Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hacer Karatas is active.

Publication


Featured researches published by Hacer Karatas.


Molecular Cell | 2014

Targeting MLL1 H3K4 methyltransferase activity in mixed-lineage leukemia.

Fang Cao; Elizabeth C. Townsend; Hacer Karatas; Jing Xu; Li Li; Shirley Y. Lee; Liu Liu; Yong Chen; Peter Ouillette; Jidong Zhu; Jay L. Hess; Peter Atadja; Ming Lei; Zhaohui S. Qin; Sami N. Malek; Shaomeng Wang; Yali Dou

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.


Journal of the American Chemical Society | 2013

High-Affinity, Small-Molecule Peptidomimetic Inhibitors of MLL1/WDR5 Protein–Protein Interaction

Hacer Karatas; Elizabeth C. Townsend; Fang Cao; Yong Chen; Denzil Bernard; Liu Liu; Ming Lei; Yali Dou; Shaomeng Wang

Mixed lineage leukemia 1 (MLL1) is a histone H3 lysine 4 (H3K4) methyltransferase, and targeting the MLL1 enzymatic 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 enzymatic activity. In the present study, we designed a large number of peptidomimetics to target the MLL1/WDR5 interaction based upon -CO-ARA-NH-, the minimum binding motif derived from MLL1. Our study led to the design of high-affinity peptidomimetics, which bind to WDR5 with K(i) < 1 nM and function as potent antagonists of MLL1 activity in a fully reconstituted in vitro H3K4 methyltransferase assay. Determination 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 compound effectively decreases the expression of HoxA9 and Meis-1, two critical 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-molecule inhibitors of the WDR5/MLL1 protein-protein interaction as a novel therapeutic approach for acute leukemia harboring MLL1 fusion proteins.


Journal of Medicinal Chemistry | 2013

Structure-Based Design of High-Affinity Macrocyclic Peptidomimetics to Block the Menin-Mixed Lineage Leukemia 1 (MLL1) Protein-Protein Interaction.

Haibin Zhou; Liu Liu; Jing Huang; Denzil Bernard; Hacer Karatas; Alexandro Navarro; Ming Lei; Shaomeng Wang

Menin is an essential oncogenic cofactor for mixed lineage leukemia 1 (MLL1)-mediated leukemogenesis through its direct interaction with MLL1. Targeting the menin-MLL1 protein-protein interaction represents a promising strategy to block MLL1-mediated leukemogenesis. Employing a structure-based approach and starting from a linear MLL1 octapeptide, we have designed a class of potent macrocyclic peptidomimetic inhibitors of the menin-MLL1 interaction. The most potent macrocyclic peptidomimetic (MCP-1), 34, binds to menin with a K(i) value of 4.7 nM and is >600 times more potent than the corresponding acyclic peptide. Compound 34 is also less peptide-like and has a lower molecular weight than the initial MLL1 peptide. Therefore, compound 34 serves as a promising lead structure for the design of potent and cell-permeable inhibitors of the menin-MLL1 interaction.


Journal of Medicinal Chemistry | 2010

Analysis of the Binding of Mixed Lineage Leukemia 1 (MLL1) and Histone 3 Peptides to WD Repeat Domain 5 (WDR5) for the Design of Inhibitors of the MLL1−WDR5 Interaction

Hacer Karatas; Elizabeth C. Townsend; Denzil Bernard; Yali Dou; Shaomeng Wang

MLL1 is a histone 3 lysine 4 (H3K4) methyltransferase and a promising new cancer therapeutic target. The catalytic activity of MLL1 is regulated by the formation of a core complex consisting of MLL1, WDR5, RbBP5, and Ash2L. The interaction between WDR5 and MLL1 plays an essential role in regulation of the H3K4 methyltransferase activity of MLL1 and targeting this interaction using small molecules may represent an attractive therapeutic strategy. In this study, we have defined the essential elements in MLL1 required for its high-affinity binding to WDR5. Our data showed that the minimal elements crucial for high-affinity binding of MLL1 to WDR5 are -CO-ARA-NH- motif and two intramolecular hydrogen bonds that stabilize the conformation of this motif. Two 3-mer peptides, Ac-ARA-NH(2) and Ac-ART-NH(2), were designed based upon MLL1 and H3 sequences and achieved K(i) values of 120 and 20 nM to WDR5, respectively. Our study provides a concrete basis for the design of potent peptidomimetics and nonpeptidic compounds to inhibit MLL1 activity by targeting the MLL1 and WDR5 interaction.


Cell Stem Cell | 2016

MLL1 Inhibition Reprograms Epiblast Stem Cells to Naive Pluripotency

Hui Zhang; Srimonta Gayen; Jie Xiong; Bo Zhou; Avinash Kumar Shanmugam; Yuqing Sun; Hacer Karatas; Liu Liu; Rajesh C. Rao; Shaomeng Wang; Alexey I. Nesvizhskii; Sundeep Kalantry; Yali Dou

The interconversion between naive and primed pluripotent states is accompanied by drastic epigenetic rearrangements. However, it is unclear whether intrinsic epigenetic events can drive reprogramming to naive pluripotency or if distinct chromatin states are instead simply a reflection of discrete pluripotent states. Here, we show that blocking histone H3K4 methyltransferase MLL1 activity with the small-molecule inhibitor MM-401 reprograms mouse epiblast stem cells (EpiSCs) to naive pluripotency. This reversion is highly efficient and synchronized, with more than 50% of treated EpiSCs exhibiting features of naive embryonic stem cells (ESCs) within 3 days. Reverted ESCs reactivate the silenced X chromosome and contribute to embryos following blastocyst injection, generating germline-competent chimeras. Importantly, blocking MLL1 leads to global redistribution of H3K4me1 at enhancers and represses lineage determinant factors and EpiSC markers, which indirectly regulate ESC transcription circuitry. These findings show that discrete perturbation of H3K4 methylation is sufficient to drive reprogramming to naive pluripotency.


Cell discovery | 2016

Targeting Mll1 H3K4 methyltransferase activity to guide cardiac lineage specific reprogramming of fibroblasts

Liu Liu; Ienglam Lei; Hacer Karatas; Yangbing Li; Li Wang; Leonid Gnatovskiy; Yali Dou; Shaomeng Wang; Li Qian; Zhong Wang

Generation of induced cardiomyocytes (iCMs) directly from fibroblasts offers a great opportunity for cardiac disease modeling and cardiac regeneration. A major challenge of iCM generation is the low conversion rate. To address this issue, we attempted to identify small molecules that could potentiate the reprogramming ability towards cardiac fate by removing inhibitory roadblocks. Using mouse embryonic fibroblasts as the starting cell source, we first screened 47 cardiac development related epigenetic and transcription factors, and identified an unexpected role of H3K4 methyltransferase Mll1 and related factor Men1 in inhibiting iCM reprogramming. We then applied small molecules (MM408 and MI503) of Mll1 pathway inhibitors and observed an improved efficiency in converting embryonic fibroblasts and cardiac fibroblasts into functional cardiomyocyte-like cells. We further observed that these inhibitors directly suppressed the expression of Mll1 target gene Ebf1 involved in adipocyte differentiation. Consequently, Mll1 inhibition significantly decreased the formation of adipocytes during iCM induction. Therefore, Mll1 inhibitors likely increased iCM efficiency by suppressing alternative lineage gene expression. Our studies show that targeting Mll1 dependent H3K4 methyltransferase activity provides specificity in the process of cardiac reprogramming. These findings shed new light on the molecular mechanisms underlying cardiac conversion of fibroblasts and provide novel targets and small molecules to improve iCM reprogramming for clinical applications.


Journal of Medicinal Chemistry | 2017

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

Hacer Karatas; Yangbing Li; Liu Liu; Jiao Ji; Shirley Y. Lee; Yong Chen; Jiuling Yang; Liyue Huang; Denzil Bernard; Jing Xu; Elizabeth C. Townsend; Fang Cao; Xu Ran; Xiaoqin Li; Bo Wen; Duxin Sun; Jeanne A. Stuckey; Ming Lei; Yali Dou; Shaomeng Wang

We report herein the design, synthesis, and evaluation of macrocyclic peptidomimetics that bind to WD repeat domain 5 (WDR5) and block the WDR5-mixed lineage leukemia (MLL) protein-protein interaction. Compound 18 (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. Compound 18 potently and selectively inhibits cell growth in human leukemia cell lines harboring MLL translocations and is >40 times better than the previously reported compound MM-401. Cocrystal structures of 16 and 18 complexed with WDR5 provide structural basis for their high affinity binding to WDR5. Additionally, we have developed and optimized a new AlphaLISA-based MLL HMT functional assay to facilitate the functional evaluation of these designed compounds. Compound 18 represents the most potent inhibitor of the WDR5-MLL interaction reported to date, and further optimization of 18 may yield a new therapy for acute leukemia.


Cell discovery | 2016

MLL1 and MLL1 fusion proteins have distinct functions in regulating leukemic transcription program

Jing Xu; Li Li; Jie Xiong; Aaron denDekker; Andrew Ye; Hacer Karatas; Liu Liu; He Wang; Zhaohui S. Qin; Shaomeng Wang; Yali Dou

Mixed lineage leukemia protein-1 (MLL1) has a critical role in human MLL1 rearranged leukemia (MLLr) and is a validated therapeutic target. However, its role in regulating global gene expression in MLLr cells, as well as its interplay with MLL1 fusion proteins remains unclear. Here we show that despite shared DNA-binding and cofactor interacting domains at the N terminus, MLL1 and MLL-AF9 are recruited to distinct chromatin regions and have divergent functions in regulating the leukemic transcription program. We demonstrate that MLL1, probably through C-terminal interaction with WDR5, is recruited to regulatory enhancers that are enriched for binding sites of E-twenty-six (ETS) family transcription factors, whereas MLL-AF9 binds to chromatin regions that have no H3K4me1 enrichment. Transcriptome-wide changes induced by different small molecule inhibitors also highlight the distinct functions of MLL1 and MLL-AF9. Taken together, our studies provide novel insights on how MLL1 and MLL fusion proteins contribute to leukemic gene expression, which have implications for developing effective therapies in the future.


Cancer Research | 2012

Abstract LB-256: Targeting MLL1 methyltransferase activity for treatment of acute leukemia with MLL1 rearrangement

Elizabeth C. Townsend; Fang Cao; Hacer Karatas; Shaomeng Wang; Yali Dou

Translocation of the MLL1 gene, which encodes for the histone methyltransferase, MLL1, is found in approximately 10% of all cases of acute leukemia and is generally associated with poor patient outcomes. Recent research has pointed to an essential function for the wild-type copy of MLL1 in these leukemias. In these cells, abnormal upregulation of MLL1-directed histone methylation and subsequent overexpression of MLL1 gene targets is an essential step in the development and progression of leukemia. Efficient MLL1-catalyzed histone methylation can only be achieved when MLL1 associates with the complex of WDR5, RbBP5 and Ash2L. MLL1 complex assembly is nucleated by a highly conserved interaction between the MLL1 SET domain and WDR5. We hypothesize that blocking histone methyltransferase activity of wild-type MLL1 through inhibition of the essential WDR5-MLL1 interaction will prevent abnormal gene upregulation and transformation in leukemias with MLL1 rearrangement. Using rational design, we have developed potent inhibitors for WDR5. We have demonstrated that the lead compound, MM-401, can disrupt the interaction of MLL1 with WDR5, and prevent assembly of the MLL1 complex. We have also shown that MM-401 specifically inhibits MLL1 complex activity, with an IC50=320nM. Using MLL1 fusion-transduced mouse bone marrow cells as a model, we have shown that treating leukemia cells with MM-401 inhibits expression of the MLL1 target oncogene, HoxA9, in a concentration-dependent fashion. Treatment of these cells with MM-401 also promotes differentiation of leukemia blasts in a dose dependent manner. Furthermore, MM-401 specifically impairs cell growth and viability of human leukemia cell lines with MLL1 rearrangement but does not alter growth of leukemia cell lines with mutations or translocations targeting other oncogenes. In conclusion, we have developed a potent inhibitor MM-401, which is the first in a novel class of inhibitors designed to target MLL1 methyltransferase activity, one of the most important chromatin modifying enzymes for transcription activation. We have further demonstrated that pharmacological inhibition of the MLL1 methyltransferase activity specifically targets leukemias with MLL1 rearrangement, and shifts leukemia cells away from the undifferentiated, rapidly proliferating state that is characteristic of aggressive AMLs. These results point to a promising new therapeutic target for leukemia treatment. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr LB-256. doi:1538-7445.AM2012-LB-256


Archive | 2014

BET BROMODOMAIN INHIBITORS AND THERAPEUTIC METHODS USING THE SAME

Shaomeng Wang; Xu Ran; Yujun Zhao; Chao Yie Yang; Liu Liu; Longchuan Bai; Donna McEachern; Jeanne A. Stuckey; Jennifer L. Meagher; Duxin Sun; Xiaoqin Li; Bing Zhou; Hacer Karatas; Ruijuan Luo; Arul M. Chinnaiyan; Irfan A. Asangani

Collaboration


Dive into the Hacer Karatas's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Liu Liu

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Yali Dou

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fang Cao

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jing Xu

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Ming Lei

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yong Chen

Chinese Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge