle EM can enrich the understanding of protein-protein interactions at the single-mol. level and potentially capture states that are unobservable with ensemble methods because they are below the limit of detection or not conducted on an appropriate time scale. Using the HIV-1 envelope glycoprotein (Env) and its interaction with receptor CD4-binding site neutralizing antibodies as a model system, the authors both corroborate ensemble kinetics-derived parameters and demonstrate how time-course EM can further dissect stoichiometric states of complexes that are not readily observable with other methods. Visualization of the kinetics and stoichiometry of Env-antibody complexes demonstrated the applicability of this approach to qual. and semi-quant. differentiate two highly similar neutralizing antibodies. Furthermore, implementation of machine-learning techniques for sorting class avs. of these complexes into discrete subclasses of particles helped reduce human bias. The authors' data provide proof of concept that single-particle EM can be used to generate a "visual" kinetic profile that should be amenable to studying many other protein-protein interactions, is relatively simple and complementary to well-established biophys. approaches. Moreover, the authors' method provides crit. insights into broadly neutralizing antibody recognition of Env, which may inform vaccine immunogen design and immunotherapeutic development.
- 66Chen, C.; Liu, Y.; Rappaport, A. R.; Kitzing, T.; Schultz, N.; Zhao, Z.; Shroff, A. S.; Dickins, R. A.; Vakoc, C. R.; Bradner, J. E.; Stock, W.; LeBeau, M. M.; Shannon, K. M.; Kogan, S.; Zuber, J.; Lowe, S. W. MLL3 is a haploinsufficient 7q tumor suppressor in acute myeloid leukemia. Cancer Cell 2014, 25, 652– 665, DOI: 10.1016/j.ccr.2014.03.01666MLL3 Is a Haploinsufficient 7q Tumor Suppressor in Acute Myeloid LeukemiaChen, Chong; Liu, Yu; Rappaport, Amy R.; Kitzing, Thomas; Schultz, Nikolaus; Zhao, Zhen; Shroff, Aditya S.; Dickins, Ross A.; Vakoc, Christopher R.; Bradner, James E.; Stock, Wendy; LeBeau, Michelle M.; Shannon, Kevin M.; Kogan, Scott; Zuber, Johannes; Lowe, Scott W.Cancer Cell (2014), 25 (5), 652-665CODEN: CCAECI; ISSN:1535-6108. (Elsevier Inc.)Recurring deletions of chromosome 7 and 7q [-7/del(7q)] occur in myelodysplastic syndromes and acute myeloid leukemia (AML) and are assocd. with poor prognosis. However, the identity of functionally relevant tumor suppressors on 7q remains unclear. Using RNAi and CRISPR/Cas9 approaches, we show that an ∼50% redn. in gene dosage of the mixed lineage leukemia 3 (MLL3) gene, located on 7q36.1, cooperates with other events occurring in -7/del(7q) AMLs to promote leukemogenesis. Mll3 suppression impairs the differentiation of HSPC. Interestingly, Mll3-suppressed leukemias, like human -7/del(7q) AMLs, are refractory to conventional chemotherapy but sensitive to the BET inhibitor JQ1. Thus, our mouse model functionally validates MLL3 as a haploinsufficient 7q tumor suppressor and suggests a therapeutic option for this aggressive disease.
- 67Wu, H.-T.; Liu, Y.-E.; Hsu, K.-W.; Wang, Y.-F.; Chan, Y.-C.; Chen, Y.; Chen, D.-R. MLL3 Induced by Luteolin Causes Apoptosis in Tamoxifen-Resistant Breast Cancer Cells through H3K4 Monomethylation and Suppression of the PI3K/AKT/mTOR Pathway. Am. J. Chin. Med. 2020, 48, 1221– 1241, DOI: 10.1142/s0192415x2050060367MLL3 Induced by Luteolin Causes Apoptosis in Tamoxifen-Resistant Breast Cancer Cells through H3K4 Monomethylation and Suppression of the PI3K/AKT/mTOR PathwayWu, Han-Tsang; Liu, Yi-En; Hsu, Kai-Wen; Wang, Yu-Fen; Chan, Ya-Chi; Chen, Yeh; Chen, Dar-RenAmerican Journal of Chinese Medicine (2020), 48 (5), 1221-1241CODEN: AJCMBA; ISSN:0192-415X. (World Scientific Publishing Co. Pte. Ltd.)Tamoxifen is one of the most common hormone therapy drug for estrogen receptor pos. breast cancer. Tumor cells with drug resistance often cause recurrence and metastasis in cancer patients. Luteolin is a natural compd. found from various types of vegetables and exhibit anticancer activity in different cancers. This study demonstrated that luteolin inhibits the proliferation and induces apoptosis of tamoxifen-resistant ER-pos. breast cancer cells. Luteolin also causes cell cycle arrest at the G2/M phase and decreases mitochondrial membrane potential. Besides, luteolin reduces the levels of activated PI3K/AKT/mTOR signaling pathway. The combination treatment of luteolin and PI3K, AKT, or mTOR inhibitors synergistically increases apoptosis in tamoxifen-resistant ER-pos. breast cancer cells. Ras gene family (K-Ras, H-Ras, and N-Ras), an activator of PI3K, was transcriptionally repressed by luteolin via induction of tumor suppressor mixed-lineage leukemia 3 expression. MLL3 increases the level of monomethylation of Histone 3 Lysine 4 on the enhancer and promoter region of Ras genes, thus causes repression of Ras expressions. Our finding implies that luteolin was a promising natural agent against tamoxifen resistance of breast cancer.
- 68Rampias, T.; Karagiannis, D.; Avgeris, M.; Polyzos, A.; Kokkalis, A.; Kanaki, Z.; Kousidou, E.; Tzetis, M.; Kanavakis, E.; Stravodimos, K.; Manola, K. N.; Pantelias, G. E.; Scorilas, A.; Klinakis, A. The lysine-specific methyltransferase KMT2C/MLL3 regulates DNA repair components in cancer. EMBO Rep. 2019, 20, e46821 DOI: 10.15252/embr.201846821There is no corresponding record for this reference.
- 69Wong, S. H. K.; Goode, D. L.; Iwasaki, M.; Wei, M. C.; Kuo, H.-P.; Zhu, L.; Schneidawind, D.; Duque-Afonso, J.; Weng, Z.; Cleary, M. L. The H3K4-Methyl Epigenome Regulates Leukemia Stem Cell Oncogenic Potential. Cancer Cell 2015, 28, 198– 209, DOI: 10.1016/j.ccell.2015.06.00369The H3K4-Methyl Epigenome Regulates Leukemia Stem Cell Oncogenic PotentialWong, Stephen H. K.; Goode, David L.; Iwasaki, Masayuki; Wei, Michael C.; Kuo, Hsu-Ping; Zhu, Li; Schneidawind, Dominik; Duque-Afonso, Jesus; Weng, Ziming; Cleary, Michael L.Cancer Cell (2015), 28 (2), 198-209CODEN: CCAECI; ISSN:1535-6108. (Elsevier Inc.)The genetic programs that maintain leukemia stem cell (LSC) self-renewal and oncogenic potential have been well defined; however, the comprehensive epigenetic landscape that sustains LSC cellular identity and functionality is less well established. We report that LSCs in MLL-assocd. leukemia reside in an epigenetic state of relative genome-wide high-level H3K4me3 and low-level H3K79me2. LSC differentiation is assocd. with reversal of these broad epigenetic profiles, with concomitant downregulation of crucial MLL target genes and the LSC maintenance transcriptional program that is driven by the loss of H3K4me3, but not H3K79me2. The H3K4-specific demethylase KDM5B neg. regulates leukemogenesis in murine and human MLL-rearranged AML cells, demonstrating a crucial role for the H3K4 global methylome in detg. LSC fate.
- 70Shinsky, S. A.; Cosgrove, M. S. Unique Role of the WD-40 Repeat Protein 5 (WDR5) Subunit within the Mixed Lineage Leukemia 3 (MLL3) Histone Methyltransferase Complex. J. Biol. Chem. 2015, 290, 25819– 25833, DOI: 10.1074/jbc.m115.68414270Unique role of the WD-40 repeat protein 5 (WDR5) subunit within the mixed lineage leukemia 3 (MLL3) histone methyltransferase complexShinsky, Stephen A.; Cosgrove, Michael S.Journal of Biological Chemistry (2015), 290 (43), 25819-25833CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The MLL3 (mixed lineage leukemia 3) protein is a member of the human SET1 family of histone H3 lysine 4 methyltransferases and contains the conserved WDR5 interaction (Win) motif and the catalytic suppressor of variegation, enhancer of zeste, trithorax (SET) domain. The human SET1 family includes MLL1-4 and SETd1A/B, which all interact with a conserved subcomplex contg. WDR5, RbBP5, Ash2L, and DPY-30 (WRAD) to form the minimal core complex required for full methyltransferase activity. However, recent evidence suggests that the WDR5 subunit may not be utilized in an identical manner within all SET1 family core complexes. Although the roles of WDR5 within the MLL1 core complex have been extensively studied, not much is known about the roles of WDR5 in other SET1 family core complexes. In this investigation, we set out to characterize the roles of the WDR5 subunit in the MLL3 core complex. We found that unlike MLL1, the MLL3 SET domain assembles with the RbBP5/Ash2L heterodimer independently of the Win motif-WDR5 interaction. Furthermore, we obsd. that WDR5 inhibits the monomethylation activity of the MLL3 core complex, which is dependent on the Win motif. We also found evidence suggesting that the WRAD subcomplex catalyzes weak H3K4 monomethylation within the context of the MLL3 core complex. Furthermore, soln. structures of the MLL3 core complex assembled with and without WDR5 by small angle x-ray scattering show similar overall topologies. Together, this work demonstrates a unique role for WDR5 in modulating the enzymic activity of the MLL3 core complex.
- 71Wysocka, J.; Swigut, T.; Milne, T. A.; Dou, Y.; Zhang, X.; Burlingame, A. L.; Roeder, R. G.; Brivanlou, A. H.; Allis, C. D. WDR5 associates with histone H3 methylated at K4 and is essential for H3 K4 methylation and vertebrate development. Cell 2005, 121, 859– 872, DOI: 10.1016/j.cell.2005.03.03671WDR5 associates with histone H3 methylated at K4 and is essential for H3 K4 methylation and vertebrate developmentWysocka, Joanna; Swigut, Tomek; Milne, Thomas A.; Dou, Yali; Zhang, Xin; Burlingame, Alma L.; Roeder, Robert G.; Brivanlou, Ali H.; Allis, C. DavidCell (Cambridge, MA, United States) (2005), 121 (6), 859-872CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Histone H3 lysine 4 (K4) methylation has been linked to the transcriptional activation in a variety of eukaryotic species. Here authors show that a common component of MLL1, MLL2, and hSet1 H3 K4 methyltransferase complexes, the WD40-repeat protein WDR5, directly assocs. with histone H3 di- and trimethylated at K4 and with H3-K4-dimethylated nucleosomes. WDR5 is required for binding of the methyltransferase complex to the K4-dimethylated H3 tail as well as for global H3 K4 trimethylation and HOX gene activation in human cells. WDR5 is essential for vertebrate development, in that WDR5-depleted X. laevis tadpoles exhibit a variety of developmental defects and abnormal spatial Hox gene expression. These results are the first demonstration that a WD40-repeat protein acts as a module for recognition of a specific histone modification and suggest a mechanism for reading and writing an epigenetic mark for gene activation.
- 72Itan, Y.; Casanova, J.-L. Can the impact of human genetic variations be predicted?. Proc. Natl. Acad. Sci. U.S.A. 2015, 112, 11426– 11427, DOI: 10.1073/pnas.151505711272Can the impact of human genetic variations be predicted?Itan, Yuval; Casanova, Jean-LaurentProceedings of the National Academy of Sciences of the United States of America (2015), 112 (37), 11426-11427CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)There is no expanded citation for this reference.
- 73Ali, A.; Veeranki, S. N.; Tyagi, S. A SET-domain-independent role of WRAD complex in cell-cycle regulatory function of mixed lineage leukemia. Nucleic Acids Res. 2014, 42, 7611– 7624, DOI: 10.1093/nar/gku45873A SET-domain-independent role of WRAD complex in cell-cycle regulatory function of mixed lineage leukemiaAli, Aamir; Veeranki, Sailaja Naga; Tyagi, ShwetaNucleic Acids Research (2014), 42 (12), 7611-7624CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)MLL, the trithorax ortholog, is a well-characterized histone 3 lysine 4 methyltransferase that is crucial for proper regulation of the Hox genes during embryonic development. Chromosomal translocations, disrupting the Mll gene, lead to aggressive leukemia with poor prognosis. However, the functions of MLL in cellular processes like cell-cycle regulation are not well studied. Here we show that the MLL has a regulatory role during multiple phases of the cell cycle. RNAi-mediated knockdown reveals that MLL regulates S-phase progression and, proper segregation and cytokinesis during M phase. Using deletions and mutations, we narrow the cell-cycle regulatory role to the C subunit of MLL. Our anal. reveals that the transactivation domain and not the SET domain is important for the S-phase function of MLL. Surprisingly, disruption of MLL-WRAD interaction is sufficient to disrupt proper mitotic progression. These mitotic functions of WRAD are independent of SET domain of MLL and, therefore, define a new role of WRAD in subset of MLL functions. Finally, we address the overlapping and unique roles of the different SET family members in the cell cycle.
- 74Movileanu, L.; Cheley, S.; Howorka, S.; Braha, O.; Bayley, H. Location of a Constriction in the Lumen of a Transmembrane Pore by Targeted Covalent Attachment of Polymer Molecules. J. Gen. Physiol. 2001, 117, 239– 252, DOI: 10.1085/jgp.117.3.23974Location of a constriction in the lumen of a transmembrane pore by targeted covalent attachment of polymer moleculesMovileanu, Liviu; Cheley, Stephen; Howorka, Stefan; Braha, Orit; Bayley, HaganJournal of General Physiology (2001), 117 (3), 239-251CODEN: JGPLAD; ISSN:0022-1295. (Rockefeller University Press)Few methods exist for obtaining the internal dimensions of transmembrane pores for which 3-D structures are lacking or for showing that structures detd. by crystallog. reflect the internal dimensions of pores in lipid bilayers. Several approaches, involving polymer penetration and transport, have revealed limiting diams. for various pores. But, in general, these approaches do not indicate the locations of constrictions in the channel lumen. Here, the authors combine cysteine mutagenesis and chem. modification with sulfhydryl-reactive polymers to locate the constriction in the lumen of the staphylococcal α-hemolysin pore, a model protein of known structure. The rates of reaction of each of four polymeric regents (MePEG-OPSS) of different masses towards individual single cysteine mutants, comprising a set with cysteines distributed over the length of the lumen of the pore, were detd. by macroscopic current recording. The rates for the three larger polymers (1.8, 2.5, and 5.0 kDa) were normalized with respect to the rates of reaction with a 1.0 kDa polymer for each of the seven positions in the lumen. The rate of reaction of the 5.0 kDa polymer dropped dramatically at the centrally located Cys-111 residue and positions distal to Cys-111, whether the reagent was applied from the trans or the cis side of the bilayer. This semi-quant. anal. sufficed to demonstrate that a constriction is located at the midpoint of the pore lumen, as predicted by the crystal structure, and although the constriction allows a 2.5 kDa polymer to pass, transport of a 5.0 kDa mol. is greatly restricted. In addn., PEG chains gave greater redns. in pore conductance when covalently attached to the narrower regions of the lumen, permitting further definition of the interior of the pore. The procedures described here should be applicable to other pores and to related structures such as the vestibules of ion channels.
- 75Jarmoskaite, I.; AlSadhan, I.; Vaidyanathan, P. P.; Herschlag, D. How to measure and evaluate binding affinities. eLife 2020, 9, e57264 DOI: 10.7554/eLife.5726475How to measure and evaluate binding affinitiesJarmoskaite, Inga; Alsadhan, Ishraq; Vaidyanathan, Pavanapuresan P.; Herschlag, DanieleLife (2020), 9 (), e57264CODEN: ELIFA8; ISSN:2050-084X. (eLife Sciences Publications Ltd.)Quant. measurements of biomol. assocns. are central to biol. understanding and are needed to build and test predictive and mechanistic models. Given the advances in high-throughput technologies and the projected increase in the availability of binding data, we found it esp. timely to evaluate the current stds. for performing and reporting binding measurements. A review of 100 studies revealed that in most cases essential controls for establishing the appropriate incubation time and concn. regime were not documented, making it impossible to det. measurement reliability. Moreover, several reported affinities could be concluded to be incorrect, thereby impacting biol. interpretations. Given these challenges, we provide a framework for a broad range of researchers to evaluate, teach about, perform, and clearly document high-quality equil. binding measurements. We apply this framework and explain underlying fundamental concepts through exptl. examples with the RNA-binding protein Puf4.
- 76Wolfe, A. J.; Hsueh, Y.-C.; Blanden, A. R.; Mohammad, M. M.; Pham, B.; Thakur, A. K.; Loh, S. N.; Chen, M.; Movileanu, L. Interrogating Detergent Desolvation of Nanopore-Forming Proteins by Fluorescence Polarization Spectroscopy. Anal. Chem. 2017, 89, 8013– 8020, DOI: 10.1021/acs.analchem.7b0133976Interrogating Detergent Desolvation of Nanopore-Forming Proteins by Fluorescence Polarization SpectroscopyWolfe, Aaron J.; Hsueh, Yi-Ching; Blanden, Adam R.; Mohammad, Mohammad M.; Pham, Bach; Thakur, Avinash K.; Loh, Stewart N.; Chen, Min; Movileanu, LiviuAnalytical Chemistry (Washington, DC, United States) (2017), 89 (15), 8013-8020CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Understanding how membrane proteins interact with detergents is of fundamental and practical significance in structural and chem. biol. as well as in nanobiotechnol. Current methods for inspecting protein-detergent complex (PDC) interfaces require high concns. of protein and are of low throughput. Here, the authors describe a scalable, spectroscopic approach that uses nanomolar protein concns. in native solns. This approach, which is based on steady-state fluorescence polarization (FP) spectroscopy, kinetically resolves the dissocn. of detergents from membrane proteins and protein unfolding. For satisfactorily solubilizing detergents, at concns. much greater than the crit. micelle concn. (CMC), the fluorescence anisotropy was independent of detergent concn. In contrast, at detergent concns. comparable with or below the CMC, the anisotropy readout underwent a time-dependent decrease, showing a specific and sensitive protein unfolding signature. Functionally reconstituted membrane proteins into a bilayer membrane confirmed predictions made by these FP-based detns. with respect to varying refolding conditions. From a practical point of view, this 96-well anal. approach will facilitate a massively parallel assessment of the PDC interfacial interactions under a fairly broad range of micellar and environmental conditions. The authors expect that these studies will potentially accelerate research in membrane proteins pertaining to their extn., solubilization, stabilization, and crystn., as well as reconstitution into bilayer membranes.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acschembio.2c00273.
Sequence of SET1Win peptide ligands; location of somatic cancer mutations from tumor samples with N < 500; results of mutation clustering for different mutation subsets; list of tumor locations associated with missense WDR5 mutations within and around the Win binding site; list of noncovalent bonds at the WDR5-SET1Win interface; location of surface WDR5 mutations within the A and B pockets; BLI measurements of very weak interactions; kinetic rate constants of association and dissociation of WDR5 mutants with SET1Win ligands; equilibrium dissociation constants of WDR5 mutants with SET1Win ligands; structural information on the effect of the S175L mutation; steady-state FP spectroscopy curves for the interactions of WDR5 mutants with SET1win ligands; equilibrium dissociation constants of WDR5 mutants with SET1Win ligands using steady-state FP spectroscopy; quantitative comparisons of affinity data acquired with BLI and FP; and supporting references (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.