Network


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

Hotspot


Dive into the research topics where Ye Che is active.

Publication


Featured researches published by Ye Che.


Proteins | 2012

Thermodynamic analysis of water molecules at the surface of proteins and applications to binding site prediction and characterization

Thijs Beuming; Ye Che; Robert Abel; Byungchan Kim; Veerabahu Shanmugasundaram; Woody Sherman

Water plays an essential role in determining the structure and function of all biological systems. Recent methodological advances allow for an accurate and efficient estimation of the thermodynamic properties of water molecules at the surface of proteins. In this work, we characterize these thermodynamic properties and relate them to various structural and functional characteristics of the protein. We find that high‐energy hydration sites often exist near protein motifs typically characterized as hydrophilic, such as backbone amide groups. We also find that waters around alpha helices and beta sheets tend to be less stable than waters around loops. Furthermore, we find no significant correlation between the hydration site‐free energy and the solvent accessible surface area of the site. In addition, we find that the distribution of high‐energy hydration sites on the protein surface can be used to identify the location of binding sites and that binding sites of druggable targets tend to have a greater density of thermodynamically unstable hydration sites. Using this information, we characterize the FKBP12 protein and show good agreement between fragment screening hit rates from NMR spectroscopy and hydration site energetics. Finally, we show that water molecules observed in crystal structures are less stable on average than bulk water as a consequence of the high degree of spatial localization, thereby resulting in a significant loss in entropy. These findings should help to better understand the characteristics of waters at the surface of proteins and are expected to lead to insights that can guide structure‐based drug design efforts. Proteins 2011.


Journal of Medicinal Chemistry | 2014

Chemical and computational methods for the characterization of covalent reactive groups for the prospective design of irreversible inhibitors.

Mark Edward Flanagan; Joseph A. Abramite; Dennis P. Anderson; Ann Aulabaugh; Upendra P. Dahal; Adam M. Gilbert; Chao Li; Justin Ian Montgomery; Stacey R. Oppenheimer; Tim Ryder; Brandon P. Schuff; Daniel P. Uccello; Gregory S. Walker; Yan Wu; Matthew Frank Brown; Jinshan M. Chen; Matthew Merrill Hayward; Mark C. Noe; R. Scott Obach; Laurence Philippe; Veerabahu Shanmugasundaram; Michael J. Shapiro; Jeremy T. Starr; Justin G. Stroh; Ye Che

Interest in drugs that covalently modify their target is driven by the desire for enhanced efficacy that can result from the silencing of enzymatic activity until protein resynthesis can occur, along with the potential for increased selectivity by targeting uniquely positioned nucleophilic residues in the protein. However, covalent approaches carry additional risk for toxicities or hypersensitivity reactions that can result from covalent modification of unintended targets. Here we describe methods for measuring the reactivity of covalent reactive groups (CRGs) with a biologically relevant nucleophile, glutathione (GSH), along with kinetic data for a broad array of electrophiles. We also describe a computational method for predicting electrophilic reactivity, which taken together can be applied to the prospective design of thiol-reactive covalent inhibitors.


Journal of Medicinal Chemistry | 2012

Potent Inhibitors of LpxC for the Treatment of Gram-Negative Infections

Matthew Frank Brown; Usa Reilly; Joseph A. Abramite; Robert M. Oliver; Rose Barham; Ye Che; Jinshan Michael Chen; Elizabeth M. Collantes; Seung Won Chung; Charlene R. Desbonnet; Jonathan L. Doty; Matthew Doroski; Juntyma J. Engtrakul; Thomas M. Harris; Michael D. Huband; John D. Knafels; Karen L. Leach; Shenping Liu; Anthony Marfat; Andrea Marra; Eric McElroy; Michael Melnick; Carol A. Menard; Justin Ian Montgomery; Lisa Mullins; Mark C. Noe; John P. O’Donnell; Joseph Penzien; Mark Stephen Plummer; Loren M. Price

In this paper, we present the synthesis and SAR as well as selectivity, pharmacokinetic, and infection model data for representative analogues of a novel series of potent antibacterial LpxC inhibitors represented by hydroxamic acid.


Journal of Medicinal Chemistry | 2012

Pyridone Methylsulfone Hydroxamate LpxC Inhibitors for the Treatment of Serious Gram-Negative Infections

Justin Ian Montgomery; Matthew Frank Brown; Usa Reilly; Loren M. Price; Joseph A. Abramite; Rose Barham; Ye Che; Jinshan Michael Chen; Seung Won Chung; E.M Collantes; Charlene R. Desbonnet; M Doroski; Jonathan L. Doty; J.J Engtrakul; Thomas M. Harris; Michael D. Huband; John D. Knafels; Karen L. Leach; Shenping Liu; Anthony Marfat; Laura A. McAllister; Eric McElroy; Carol A. Menard; Mark J. Mitton-Fry; Lisa Mullins; Mark C. Noe; J O'Donnell; Robert M. Oliver; Joseph Penzien; Mark Stephen Plummer

The synthesis and biological activity of a new series of LpxC inhibitors represented by pyridone methylsulfone hydroxamate 2a is presented. Members of this series have improved solubility and free fraction when compared to compounds in the previously described biphenyl methylsulfone hydroxamate series, and they maintain superior Gram-negative antibacterial activity to comparator agents.


Journal of Medicinal Chemistry | 2014

Siderophore Receptor-Mediated Uptake of Lactivicin Analogues in Gram-Negative Bacteria

Jeremy T. Starr; Matthew Frank Brown; Lisa M. Aschenbrenner; Nicole Caspers; Ye Che; Brian S. Gerstenberger; Michael D. Huband; John D. Knafels; M. Megan Lemmon; Chao Li; Sandra P. McCurdy; Eric McElroy; Mark R. Rauckhorst; Andrew P. Tomaras; Jennifer A. Young; Richard P. Zaniewski; Veerabahu Shanmugasundaram; Seungil Han

Multidrug-resistant Gram-negative pathogens are an emerging threat to human health, and addressing this challenge will require development of new antibacterial agents. This can be achieved through an improved molecular understanding of drug-target interactions combined with enhanced delivery of these agents to the site of action. Herein we describe the first application of siderophore receptor-mediated drug uptake of lactivicin analogues as a strategy that enables the development of novel antibacterial agents against clinically relevant Gram-negative bacteria. We report the first crystal structures of several sideromimic conjugated compounds bound to penicillin binding proteins PBP3 and PBP1a from Pseudomonas aeruginosa and characterize the reactivity of lactivicin and β-lactam core structures. Results from drug sensitivity studies with β-lactamase enzymes are presented, as well as a structure-based hypothesis to reduce susceptibility to this enzyme class. Finally, mechanistic studies demonstrating that sideromimic modification alters the drug uptake process are discussed.


Bioorganic & Medicinal Chemistry Letters | 2012

Heterocyclic methylsulfone hydroxamic acid LpxC inhibitors as Gram-negative antibacterial agents

Laura A. McAllister; Justin Ian Montgomery; Joseph A. Abramite; Usa Reilly; Matthew Frank Brown; Jinshan M. Chen; Rose Barham; Ye Che; Seung Won Chung; Carol A. Menard; Mark J. Mitton-Fry; Lisa Mullins; Mark C. Noe; John P. O’Donnell; Robert M. Oliver; Joseph Penzien; Mark Stephen Plummer; Loren M. Price; Veerabahu Shanmugasundaram; Andrew P. Tomaras; Daniel P. Uccello

The synthesis and antibacterial activity of heterocyclic methylsulfone hydroxamates is presented. Compounds in this series are potent inhibitors of the LpxC enzyme, a key enzyme involved in the production of lipopolysaccharide (LPS) found in the outer membrane of Gram-negative bacteria. SAR evaluation of compounds in this series revealed analogs with potent antibacterial activity against challenging Gram-negative species such as Pseudomonas aeruginosa and Klebsiella pneumoniae.


Journal of Medicinal Chemistry | 2017

Design of a Janus Kinase 3 (JAK3) Specific Inhibitor 1-((2S,5R)-5-((7H-Pyrrolo[2,3-d]pyrimidin-4-yl)amino)-2-methylpiperidin-1-yl)prop-2-en-1-one (PF-06651600) Allowing for the Interrogation of JAK3 Signaling in Humans

Atli Thorarensen; Martin E. Dowty; Mary Ellen Banker; Brian Juba; Jason Jussif; Tsung Lin; Fabien Vincent; Robert M. Czerwinski; Agustin Casimiro-Garcia; Ray Unwalla; John I. Trujillo; Sidney Xi Liang; Paul Balbo; Ye Che; Adam M. Gilbert; Matthew Frank Brown; Matthew Merrill Hayward; Justin Ian Montgomery; Louis Leung; Xin Yang; Sarah Soucy; Martin Hegen; Jotham Wadsworth Coe; Jonathan Langille; Felix Vajdos; Jill Chrencik; Jean-Baptiste Telliez

Significant work has been dedicated to the discovery of JAK kinase inhibitors resulting in several compounds entering clinical development and two FDA approved NMEs. However, despite significant effort during the past 2 decades, identification of highly selective JAK3 inhibitors has eluded the scientific community. A significant effort within our research organization has resulted in the identification of the first orally active JAK3 specific inhibitor, which achieves JAK isoform specificity through covalent interaction with a unique JAK3 residue Cys-909. The relatively rapid resynthesis rate of the JAK3 enzyme presented a unique challenge in the design of covalent inhibitors with appropriate pharmacodynamics properties coupled with limited unwanted off-target reactivity. This effort resulted in the identification of 11 (PF-06651600), a potent and low clearance compound with demonstrated in vivo efficacy. The favorable efficacy and safety profile of this JAK3-specific inhibitor 11 led to its evaluation in several human clinical studies.


Angewandte Chemie | 2016

Discovery of a Highly Selective Glycogen Synthase Kinase-3 Inhibitor (PF-04802367) That Modulates Tau Phosphorylation in the Brain: Translation for PET Neuroimaging.

Steven H. Liang; Jinshan Michael Chen; Marc D. Normandin; Jeanne S. Chang; George Chang; Christine Taylor; Patrick Trapa; Mark Stephen Plummer; Kimberly Suzanne Para; Edward L. Conn; Lori L. Lopresti-Morrow; Lorraine Lanyon; James M. Cook; Karl E.G. Richter; Charlie E Nolan; Joel B. Schachter; Fouad Janat; Ye Che; Veerabahu Shanmugasundaram; Bruce Allen Lefker; Bradley E. Enerson; E. Livni; Lu Wang; Nicolas Guehl; Debasis Patnaik; Florence F. Wagner; Roy H. Perlis; Edward B. Holson; Stephen J. Haggarty; Georges El Fakhri

Glycogen synthase kinase-3 (GSK-3) regulates multiple cellular processes in diabetes, oncology, and neurology. N-(3-(1H-1,2,4-triazol-1-yl)propyl)-5-(3-chloro-4-methoxyphenyl)oxazole-4-carboxamide (PF-04802367 or PF-367) has been identified as a highly potent inhibitor, which is among the most selective antagonists of GSK-3 to date. Its efficacy was demonstrated in modulation of tau phosphorylation in vitro and in vivo. Whereas the kinetics of PF-367 binding in brain tissues are too fast for an effective therapeutic agent, the pharmacokinetic profile of PF-367 is ideal for discovery of radiopharmaceuticals for GSK-3 in the central nervous system. A (11) C-isotopologue of PF-367 was synthesized and preliminary PET imaging studies in non-human primates confirmed that we have overcome the two major obstacles for imaging GSK-3, namely, reasonable brain permeability and displaceable binding.


ACS Chemical Biology | 2016

Discovery of a Selective Covalent Inhibitor of Lysophospholipase-like 1 (LYPLAL1) as a Tool to Evaluate the Role of this Serine Hydrolase in Metabolism.

Kay Ahn; Markus Boehm; Matthew Frank Brown; Jessica Calloway; Ye Che; Jinshan Chen; Kimberly F. Fennell; Kieran F. Geoghegan; Adam M. Gilbert; Jemy A. Gutierrez; Amit S. Kalgutkar; Adhiraj Lanba; Chris Limberakis; Thomas V. Magee; Inish O’Doherty; Robert M. Oliver; Brandon Pabst; Jayvardhan Pandit; Kevin D. Parris; Jeffrey A. Pfefferkorn; Timothy P. Rolph; Rushi Patel; Brandon P. Schuff; Veerabahu Shanmugasundaram; Jeremy T. Starr; Alison H. Varghese; Nicholas B. Vera; Cecile Vernochet; Jiangli Yan

Lysophospholipase-like 1 (LYPLAL1) is an uncharacterized metabolic serine hydrolase. Human genome-wide association studies link variants of the gene encoding this enzyme to fat distribution, waist-to-hip ratio, and nonalcoholic fatty liver disease. We describe the discovery of potent and selective covalent small-molecule inhibitors of LYPLAL1 and their use to investigate its role in hepatic metabolism. In hepatocytes, selective inhibition of LYPLAL1 increased glucose production supporting the inference that LYPLAL1 is a significant actor in hepatic metabolism. The results provide an example of how a selective chemical tool can contribute to evaluating a hypothetical target for therapeutic intervention, even in the absence of complete biochemical characterization.


Bioorganic & Medicinal Chemistry Letters | 2018

Inducing protein-protein interactions with molecular glues

Ye Che; Adam M. Gilbert; Veerabahu Shanmugasundaram; Mark C. Noe

The drugable proteome is limited by the number of functional binding sites that can bind small molecules and respond with a therapeutic effect. Orthosteric and allosteric modulators of enzyme function or receptor signaling are well-established mechanisms of drug action. Drugs that perturb protein-protein interactions have only recently been launched. This approach is more difficult due to the extensive contact surfaces that must be perturbed antagonistically. Compounds that promote novel protein-protein interactions promise to dramatically expand opportunities for therapeutic intervention. This approach is precedented with natural products (rapamycin, FK506, sanglifehrin A), synthetic small molecules (thalidomide and IMiD derivatives) and indisulam analogues.

Collaboration


Dive into the Ye Che's collaboration.

Researchain Logo
Decentralizing Knowledge