21 results on '"Kaniskan, H. Ümit"'
Search Results
2. Discovery of Potent and Selective Allosteric Inhibitors of Protein Arginine Methyltransferase 3 (PRMT3).
- Author
-
Kaniskan, H. Ümit, Eram, Mohammad S., Kehao Zhao, Szewczyk, Magdalena M., Xiaobao Yang, Xiao Luo, Sean Xiao, Miao Dai, Feng He, Irene Zang, Ying Lin, Fengling Li, Dobrovetsky, Elena, Smil, David, Sun-Joon Min, Lin-Jones, Jennifer, Schapira, Matthieu, Atadja, Peter, En Li, and Barsyte-Lovejoy, Dalia
- Subjects
- *
PROTEIN arginine methyltransferases , *ALLOSTERIC regulation , *STRUCTURE-activity relationship in pharmacology , *RIBOSOMES , *DRUG development - Abstract
PRMT3 catalyzes the asymmetric dimethylation of arginine residues of various proteins. It is crucial for maturation of ribosomes and has been implicated in several diseases. We recently disclosed a highly potent, selective, and cell-active allosteric inhibitor of PRMT3, compound 4. Here, we report comprehensive structure-activity relationship studies that target the allosteric binding site of PRMT3. We conducted design, synthesis, and evaluation of novel compounds in biochemical, selectivity, and cellular assays that culminated in the discovery of 4 and other highly potent (IC50 values: ~10-36 nM), selective, and cell-active allosteric inhibitors of PRMT3 (compounds 29, 30, 36, and 37). In addition, we generated compounds that are very close analogs of these potent inhibitors but displayed drastically reduced potency as negative controls (compounds 49-51). These inhibitors and negative controls are valuable chemical tools for the biomedical community to further investigate biological functions and disease associations of PRMT3. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
3. Recent advances in developing degraders & inhibitors of lysine methyltransferases.
- Author
-
Velez, Julia, Kaniskan, H. Ümit, and Jin, Jian
- Subjects
- *
LYSINE , *PROTEOLYSIS , *METHYLTRANSFERASES - Abstract
Over the last several decades, there has been continued interest in developing novel therapeutic approaches targeting protein lysine methyltransferases (PKMTs). Along with PKMT inhibitors, targeted protein degradation (TPD) has emerged as a promising strategy to attenuate aberrant PKMT activity. Particularly, proteolysis targeting chimeras (PROTACs) effectively eliminate PKMTs of interest, suppressing all enzymatic and non-enzymatic functions. PROTACs and other TPD approaches add new depth to PKMT research and novel therapeutics discovery. This review focuses on recent advances in PKMT degrader and inhibitor development over the last several years. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
4. Recent progress in developing selective inhibitors of protein methyltransferases.
- Author
-
Kaniskan, H. Ümit and Jin, Jian
- Subjects
- *
METHYLTRANSFERASES , *ENZYME inhibitors , *HISTONE methylation , *GENE expression , *CHROMATIN - Abstract
Mounting evidence suggests that protein methyltransferases (PMTs), which catalyze methylation of histones as well as non-histone proteins, play a crucial role in diverse biological pathways and human diseases. In particular, PMTs have been recognized as major players in regulating gene expression and chromatin state. There has been an increasingly growing interest in these enzymes as potential therapeutic targets and over the past two years tremendous progress has been made in the discovery of selective, small molecule inhibitors of protein lysine and arginine methyltransferases. Inhibitors of PMTs have been used extensively in oncology studies as tool compounds, and inhibitors of EZH2, DOT1L and PRMT5 are currently in clinical trials. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
5. A Potent, Selective and Cell-Active Allosteric Inhibitor of Protein Arginine Methyltransferase 3 (PRMT3).
- Author
-
Kaniskan, H. Ümit, Szewczyk, Magdalena M., Yu, Zhengtian, Eram, Mohammad S., Yang, Xiaobao, Schmidt, Keith, Luo, Xiao, Dai, Miao, He, Feng, Zang, Irene, Lin, Ying, Kennedy, Steven, Li, Fengling, Dobrovetsky, Elena, Dong, Aiping, Smil, David, Min, Sun ‐ Joon, Landon, Melissa, Lin ‐ Jones, Jennifer, and Huang, Xi ‐ Ping
- Subjects
- *
PROTEIN arginine methyltransferases , *HISTONE methylation , *ENZYME inhibitors , *X-ray diffraction , *ALLOSTERIC enzymes - Abstract
PRMT3 catalyzes the asymmetric dimethylation of arginine residues of various proteins. It is essential for maturation of ribosomes, may have a role in lipogenesis, and is implicated in several diseases. A potent, selective, and cell-active PRMT3 inhibitor would be a valuable tool for further investigating PRMT3 biology. Here we report the discovery of the first PRMT3 chemical probe, SGC707, by structure-based optimization of the allosteric PRMT3 inhibitors we reported previously, and thorough characterization of this probe in biochemical, biophysical, and cellular assays. SGC707 is a potent PRMT3 inhibitor (IC50=31±2 n M, KD=53±2 n M) with outstanding selectivity (selective against 31 other methyltransferases and more than 250 non-epigenetic targets). The mechanism of action studies and crystal structure of the PRMT3-SGC707 complex confirm the allosteric inhibition mode. Importantly, SGC707 engages PRMT3 and potently inhibits its methyltransferase activity in cells. It is also bioavailable and suitable for animal studies. This well-characterized chemical probe is an excellent tool to further study the role of PRMT3 in health and disease. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
6. A Potent, Selective and Cell-Active Allosteric Inhibitor of Protein Arginine Methyltransferase 3 (PRMT3).
- Author
-
Kaniskan, H. Ümit, Szewczyk, Magdalena M., Yu, Zhengtian, Eram, Mohammad S., Yang, Xiaobao, Schmidt, Keith, Luo, Xiao, Dai, Miao, He, Feng, Zang, Irene, Lin, Ying, Kennedy, Steven, Li, Fengling, Dobrovetsky, Elena, Dong, Aiping, Smil, David, Min, Sun‐Joon, Landon, Melissa, Lin‐Jones, Jennifer, and Huang, Xi‐Ping
- Subjects
- *
PROTEIN arginine methyltransferases , *ARGININE , *ALLOSTERIC enzymes , *METHYLATION , *LIPID synthesis - Abstract
PRMT3 catalyzes the asymmetric dimethylation of arginine residues of various proteins. It is essential for maturation of ribosomes, may have a role in lipogenesis, and is implicated in several diseases. A potent, selective, and cell-active PRMT3 inhibitor would be a valuable tool for further investigating PRMT3 biology. Here we report the discovery of the first PRMT3 chemical probe, SGC707, by structure-based optimization of the allosteric PRMT3 inhibitors we reported previously, and thorough characterization of this probe in biochemical, biophysical, and cellular assays. SGC707 is a potent PRMT3 inhibitor (IC50=31±2 n M, KD=53±2 n M) with outstanding selectivity (selective against 31 other methyltransferases and more than 250 non-epigenetic targets). The mechanism of action studies and crystal structure of the PRMT3-SGC707 complex confirm the allosteric inhibition mode. Importantly, SGC707 engages PRMT3 and potently inhibits its methyltransferase activity in cells. It is also bioavailable and suitable for animal studies. This well-characterized chemical probe is an excellent tool to further study the role of PRMT3 in health and disease. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
7. Selective Inhibitors of ProteinMethyltransferases.
- Author
-
Kaniskan, H. Ümit, Konze, Kyle D., and Jian Jin
- Subjects
- *
METHYLTRANSFERASES , *ENZYME inhibitors , *CATALYSIS , *HISTONE methylation , *GENETIC regulation , *DRUG development - Abstract
Mountingevidence suggests that protein methyltransferases (PMTs),which catalyze methylation of histone and nonhistone proteins, playa crucial role in diverse biological processes and human diseases.In particular, PMTs have been recognized as major players in regulatinggene expression and chromatin state. PMTs are divided into two categories:protein lysine methyltransferases (PKMTs) and protein arginine methyltransferases(PRMTs). There has been a steadily growing interest in these enzymesas potential therapeutic targets and therefore discovery of PMT inhibitorshas also been pursued increasingly over the past decade. Here, wepresent a perspective on selective, small-molecule inhibitors of PMTswith an emphasis on their discovery, characterization, and applicabilityas chemical tools for deciphering the target PMTs’ physiologicalfunctions and involvement in human diseases. We highlight the currentstate of PMT inhibitors and discuss future directions and opportunitiesfor PMT inhibitor discovery. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
8. Chemically induced degradation of epigenetic targets.
- Author
-
Kabir, Md, Yu, Xufen, Kaniskan, H. Ümit, and Jin, Jian
- Subjects
- *
EPIGENETICS , *TUMOR growth , *SMALL molecules , *CANCER invasiveness , *TREATMENT effectiveness - Abstract
Proteolysis-targeting chimeras (PROTACs) are heterobifunctional small molecules that induce the ternary complex formation between a protein-of-interest (POI) and an E3 ligase, leading to targeted polyubiquitination and degradation of the POI. Particularly, PROTACs have the distinct advantage of targeting both canonical and noncanonical functions of epigenetic targets over traditional inhibitors, which typically target canonical functions only, resulting in greater therapeutic efficacy. In this review, we methodically analyze published PROTAC degraders of epigenetic writer, reader, and eraser proteins and their in vitro and in vivo effects. We highlight the mechanism of action of these degraders and their advantages in targeting both canonical and noncanonical functions of epigenetic targets in the context of cancer treatment. Furthermore, we present a future outlook for this exciting field. Overall, pharmacological degradation of epigenetic targets has emerged as an effective and attractive strategy to thwart cancer progression and growth. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
9. Asymmetric [C + NC + CC] Coupling Entry to the Naphthyridinomycin Natural Product Family: Formal Total Synthesis of Cyanocycline A and Bioxalomycin β2.
- Author
-
Garner, Philip, Kaniskan, H. Ümit, Keyari, Charles M., and Weerasinghe, Laksiri
- Subjects
- *
CELL migration , *NATURAL products , *ALDEHYDES , *PYRROLIDINE , *ACRYLATES - Abstract
A full account of our [C + NC + CC] coupling approach to the naphthyridinomycin family of natural products is presented, culminating in formal total syntheses of cyanocycline A and bioxalomycin β2. The key complexity-building reaction in the synthesis involves the AgI-catalyzed endo-selective [C + NC + CC] coupling of aldehyde 7, (S)-glycyl sultam 8, and methyl acrylate (9) to provide the highly functionalized pyrrolidine 6, which was carried forward to an advanced intermediate (compound 33) in Fukuyama's synthesis of cyanocycline A. Since cyanocycline A has been converted to bioxalomycin β2, this constitutes a formal synthesis of the latter natural product as well. The multicomponent reaction-based strategy reduces the number of steps previously needed to assemble these complex molecular targets by one-third. This work highlights the utility of the asymmetric [C + NC + CC] coupling reaction in the context of a complex pyrrolidine-containing target and provides an illustrative guide for its application to other synthesis problems. The synthesis also fueled collaborative biological and biochemical research that identified a unique small molecule inhibitor of cell migration (compound 30). [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
10. Synthesis of Highly Functionalized Pyrrolidines via a Mild One-Pot, Three-Component 1,3-Dipolar Cycloaddition Process.
- Author
-
Garner, Philip and Kaniskan, H. Ümit
- Subjects
- *
CHEMISTRY education , *RING formation (Chemistry) , *ORGANIC compounds , *ALKENES , *IMINES , *YLIDES - Abstract
A simple and efficient one-pot, three-component synthesis of highly functionalized pyrrolidines via cascade imine ↵ azomethine ylide ↵ 1,3-dipolar cycloadditions is reported. Admixing a variety of aldehydes, dimethyl 2-aminomalonate, and electron deficient alkenes in THF leads to the clean production of pyrrolidines in good to excellent yields. The mild reaction conditions enabled the generation of previously inaccessible azomethine ylides from enolizable aldehydes. Endo selectivity was exclusive with N-phenyl maleimide and maleic anhydride. Good chemo-, regio-, and stereoselectivities were observed with methyl acrylate, though catalysis by Ag(I) was necessary with this dipolarophile. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
11. A stereodivergent cascade imine→azomethine ylide→1,3-dipolar cycloadditive approach to α-chiral pyrrolidines
- Author
-
Garner, Philip and Kaniskan, H. Ümit
- Subjects
- *
CHIRALITY , *PYRROLIDINE , *RING formation (Chemistry) , *CHEMICAL reactions - Abstract
Abstract: Stereodivergent [3+2] cycloadditions of chiral α-amino azomethine ylides leading to highly functionalized pyrrolidines are reported. The marriage of substrate conformational preferences and either an inter- or intramolecular cycloaddition manifold leads to either the l (syn) or u (anti) relationship between the pyrrolidine and α-stereocenters. The latter result may be applicable to a new approach to the bioxalomycin family of antibiotics. [Copyright &y& Elsevier]
- Published
- 2005
- Full Text
- View/download PDF
12. Synthesis of 3-Azabicyclo[3.3.0]octane Framework and a Novel Tricyclicaminoether.
- Author
-
Kaniskan, H. Ümit and Dogan, Özdemir
- Subjects
- *
CHEMICAL reagents , *LACTONES , *MACROLIDE antibiotics , *EPOXY compounds , *KETONES - Abstract
The efficient synthesis of 3-azabicyclo[3.3.0]octane derivative 1 and a novel tricyclicaminoether 11 has been achieved in seven steps using commercially available starting materials and commonly used reagents. The key steps are the intramolecular ring opening of epoxide 7 providing tricyclic aminoalcohol 3 and Baeyer-Villiger oxidation of ketone 8 providing the corresponding lactone 2. Both reactions proceeded regiospecifically. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
13. A potent and selective ENL degrader suppresses oncogenic gene expression and leukemia progression.
- Author
-
Zhaoyu Xue, Lihuai Qin, Hongwen Xuan, Kaixiu Luo, Mengying Huang, Ling Xie, Yangzhou Su, Longxia Xu, Harsh, Josiah, Dale, Brandon, Xiaobing Shi, Xian Chen, Kaniskan, H. Ümit, Jian Jin, and Hong Wen
- Subjects
- *
GENE expression , *NEPHROBLASTOMA , *LEUKEMIA , *ACUTE leukemia , *MUTANT proteins - Abstract
The histone acylation reader eleven-nineteen leukemia (ENL) plays a pivotal role in sustaining oncogenesis in acute leukemias, particularly in mixed-lineage leukemia-rearranged (MLL-r) leukemia. ENL relies on its reader domain to recognize histone lysine acylation promoting oncogenic gene expression and leukemia progression. Here, we report the development of MS41, a highly potent and selective von Hippel-Lindau-recruiting ENL degrader that effectively inhibits the growth of ENL-dependent leukemia cells. MS41-induced ENL degradation reduces the chromatin occupancy of ENL-associated transcription elongation machinery, resulting in the suppression of key oncogenic gene expression programs and the activation of differentiation genes. MS41 is well-tolerated in vivo and substantially suppresses leukemia progression in a xenograft mouse model of MLL-r leukemia. Notably, MS41 also induces the degradation of mutant ENL proteins identified in Wilms' tumors. Our findings emphasize the therapeutic potential of pharmacological ENL degradation for treating ENL-dependent cancers, making MS41 not only a valuable chemical probe but also potential anticancer therapeutic for further development. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. An Efficient Synthetic Approach to Cyanocycline A and Bioxalomycin β2 via [C+NC+CC] Coupling.
- Author
-
Kaniskan, H. Ümit and Garner, Philip
- Subjects
- *
PYRROLIDINE , *ORGANIC compounds , *NANOSTRUCTURED materials , *CHEMICAL research , *MOLECULAR biology - Abstract
The article discusses the effectiveness of the coupling reactions technology in simplifying the synthesis of the cyanocyclines and bioxalomycins. According to the article, the said synthetic approach provides rapid access to a key pyrrolidine intermediate structure that is converted to the target using established chemistry. Moreover, the key coupling reaction was described to be effected by combining aldehyde 5 and L-glycylsultam 6 in methyl acrylate with 10 mol % AgOAc at room temperature.
- Published
- 2007
- Full Text
- View/download PDF
15. Analogs of Tetrahydroisoquinoline Natural Products That Inhibit Cell Migration and Target Galectin-3 Outside of Its Carbohydrate-binding Site.
- Author
-
Kahsai, Alem W., Cui, Junru, Kaniskan, H. Ümit, Garners, Philip P., and Fenteany, Gabriel
- Subjects
- *
CELL migration , *ORGANIC compounds , *NUCLEIC acids , *CELL adhesion , *RNA - Abstract
Cell migration is central to a number of normal and disease processes. Small organic molecules that inhibit cell migration have potential as both research probes and therapeutic agents. We have identified two tetrahydroisoquinoline natural product analogs with antimigratory activities on Madin-Darby canine kidney epithelial cells: a semisynthetic derivative of quinocarmycin (also known as quinocarcin), DX-52-1, and a more complex synthetic molecule, HUK-921, related to the naphthyridinomycin family. It has been assumed that the cellular effects of reactive tetrahydroisoquinolines result from the alkylation of DNA. We have reported previously that the primary target of DX-52-1 relevant to cell migration appears to be the membrane-cytoskeleton linker protein radixin. Here we extend the analysis of the protein targets of DX-52-1, reporting that the multifunctional carbohydrate-binding protein galectin-3 is a secondary target of DX-52-1 that may also be relevant to the antimigratory effects of both DX-52-1 and HUK-921. All known inhibitors of galectin-3 target β-galactoside-binding site in the carbohydrate recognition domain. However, we found that DX-52-1 and HUK-921 bind galectin-3 outside of β-galactoside-binding site. Intriguingly HUK-921, although a less potent inhibitor of cell migration than DX-52-1, had far greater selectivity for galectin-3 over radixin, exhibiting little binding to radixin, both in vitro and in cells. Overexpression of galectin-3 in cells led to a dramatic increase in cell adhesion on different extracellular matrix substrata as well as changes in cell-cell adhesion and cell motility. Galectin-3-overexpressing cells had greatly reduced sensitivity to DX-52-1 and HUK-921, and these compounds caused a change in localization of the overexpressed galectin-3 and reversion of the cells to a more normal morphology. The converse manipulation, RNA interference-based silencing of galectin-3 expression, resulted in reduced cell-matrix adhesion and cell migration. In aggregate, the data suggest that DX-52-1 and HUK-921 inhibit a carbohydrate binding-independent function of galectin-3 that is involved in cell migration. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
16. Discovery of a novel, highly potent EZH2 PROTAC degrader for targeting non-canonical oncogenic functions of EZH2.
- Author
-
Velez, Julia, Dale, Brandon, Park, Kwang-Su, Kaniskan, H. Ümit, Yu, Xufen, and Jin, Jian
- Subjects
- *
BREAST , *ACUTE myeloid leukemia , *TRIPLE-negative breast cancer , *UBIQUITIN ligases , *CELL growth , *CELL lines - Abstract
Aberrant expression of EZH2, the main catalytic subunit of PRC2, has been implicated in numerous cancers, including leukemia, breast, and prostate. Recent studies have highlighted non-catalytic oncogenic functions of EZH2, which EZH2 catalytic inhibitors cannot attenuate. Therefore, proteolysis-targeting chimera (PROTAC) degraders have been explored as an alternative therapeutic approach to suppress both canonical and non-canonical oncogenic activity. Here we present MS8847, a novel, highly potent EZH2 PROTAC degrader that recruits the E3 ligase von Hippel-Lindau (VHL). MS8847 degrades EZH2 in a concentration-, time-, and ubiquitin–proteasome system (UPS)-dependent manner. Notably, MS8847 induces superior EZH2 degradation and anti-proliferative effects in MLL-rearranged (MLL-r) acute myeloid leukemia (AML) cells compared to previously published EZH2 PROTAC degraders. Moreover, MS8847 degrades EZH2 and inhibits cell growth in triple-negative breast cancer (TNBC) cell lines, displays efficacy in a 3D TNBC in vitro model, and has a pharmacokinetic (PK) profile suitable for in vivo efficacy studies. Overall, MS8847 is a valuable chemical tool for the biomedical community to investigate canonical and non-canonical oncogenic functions of EZH2. [Display omitted] • A novel and highly effective EZH2 PROTAC degrader, MS8847, was discovered. • MS8847 potently degrades EZH2 in a ubiquitin-proteasome system-dependent manner. • MS8847 targets both canonical and non-canonical oncogenic functions of EZH2. • MS8847 effectively inhibits the growth of AML and TNBC cells. • MS8847 is bioavailable in mice and suitable for in vivo efficacy studies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Epigenetic balance ensures mechanistic control of MLL amplification and rearrangement.
- Author
-
Gray, Zach H., Chakraborty, Damayanti, Duttweiler, Reuben R., Alekbaeva, Gulnaz D., Murphy, Sedona E., Chetal, Kashish, Ji, Fei, Ferman, Benjamin I., Honer, Madison A., Wang, Zhentian, Myers, Cynthia, Sun, Renhong, Kaniskan, H. Ümit, Toma, Monika Maria, Bondarenko, Elena A., Santoro, John N., Miranda, Christopher, Dillingham, Megan E., Tang, Ran, and Gozani, Or
- Subjects
- *
DOXORUBICIN , *EPIGENETICS , *DRUG target , *DEMETHYLASE , *GENE amplification , *LYSINE - Abstract
MLL/KMT2A amplifications and translocations are prevalent in infant, adult, and therapy-induced leukemia. However, the molecular contributor(s) to these alterations are unclear. Here, we demonstrate that histone H3 lysine 9 mono- and di-methylation (H3K9me1/2) balance at the MLL/KMT2A locus regulates these amplifications and rearrangements. This balance is controlled by the crosstalk between lysine demethylase KDM3B and methyltransferase G9a/EHMT2. KDM3B depletion increases H3K9me1/2 levels and reduces CTCF occupancy at the MLL/KMT2A locus, in turn promoting amplification and rearrangements. Depleting CTCF is also sufficient to generate these focal alterations. Furthermore, the chemotherapy doxorubicin (Dox), which associates with therapy-induced leukemia and promotes MLL/KMT2A amplifications and rearrangements, suppresses KDM3B and CTCF protein levels. KDM3B and CTCF overexpression rescues Dox-induced MLL/KMT2A alterations. G9a inhibition in human cells or mice also suppresses MLL/KMT2A events accompanying Dox treatment. Therefore, MLL/KMT2A amplifications and rearrangements are controlled by epigenetic regulators that are tractable drug targets, which has clinical implications. [Display omitted] • KDM3B and G9a regulate MLL/KMT2A copy gains and rearrangements through H3K9me1/2 • CTCF depletion and reduced binding associate with KMT2A copy gains and break aparts • Doxorubicin reduces KDM3B and CTCF levels, promoting MLL/KMT2A alterations • G9a inhibition suppresses Dox-induced MLL/KMT2A alterations in mice and human cells An imbalance in histone modifications contributes to both transient amplifications and integrated rearrangements and amplifications observed in leukemia, which can be induced by a commonly used chemotherapy but prevented by chemical intervention. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
18. Discovery of a Potent, Selective, and Cell-Active Dual Inhibitor of Protein Arginine Methyltransferase 4 and Protein Arginine Methyltransferase 6.
- Author
-
Yudao Shen, Szewczyk, Magdalena M., Eram, Mohammad S., Smil, David, Kaniskan, H. Ümit, Ferreira de Freitas, Renato, Senisterra, Guillermo, Fengling Li, Schapira, Matthieu, Brown, Peter J., Arrowsmith, Cheryl H., Barsyte-Lovejoy, Dalia, Jing Liu, Vedadi, Masoud, and Jian Jin
- Subjects
- *
PROTEIN arginine methyltransferases , *ENZYME inhibitors , *STRUCTURE-activity relationship in pharmacology , *EPIGENETICS , *CHEMICAL biology - Abstract
Well-characterized selective inhibitors of protein arginine methyltransferases (PRMTs) are invaluable chemical tools for testing biological and therapeutic hypotheses. Based on 4, a fragment-like inhibitor of type I PRMTs, we conducted structure-activity relationship (SAR) studies and explored three regions of this scaffold. The studies led to the discovery of a potent, selective, and cell-active dual inhibitor of PRMT4 and PRMT6, 17 (MS049). As compared to 4, 17 displayed much improved potency for PRMT4 and PRMT6 in both biochemical and cellular assays. It was selective for PRMT4 and PRMT6 over other PRMTs and a broad range of other epigenetic modifiers and nonepigenetic targets. We also developed 46 (MS049N), which was inactive in biochemical and cellular assays, as a negative control for chemical biology studies. Considering possible overlapping substrate specificity of PRMTs, 17 and 46 are valuable chemical tools for dissecting specific biological functions and dysregulation of PRMT4 and PRMT6 in health and disease. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
19. Structure-Activity Relationship Studies for Enhancer of Zeste Homologue 2 (EZH2) and Enhancer of Zeste Homologue 1 (EZH1) Inhibitors.
- Author
-
Xiaobao Yang, Fengling Li, Konze, Kyle D., Meslamani, Jamel, Ma, Anqi, Brown, Peter J., Ming-Ming Zhou, Arrowsmith, Cheryl H., Kaniskan, H. Ümit, Vedadi, Masoud, and Jian Jin
- Subjects
- *
HISTONE methylation , *STRUCTURE-activity relationships , *POLYCOMB group proteins , *CATALYSIS , *LEUKEMIA - Abstract
EZH2 or EZH1 (enhancer of zeste homologue 2 or 1) is the catalytic subunit of polycomb repressive complex 2 (PRC2) that catalyzes methylation of histone H3 lysine 27 (H3K27). PRC2 hyperactivity and/or hypertrimethylation of H3K27 are associated with numerous human cancers, therefore inhibition of PRC2 complex has emerged as a promising therapeutic approach. Recent studies have shown that EZH2 and EZH1 are not functionally redundant and inhibition of both EZH2 and EZH1 is necessary to block the progression of certain cancers such as mixed-lineage leukemia (MLL)-rearranged leukemias. Despite the significant advances in discovery of EZH2 inhibitors, there has not been a systematic structure-activity relationship (SAR) study to investigate the selectivity between EZH2 and EZH1 inhibition. Here, we report our SAR studies that focus on modifications to various regions of the EZH2/1 inhibitor UNC1999 (5) to investigate the impact of the structural changes on EZH2 and EZH1 inhibition and selectivity. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
20. A NSD3-targeted PROTAC suppresses NSD3 and cMyc oncogenic nodes in cancer cells.
- Author
-
Xu, Chenxi, Meng, Fanye, Park, Kwang-Su, Storey, Aaron J., Gong, Weida, Tsai, Yi-Hsuan, Gibson, Elisa, Byrum, Stephanie D., Li, Dongxu, Edmondson, Rick D., Mackintosh, Samuel G., Vedadi, Masoud, Cai, Ling, Tackett, Alan J., Kaniskan, H. Ümit, Jin, Jian, and Wang, Gang Greg
- Subjects
- *
UBIQUITIN ligases , *PROTEIN domains , *HEMATOLOGIC malignancies , *TUMOR growth , *GENE expression , *CANCER cells - Abstract
Nuclear receptor binding SET domain protein 3 (NSD3), a gene located within the 8p11-p12 amplicon frequently detected in human cancers, encodes a chromatin modulator and an attractive onco-target. However, agents that effectively suppress NSD3-mediated oncogenic actions are currently lacking. We report the NSD3-targeting proteolysis targeting chimera (PROTAC), MS9715, which achieves effective and specific targeting of NSD3 and associated cMyc node in tumor cells. MS9715 is designed by linking BI-9321, a NSD3 antagonist, which binds NSD3's PWWP1 domain, with an E3 ligase VHL ligand. Importantly, MS9715, but not BI-9321, effectively suppresses growth of NSD3-dependent hematological cancer cells. Transcriptomic profiling demonstrates that MS9715, but not BI-9321, effectively suppresses NSD3-and cMyc-associated gene expression programs, resembling effects of the CRISPR-Cas9-mediated knockout of NSD3. Collectively, these results suggest that pharmacological degradation of NSD3 as an attractive therapeutic strategy, which co-suppresses NSD3- and cMyc-related oncogenic nodes, is superior to blocking the PWWP1 domain of NSD3. [Display omitted] • We discover the NSD3 PROTAC, MS9715, which effectively degrades NSD3 • MS9715 suppresses both NSD3- and cMyc-related oncogenic nodes in tumor cells • MS9715 is superior to the reported NSD3 antagonist BI-9321 in targeting NSD3 • MS9715, but not BI-9321, is effective in suppressing the growth of tumor cells Xu et al. report the discovery and characterization of the NSD3 PROTAC, MS9715, which effectively and selectively depletes NSD3 and the associated cMyc oncogenic node. We provide evidence that pharmacological degradation of NSD3 is a superior therapeutic strategy to pharmacological inhibition of NSD3 for treating NSD3-dependent cancers. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
21. A promising chemical series of positive allosteric modulators of the μ-opioid receptor that enhance the antinociceptive efficacy of opioids but not their adverse effects.
- Author
-
Pryce, Kerri D., Kang, Hye Jin, Sakloth, Farhana, Liu, Yongfeng, Khan, Susan, Toth, Katalin, Kapoor, Abhijeet, Nicolais, Andrew, Che, Tao, Qin, Lihuai, Bertherat, Feodora, Kaniskan, H. Ümit, Jin, Jian, Cameron, Michael D., Roth, Bryan L., Zachariou, Venetia, and Filizola, Marta
- Subjects
- *
OPIOID receptors , *OPIOID abuse , *OPIOIDS , *DRUG administration , *LABORATORY mice , *SMALL molecules - Abstract
Positive allosteric modulators (PAMs) of the μ-opioid receptor (MOR) have been proposed to exhibit therapeutic potential by maximizing the analgesic properties of clinically used opioid drugs while limiting their adverse effects or risk of overdose as a result of using lower drug doses. We herein report in vitro and in vivo characterization of two small molecules from a chemical series of MOR PAMs that exhibit: (i) MOR PAM activity and receptor subtype selectivity in vitro , (ii) a differential potentiation of the antinociceptive effect of oxycodone, morphine, and methadone in mouse models of pain that roughly correlates with in vitro activity, and (iii) a lack of potentiation of adverse effects associated with opioid administration, such as somatic withdrawal, respiratory depression, and analgesic tolerance. This series of MOR PAMs holds promise for the development of adjuncts to opioid therapy to mitigate against overdose and opioid use disorders. • MOR PAMs hold promise for the development of adjuncts to opioid therapy. • MOR PAM activity does not appear to require biased signaling. • Differential enhancement of the antinociceptive effect of classical opioid drugs. • Lack of potentiation of tolerance, withdrawal, and respiratory depression. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.