Network


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

Hotspot


Dive into the research topics where Kevin D. Parris is active.

Publication


Featured researches published by Kevin D. Parris.


Structure | 2003

Catalytically active MAP KAP kinase 2 structures in complex with staurosporine and ADP reveal differences with the autoinhibited enzyme

Kathryn W. Underwood; Kevin D. Parris; Elizabeth Federico; Lidia Mosyak; Robert M. Czerwinski; Tania Shane; Meggin Taylor; Kristine Svenson; Yan Liu; Chu-Lai Hsiao; Scott Wolfrom; Michelle Maguire; Karl Malakian; Jean-Baptiste Telliez; Lih-Ling Lin; Ronald W. Kriz; Jasbir Seehra; William S. Somers; Mark L. Stahl

MAP KAP kinase 2 (MK2), a Ser/Thr kinase, plays a crucial role in the inflammatory process. We have determined the crystal structures of a catalytically active C-terminal deletion form of human MK2, residues 41-364, in complex with staurosporine at 2.7 A and with ADP at 3.2 A, revealing overall structural similarity with other Ser/Thr kinases. Kinetic analysis reveals that the K(m) for ATP is very similar for MK2 41-364 and p38-activated MK2 41-400. Conversely, the catalytic rate and binding for peptide substrate are dramatically reduced in MK2 41-364. However, phosphorylation of MK2 41-364 by p38 restores the V(max) and K(m) for peptide substrate to values comparable to those seen in p38-activated MK2 41-400, suggesting a mechanism for regulation of enzyme activity.


Journal of Molecular Biology | 2013

Three-Dimensional Structure and Biophysical Characterization of Staphylococcus aureus Cell Surface Antigen–Manganese Transporter MntC

Alexey Gribenko; Lidia Mosyak; Sharmistha Ghosh; Kevin D. Parris; Kristine Svenson; Justin Keith Moran; Ling Chu; Sheng Li; Tong Liu; Virgil L. Woods; Kathrin U. Jansen; Bruce A. Green; Annaliesa S. Anderson; Yury V. Matsuka

MntC is a metal-binding protein component of the Mn²⁺-specific mntABC transporter from the pathogen Staphylococcus aureus. The protein is expressed during the early stages of infection and was proven to be effective at reducing both S. aureus and Staphylococcus epidermidis infections in a murine animal model when used as a vaccine antigen. MntC is currently being tested in human clinical trials as a component of a multiantigen vaccine for the prevention of S. aureus infections. To better understand the biological function of MntC, we are providing structural and biophysical characterization of the protein in this work. The three-dimensional structure of the protein was solved by X-ray crystallography at 2.2Å resolution and suggests two potential metal binding modes, which may lead to reversible as well as irreversible metal binding. Precise Mn²⁺-binding affinity of the protein was determined from the isothermal titration calorimetry experiments using a competition approach. Differential scanning calorimetry experiments confirmed that divalent metals can indeed bind to MntC reversibly as well as irreversibly. Finally, Mn²⁺-induced structural and dynamics changes have been characterized using spectroscopic methods and deuterium-hydrogen exchange mass spectroscopy. Results of the experiments show that these changes are minimal and are largely restricted to the structural elements involved in metal coordination. Therefore, it is unlikely that antibody binding to this antigen will be affected by the occupancy of the metal-binding site by Mn²⁺.


Journal of Medicinal Chemistry | 2015

Utilizing Structures of CYP2D6 and BACE1 Complexes To Reduce Risk of Drug-Drug Interactions with a Novel Series of Centrally Efficacious BACE1 Inhibitors.

Michael Aaron Brodney; Elizabeth Mary Beck; Christopher Ryan Butler; Gabriela Barreiro; Eric F. Johnson; David Riddell; Kevin D. Parris; Charles E. Nolan; Ying Fan; Kevin Atchison; Cathleen Gonzales; Ashley Robshaw; Shawn D. Doran; Mark W. Bundesmann; Leanne M. Buzon; Jason K. Dutra; Kevin E. Henegar; Erik LaChapelle; Xinjun Hou; Bruce N. Rogers; Jayvardhan Pandit; Ricardo Lira; Luis Martinez-Alsina; Peter Mikochik; John C. Murray; Kevin Ogilvie; Loren Price; Subas M. Sakya; Aijia Yu; Yong Zhang

In recent years, the first generation of β-secretase (BACE1) inhibitors advanced into clinical development for the treatment of Alzheimer’s disease (AD). However, the alignment of drug-like properties and selectivity remains a major challenge. Herein, we describe the discovery of a novel class of potent, low clearance, CNS penetrant BACE1 inhibitors represented by thioamidine 5. Further profiling suggested that a high fraction of the metabolism (>95%) was due to CYP2D6, increasing the potential risk for victim-based drug–drug interactions (DDI) and variable exposure in the clinic due to the polymorphic nature of this enzyme. To guide future design, we solved crystal structures of CYP2D6 complexes with substrate 5 and its corresponding metabolic product pyrazole 6, which provided insight into the binding mode and movements between substrate/inhibitor complexes. Guided by the BACE1 and CYP2D6 crystal structures, we designed and synthesized analogues with reduced risk for DDI, central efficacy, and improved hERG therapeutic margins.


Bioorganic & Medicinal Chemistry | 2009

Identification and SAR of squarate inhibitors of mitogen activated protein kinase-activated protein kinase 2 (MK-2)

Frank Lovering; Steve Kirincich; Weiheng Wang; Kerry Combs; Lynn Resnick; Joan Eileen Sabalski; John A. Butera; Julie Liu; Kevin D. Parris; Jean-Baptiste Telliez

A novel series of inhibitors for mitogen activated protein kinase-activated protein kinase 2 (MK-2) are reported. These squarate based inhibitors were identified via a high-throughput screen. An MK2 co-structure with the starting ligand was obtained and a structure based approach was followed to optimize potency and selectivity.


PLOS Pathogens | 2016

High Resolution Mapping of Bactericidal Monoclonal Antibody Binding Epitopes on Staphylococcus aureus Antigen MntC.

Alexey Gribenko; Kevin D. Parris; Lidia Mosyak; Sheng Li; Luke D. Handke; Julio Cesar Hawkins; Elena Severina; Yury V. Matsuka; Annaliesa S. Anderson

The Staphylococcus aureus manganese transporter protein MntC is under investigation as a component of a prophylactic S.aureus vaccine. Passive immunization with monoclonal antibodies mAB 305-78-7 and mAB 305-101-8 produced using MntC was shown to significantly reduce S. aureus burden in an infant rat model of infection. Earlier interference mapping suggested that a total of 23 monoclonal antibodies generated against MntC could be subdivided into three interference groups, representing three independent immunogenic regions. In the current work binding epitopes for selected representatives of each of these interference groups (mAB 305-72-5 – group 1, mAB 305-78-7 – group 2, and mAB 305-101-8 – group 3) were mapped using Hydrogen-Deuterium Exchange Mass Spectrometry (DXMS). All of the identified epitopes are discontinuous, with binding surface formed by structural elements that are separated within the primary sequence of the protein but adjacent in the context of the three-dimensional structure. The approach was validated by co-crystallizing the Fab fragment of one of the antibodies (mAB 305-78-7) with MntC and solving the three-dimensional structure of the complex. X-ray results themselves and localization of the mAB 305-78-7 epitope were further validated using antibody binding experiments with MntC variants containing substitutions of key amino acid residues. These results provided insight into the antigenic properties of MntC and how these properties may play a role in protecting the hostagainst S. aureus infection by preventing the capture and transport of Mn2+, a key element that the pathogen uses to evade host immunity.


Journal of Medicinal Chemistry | 2017

Aminomethyl-Derived Beta Secretase (BACE1) Inhibitors: Engaging Gly230 without an Anilide Functionality.

Christopher Ryan Butler; Kevin Ogilvie; Luis Martinez-Alsina; Gabriela Barreiro; Elizabeth Mary Beck; Charles E. Nolan; Kevin Atchison; Eric Benvenuti; Leanne M. Buzon; Shawn D. Doran; Cathleen Gonzales; Christopher John Helal; Xinjun Hou; Mei-Hui Hsu; Eric F. Johnson; Kimberly Lapham; Lorraine Lanyon; Kevin D. Parris; Brian T. O’Neill; David Riddell; Ashley Robshaw; Felix Vajdos; Michael Aaron Brodney

A growing subset of β-secretase (BACE1) inhibitors for the treatment of Alzheimer’s disease (AD) utilizes an anilide chemotype that engages a key residue (Gly230) in the BACE1 binding site. Although the anilide moiety affords excellent potency, it simultaneously introduces a third hydrogen bond donor that limits brain availability and provides a potential metabolic site leading to the formation of an aniline, a structural motif of prospective safety concern. We report herein an alternative aminomethyl linker that delivers similar potency and improved brain penetration relative to the amide moiety. Optimization of this series identified analogues with an excellent balance of ADME properties and potency; however, potential drug–drug interactions (DDI) were predicted based on CYP 2D6 affinities. Generation and analysis of key BACE1 and CYP 2D6 crystal structures identified strategies to obviate the DDI liability, leading to compound 16, which exhibits robust in vivo efficacy as a BACE1 inhibitor.


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.


Journal of Medicinal Chemistry | 2017

Discovery of Fragment-Derived Small Molecules for in Vivo Inhibition of Ketohexokinase (KHK)

Kim Huard; Kay Ahn; Paul Amor; David A. Beebe; Kris A. Borzilleri; Boris A. Chrunyk; Steven B. Coffey; Yang Cong; Edward L. Conn; Jeffrey S. Culp; Matthew S. Dowling; Matthew Gorgoglione; Jemy A. Gutierrez; John D. Knafels; Erik LaChapelle; Jayvardhan Pandit; Kevin D. Parris; Sylvie Perez; Jeffrey A. Pfefferkorn; David A. Price; Brian Raymer; Trenton T. Ross; Andre Shavnya; Aaron Smith; Timothy A. Subashi; Gregory Tesz; Benjamin A. Thuma; Meihua Tu; John D. Weaver; Yan Weng

Increased fructose consumption and its subsequent metabolism have been implicated in hepatic steatosis, dyslipidemia, obesity, and insulin resistance in humans. Since ketohexokinase (KHK) is the principal enzyme responsible for fructose metabolism, identification of a selective KHK inhibitor may help to further elucidate the effect of KHK inhibition on these metabolic disorders. Until now, studies on KHK inhibition with small molecules have been limited due to the lack of viable in vivo pharmacological tools. Herein we report the discovery of 12, a selective KHK inhibitor with potency and properties suitable for evaluating KHK inhibition in rat models. Key structural features interacting with KHK were discovered through fragment-based screening and subsequent optimization using structure-based drug design, and parallel medicinal chemistry led to the identification of pyridine 12.


Structure | 2000

Crystal structures of substrate binding to Bacillus subtilis holo-(acyl carrier protein) synthase reveal a novel trimeric arrangement of molecules resulting in three active sites.

Kevin D. Parris; Laura Lin; Amy Tam; Rebecca Mathew; Jeffrey Hixon; Mark Stahl; Christian Fritz; Jasbir Seehra; William S. Somers


Archive | 2005

Antibodies against human interleukin-13 and uses therefor

Marion T. Kasaian; Lioudmila Tchistiakova; Geertruida M. Veldman; Kimberly Marquette; Xiang-Yang Tan; Debra D. Donaldson; Laura Lin; Tania Shane; Amy Tam; Eric Feyfant; Nancy Wood; Lori Fitz; Angela Widom; Kevin D. Parris; Samuel J. Goldman; José W. Saldanha

Collaboration


Dive into the Kevin D. Parris's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lidia Mosyak

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

John J. Dumas

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Debra D. Donaldson

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge