Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Jin-Yi Zhu.
Journal of Medicinal Chemistry | 2012
Harshani R. Lawrence; Matthew P. Martin; Yunting Luo; Roberta Pireddu; Hua Yang; Harsukh Gevariya; Sevil Ozcan; Jin-Yi Zhu; Robert Kendig; Mercedes Rodriguez; Roy Elias; Jin Q. Cheng; Said M. Sebti; Ernst Schönbrunn; Nicholas J. Lawrence
The o-carboxylic acid substituted bisanilinopyrimidine 1 was identified as a potent hit (Aurora A IC(50) = 6.1 ± 1.0 nM) from in-house screening. Detailed structure-activity relationship (SAR) studies indicated that polar substituents at the para position of the B-ring are critical for potent activity. X-ray crystallography studies revealed that compound 1 is a type I inhibitor that binds the Aurora kinase active site in a DFG-in conformation. Structure-activity guided replacement of the A-ring carboxylic acid with halogens and incorporation of fluorine at the pyrimidine 5-position led to highly potent inhibitors of Aurora A that bind in a DFG-out conformation. B-Ring modifications were undertaken to improve the solubility and cell permeability. Compounds such as 9m with water-solubilizing moieties at the para position of the B-ring inhibited the autophosphorylation of Aurora A in MDA-MB-468 breast cancer cells.
ChemBioChem | 2012
Mathew P. Martin; Riazul Alam; Stephane Betzi; Donna J. Ingles; Jin-Yi Zhu; Ernst Schönbrunn
In an attempt to identify novel small‐molecule ligands of cyclin‐dependent kinase 2 (CDK2) with potential as allosteric inhibitors, we have devised a robust and cost‐effective fluorescence‐based high‐throughput screening assay. The assay is based on the specific interaction of CDK2 with the extrinsic fluorophore 8‐anilino‐1‐naphthalene sulfonate (ANS), which binds to a large allosteric pocket adjacent to the ATP site. Hit compounds that displace ANS directly or indirectly from CDK2 are readily classified as ATP site binders or allosteric ligands through the use of staurosporine, which blocks the ATP site without displacing ANS. Pilot screening of 1453 compounds led to the discovery of 12 compounds with displacement activities (EC50 values) ranging from 6 to 44 μM, all of which were classified as ATP‐site‐directed ligands. Four new type I inhibitor scaffolds were confirmed by X‐ray crystallography. Although this small compound library contained only ATP‐site‐directed ligands, the application of this assay to large compound libraries has the potential to reveal previously unrecognized chemical scaffolds suitable for structure‐based design of CDK2 inhibitors with new mechanisms of action.
ACS Chemical Biology | 2012
Mathew P. Martin; Jin-Yi Zhu; Harshani R. Lawrence; Roberta Pireddu; Yunting Luo; Riazul Alam; Sevil Ozcan; Said M. Sebti; Nicholas J. Lawrence; Ernst Schönbrunn
Most protein kinases share a DFG (Asp-Phe-Gly) motif in the ATP site that can assume two distinct conformations, the active DFG-in and the inactive DFG-out states. Small molecule inhibitors able to induce the DFG-out state have received considerable attention in kinase drug discovery. Using a typical DFG-in inhibitor scaffold of Aurora A, a kinase involved in the regulation of cell division, we found that halogen and nitrile substituents directed at the N-terminally flanking residue Ala273 induced global conformational changes in the enzyme, leading to DFG-out inhibitors that are among the most potent Aurora A inhibitors reported to date. The data suggest an unprecedented mechanism of action, in which induced-dipole forces along the Ala273 side chain alter the charge distribution of the DFG backbone, allowing the DFG to unwind. As the ADFG sequence and three-dimensional structure is highly conserved, DFG-out inhibitors of other kinases may be designed by specifically targeting the flanking alanine residue with electric dipoles.
MedChemComm | 2012
Roberta Pireddu; Kara D. Forinash; Nan N. Sun; Mathew P. Martin; Shen-Shu Sung; Brian G. Alexander; Jin-Yi Zhu; Wayne C. Guida; Ernst Schönbrunn; Said M. Sebti; Nicholas J. Lawrence
Potent ROCK inhibitors of a new class of 1-benzyl-3-(4-pyridylthiazol-2-yl)ureas have been identified. Remarkable differences in activity were observed for ureas bearing a benzylic stereogenic center. Derivatives with hydroxy, methoxy and amino groups at the meta position of the phenyl ring give rise to the most potent inhibitors (low nM). Substitutions at the para position result in substantial loss of potency. Changes at the benzylic position are tolerated resulting in significant potency in the case of methyl and methylenehydroxy groups. X-Ray crystallography was used to establish the binding mode of this class of inhibitors and provides an explanation for the observed differences of the enantiomer series. Potent inhibition of ROCK in human lung cancer cells was shown by suppression of the levels of phosphorylation of the ROCK substrate MYPT-1.
Journal of Biological Chemistry | 2012
Jin-Yi Zhu; Yan Yang; Huijong Han; Stéphane Betzi; Sanne H. Olesen; Frank Marsilio; Ernst Schönbrunn
Background: MurA is critical for the biosynthesis of the bacterial cell wall. Results: The covalent MurA-phosphoenolpyruvate adduct was captured in different reaction states. Conclusion: The covalent adduct primes phosphoenolpyruvate for catalysis and enables feedback inhibition by UDP-N-acetylmuramic acid, the product of MurB. Significance: Cellular MurA exists in a previously unrecognized and tightly locked complex, which presumably accounts for the failure of drug discovery efforts. The enzyme MurA has been an established antibiotic target since the discovery of fosfomycin, which specifically inhibits MurA by covalent modification of the active site residue Cys-115. Early biochemical studies established that Cys-115 also covalently reacts with substrate phosphoenolpyruvate (PEP) to yield a phospholactoyl adduct, but the structural and functional consequences of this reaction remained obscure. We captured and depicted the Cys-115-PEP adduct of Enterobacter cloacae MurA in various reaction states by X-ray crystallography. The data suggest that cellular MurA predominantly exists in a tightly locked complex with UDP-N-acetylmuramic acid (UNAM), the product of the MurB reaction, with PEP covalently attached to Cys-115. The uniqueness and rigidity of this “dormant” complex was previously not recognized and presumably accounts for the failure of drug discovery efforts toward the identification of novel and effective MurA inhibitors. We demonstrate that recently published crystal structures of MurA from various organisms determined by different laboratories were indeed misinterpreted and actually contain UNAM and covalently bound PEP. The Cys-115-PEP adduct was also captured in vitro during the reaction of free MurA and substrate UDP-N-acetylglucosamine or isomer UDP-N-acetylgalactosamine. The now available series of crystal structures allows a comprehensive view of the reaction cycle of MurA. It appears that the covalent reaction of MurA with PEP fulfills dual functions by tightening the complex with UNAM for the efficient feedback regulation of murein biosynthesis and by priming the PEP molecule for instantaneous reaction with substrate UDP-N-acetylglucosamine.
Molecular Cancer Therapeutics | 2017
Stuart W. J. Ember; Que T. Lambert; Norbert Berndt; Steven Gunawan; Muhammad Ayaz; Marilena Tauro; Jin-Yi Zhu; Paula J. Cranfill; Patricia Greninger; Conor C. Lynch; Cyril H. Benes; Harshani R. Lawrence; Gary W. Reuther; Nicholas J. Lawrence; Ernst Schönbrunn
Synergistic action of kinase and BET bromodomain inhibitors in cell killing has been reported for a variety of cancers. Using the chemical scaffold of the JAK2 inhibitor TG101348, we developed and characterized single agents which potently and simultaneously inhibit BRD4 and a specific set of oncogenic tyrosine kinases including JAK2, FLT3, RET, and ROS1. Lead compounds showed on-target inhibition in several blood cancer cell lines and were highly efficacious at inhibiting the growth of hematopoietic progenitor cells from patients with myeloproliferative neoplasm. Screening across 931 cancer cell lines revealed differential growth inhibitory potential with highest activity against bone and blood cancers and greatly enhanced activity over the single BET inhibitor JQ1. Gene drug sensitivity analyses and drug combination studies indicate synergism of BRD4 and kinase inhibition as a plausible reason for the superior potency in cell killing. Combined, our findings indicate promising potential of these agents as novel chemical probes and cancer therapeutics. Mol Cancer Ther; 16(6); 1054–67. ©2017 AACR.
Journal of Medicinal Chemistry | 2017
Jin-Yi Zhu; Rebecca Cuellar; Norbert Berndt; Hee Eun Lee; Sanne H. Olesen; Mathew P. Martin; Jeffrey T. Jensen; Gunda I. Georg; Ernst Schönbrunn
Members of the Wee family of kinases negatively regulate the cell cycle via phosphorylation of CDK1 and are considered potential drug targets. Herein, we investigated the structure-function relationship of human Wee1, Wee2, and Myt1 (PKMYT1). Purified recombinant full-length proteins and kinase domain constructs differed substantially in phosphorylation states and catalytic competency, suggesting complex mechanisms of activation. A series of crystal structures reveal unique features that distinguish Wee1 and Wee2 from Myt1 and establish the structural basis of differential inhibition by the widely used Wee1 inhibitor MK-1775. Kinome profiling and cellular studies demonstrate that, in addition to Wee1 and Wee2, MK-1775 is an equally potent inhibitor of the polo-like kinase PLK1. Several previously unrecognized inhibitors of Wee kinases were discovered and characterized. Combined, the data provide a comprehensive view on the catalytic and structural properties of Wee kinases and a framework for the rational design of novel inhibitors thereof.
Cancer Research | 2012
Roberta Pireddu; Kara D. Forinash; Nan N. Sun; Mathew P. Martin; Shen-Shu Sung; Brian M. Alexander; Jin-Yi Zhu; Wayne C. Guida; Ernst Schönbrunn; Said M. Sebti; Nicholas J. Lawrence
Rho GTPase is a small G-protein which plays a critical role in signaling pathways and controls cell growth and division. Rho-associated protein kinase (known as ROCK or Rho kinase) is a Ser/Thr protein kinase activated by GTP-bound Rho and phosphorylates target proteins involved in various signal transduction pathways. Rho and the Rho-kinase signalling pathways are implicated in cell morphology, motility, smooth muscle contraction, formation of stress fibres, focal adhesion and cell transformation. ROCKs have been subject to growing attention, having been implicated in a range of therapeutic areas including cardiovascular diseases, CNS disorders, and cancer. Further, the pharmacological inhibition of ROCKs has been suggested as a promising strategy in the prevention of cell invasion, a central event in the process of metastasis. Potent ROCK inhibitors of a new class of 1-benzyl-3-(4-pyridylthiazol-2-yl)ureas have been identified. Remarkable differences in activity were observed for ureas bearing a benzylic stereogenic center. Derivatives with hydroxy, methoxy and amino groups at the meta position of the phenyl ring give rise to the most potent inhibitors (low nM). Substitutions at the para position result in substantial loss of potency. Changes at the benzylic position are tolerated resulting in significant potency in the case of methyl and methylenehydroxy groups. X-ray crystallography was used to establish the binding mode of this class of inhibitors and provides an explanation for the observed differences of the enantiomer series. Potent inhibition of ROCK in human lung cancer cells was shown by suppression of the phosphorylation levels of the ROCK substrate MYPT-1. One of the series, RKI-1447 inhibits migration, invasion and anchorage-independent tumor growth of breast cancer cells and is discussed in an accompanying poster. 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 3904. doi:1538-7445.AM2012-3904
ACS omega | 2017
Bryce K. Allen; Saurabh Mehta; Stuart W. J. Ember; Jin-Yi Zhu; Ernst Schönbrunn; Nagi G. Ayad; Stephan C. Schürer
Computational screening is a method to prioritize small-molecule compounds based on the structural and biochemical attributes built from ligand and target information. Previously, we have developed a scalable virtual screening workflow to identify novel multitarget kinase/bromodomain inhibitors. In the current study, we identified several novel N-[3-(2-oxo-pyrrolidinyl)phenyl]-benzenesulfonamide derivatives that scored highly in our ensemble docking protocol. We quantified the binding affinity of these compounds for BRD4(BD1) biochemically and generated cocrystal structures, which were deposited in the Protein Data Bank. As the docking poses obtained in the virtual screening pipeline did not align with the experimental cocrystal structures, we evaluated the predictions of their precise binding modes by performing molecular dynamics (MD) simulations. The MD simulations closely reproduced the experimentally observed protein–ligand cocrystal binding conformations and interactions for all compounds. These results suggest a computational workflow to generate experimental-quality protein–ligand binding models, overcoming limitations of docking results due to receptor flexibility and incomplete sampling, as a useful starting point for the structure-based lead optimization of novel BRD4(BD1) inhibitors.
ChemMedChem | 2016
Sanne H. Olesen; Jin-Yi Zhu; Mathew P. Martin; Ernst Schönbrunn
Increasing evidence suggests key roles for members of the mammalian Sterile20‐like (MST) family of kinases in many aspects of biology. MST3 is a member of the STRIPAK complex, the deregulation of which has recently been associated with cancer cell migration and metastasis. Targeting MST3 with small‐molecule inhibitors may be beneficial for the treatment of certain cancers, but little information exists on the potential of kinase inhibitor scaffolds to engage with MST3. In this study we screened MST3 against a library of 277 kinase inhibitors using differential scanning fluorimetry and confirmed 14 previously unknown MST3 inhibitors by X‐ray crystallography. These compounds, of which eight are in clinical trials or FDA approved, comprise nine distinct chemical scaffolds that inhibit MST3 enzymatic activity with IC50 values between 0.003 and 23 μm. The structure–activity relationships explain the differential inhibitory activity of these compounds against MST3 and the structural basis for high binding potential, the information of which may serve as a framework for the rational design of MST3‐selective inhibitors as potential therapeutics and to interrogate the function of this enzyme in diseased cells.