Network


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

Hotspot


Dive into the research topics where Mihir D. Parikh is active.

Publication


Featured researches published by Mihir D. Parikh.


ACS Chemical Biology | 2015

Rational Targeting of Active-Site Tyrosine Residues Using Sulfonyl Fluoride Probes

Erik C. Hett; Hua Xu; Kieran F. Geoghegan; Ariamala Gopalsamy; Robert E. Kyne; Carol A. Menard; Arjun Narayanan; Mihir D. Parikh; Shenping Liu; Lee R. Roberts; Ralph P. Robinson; Michael A. Tones; Lyn H. Jones

This work describes the first rational targeting of tyrosine residues in a protein binding site by small-molecule covalent probes. Specific tyrosine residues in the active site of the mRNA-decapping scavenger enzyme DcpS were modified using reactive sulfonyl fluoride covalent inhibitors. Structure-based molecular design was used to create an alkyne-tagged probe bearing the sulfonyl fluoride warhead, thus enabling the efficient capture of the protein from a complex proteome. Use of the probe in competition experiments with a diaminoquinazoline DcpS inhibitor permitted the quantification of intracellular target occupancy. As a result, diaminoquinazoline upregulators of survival motor neuron protein that are used for the treatment of spinal muscular atrophy were confirmed as inhibitors of DcpS in human primary cells. This work illustrates the utility of sulfonyl fluoride probes designed to react with specific tyrosine residues of a protein and augments the chemical biology toolkit by these probes uses in target validation and molecular pharmacology.


Bioorganic & Medicinal Chemistry Letters | 2009

Benzothiophene inhibitors of MK2. Part 2: improvements in kinase selectivity and cell potency.

David R. Anderson; Marvin Jay Meyers; Ravi G. Kurumbail; Nicole Caspers; Gennadiy I. Poda; Scott A. Long; Betsy S. Pierce; Matthew W. Mahoney; Robert J. Mourey; Mihir D. Parikh

Optimization of kinase selectivity for a set of benzothiophene MK2 inhibitors provided analogs with potencies of less than 500 nM in a cell based assay. The selectivity of the inhibitors can be rationalized by examination of X-ray crystal structures of inhibitors bound to MK2.


Journal of Medicinal Chemistry | 2016

Identification of a Chemical Probe for Family VIII Bromodomains through Optimization of a Fragment Hit

Brian S. Gerstenberger; John David Trzupek; Cynthia Tallant; Oleg Fedorov; Panagis Filippakopoulos; Paul E. Brennan; Vita Fedele; Sarah Martin; Sarah Picaud; Catherine Rogers; Mihir D. Parikh; Alexandria P. Taylor; Brian Samas; Alison O’Mahony; Ellen Berg; Gabriel Pallares; Adam Torrey; Daniel Kelly Treiber; Ivan Samardjiev; Brian T. Nasipak; Teresita Padilla-Benavides; Qiong Wu; Anthony N. Imbalzano; Jeffrey A. Nickerson; Mark Edward Bunnage; Susanne Müller; Stefan Knapp; Dafydd R. Owen

The acetyl post-translational modification of chromatin at selected histone lysine residues is interpreted by an acetyl-lysine specific interaction with bromodomain reader modules. Here we report the discovery of the potent, acetyl-lysine-competitive, and cell active inhibitor PFI-3 that binds to certain family VIII bromodomains while displaying significant, broader bromodomain family selectivity. The high specificity of PFI-3 for family VIII was achieved through a novel bromodomain binding mode of a phenolic headgroup that led to the unusual displacement of water molecules that are generally retained by most other bromodomain inhibitors reported to date. The medicinal chemistry program that led to PFI-3 from an initial fragment screening hit is described in detail, and additional analogues with differing family VIII bromodomain selectivity profiles are also reported. We also describe the full pharmacological characterization of PFI-3 as a chemical probe, along with phenotypic data on adipocyte and myoblast cell differentiation assays.


Bioorganic & Medicinal Chemistry Letters | 2010

Structure-based drug design enables conversion of a DFG-in binding CSF-1R kinase inhibitor to a DFG-out binding mode.

Marvin Jay Meyers; Matthew James Pelc; Satwik Kamtekar; Jacqueline E. Day; Gennadiy I. Poda; Molly K. Hall; Marshall L. Michener; Beverly A. Reitz; Karl J. Mathis; Betsy S. Pierce; Mihir D. Parikh; Deborah A. Mischke; Scott A. Long; John J. Parlow; David R. Anderson; Atli Thorarensen

The work described herein demonstrates the utility of structure-based drug design (SBDD) in shifting the binding mode of an HTS hit from a DFG-in to a DFG-out binding mode resulting in a class of novel potent CSF-1R kinase inhibitors suitable for lead development.


Biochemical Journal | 2014

Selectively targeting an inactive conformation of interleukin-2-inducible T-cell kinase by allosteric inhibitors

Seungil Han; R.M Czerwinski; Nicole Caspers; David Limburg; Weidong Ding; Hong Wang; J.F Ohren; Francis Rajamohan; Thomas J. McLellan; Ray Unwalla; Chulho Choi; Mihir D. Parikh; N Seth; Jason Edmonds; Christopher Phillips; S Shakya; X Li; Spaulding; Samantha J. Hughes; Andrew Simon Cook; Colin Robinson; John Paul Mathias; I Navratilova; Quintus G. Medley; D.R Anderson; Ravi G. Kurumbail; A. Aulabaugh

ITK (interleukin-2-inducible T-cell kinase) is a critical component of signal transduction in T-cells and has a well-validated role in their proliferation, cytokine release and chemotaxis. ITK is an attractive target for the treatment of T-cell-mediated inflammatory diseases. In the present study we describe the discovery of kinase inhibitors that preferentially bind to an allosteric pocket of ITK. The novel ITK allosteric site was characterized by NMR, surface plasmon resonance, isothermal titration calorimetry, enzymology and X-ray crystallography. Initial screening hits bound to both the allosteric pocket and the ATP site. Successful lead optimization was achieved by improving the contribution of the allosteric component to the overall inhibition. NMR competition experiments demonstrated that the dual-site binders showed higher affinity for the allosteric site compared with the ATP site. Moreover, an optimized inhibitor displayed non-competitive inhibition with respect to ATP as shown by steady-state enzyme kinetics. The activity of the isolated kinase domain and auto-activation of the full-length enzyme were inhibited with similar potency. However, inhibition of the activated full-length enzyme was weaker, presumably because the allosteric site is altered when ITK becomes activated. An optimized lead showed exquisite kinome selectivity and is efficacious in human whole blood and proximal cell-based assays.


Journal of Pharmacology and Experimental Therapeutics | 2010

Biochemical, Cellular, and Anti-Inflammatory Properties of a Potent, Selective, Orally Bioavailable Benzamide Inhibitor of Rho Kinase Activity

Lakshman E. Rajagopalan; Michael S. Davies; Larry E. Kahn; Christine M. Kornmeier; Hideaki Shimada; Toni A. Steiner; Ben S. Zweifel; Jay M. Wendling; Maria A. Payne; Richard F. Loeffler; Brenda L. Case; Monica B. Norton; Mihir D. Parikh; Olga V. Nemirovskiy; Robert J. Mourey; Jaime L. Masferrer; Thomas P. Misko; Stephen A. Kolodziej

Rho kinase, is the most widely studied downstream effector of the small Rho GTPase RhoA. Two Rho kinase isoforms have been described and are frequently referred to in the literature as ROCK1and ROCK2. The RhoA–Rho kinase pathway has been implicated in the recruitment of cellular infiltrates to disease loci in a number of preclinical animal models of inflammatory disease. In this study, we used biochemical enzyme assays and a cellular target biomarker assay to define PF-4950834 [N-methyl-3-{[(4-pyridin-4-ylbenzoyl)amino]methyl}benzamide] as an ATP-competitive, selective Rho kinase inhibitor. We further used PF-4950834 to study the role of Rho kinase activation in lymphocyte and neutrophil migration in addition to the endothelial cell-mediated expression of adhesion molecules and chemokines, which are essential for leukocyte recruitment. The inhibitor blocked stromal cell-derived factor-1α-mediated chemotaxis of T lymphocytes in vitro and the synthesis of vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 in activated human endothelial cells in vitro. The secretion of chemokines interleukin-8 and monocyte chemoattractant protein-1 was also inhibited in activated endothelial cells. In addition, when dosed orally, the compound potently inhibited neutrophil migration in a carrageenan-induced acute inflammation model. In summary, we have used a pharmacologic approach to link Rho kinase activation to multiple phenotypes that can contribute to leukocyte infiltration. Inhibition of this pathway therefore could be strongly anti-inflammatory and provide therapeutic benefit in chronic inflammatory diseases.


Journal of Medicinal Chemistry | 2017

Design of Potent mRNA Decapping Scavenger Enzyme (DcpS) Inhibitors with Improved Physicochemical Properties To Investigate the Mechanism of Therapeutic Benefit in Spinal Muscular Atrophy (SMA)

Ariamala Gopalsamy; Arjun Narayanan; Shenping Liu; Mihir D. Parikh; Robert E. Kyne; Olugbeminiyi O. Fadeyi; Michael A. Tones; Jonathan J. Cherry; Joseph F. Nabhan; Gregory J. LaRosa; Donna N. Petersen; Carol A. Menard; Timothy L. Foley; Stephen Noell; Yong Ren; Paula M. Loria; Jodi Maglich-Goodwin; Haojing Rong; Lyn H. Jones

The C-5 substituted 2,4-diaminoquinazoline RG3039 (compound 1), a member of a chemical series that was identified and optimized using an SMN2 promoter screen, prolongs survival and improves motor function in a mouse model of spinal muscular atrophy (SMA). It is a potent inhibitor of the mRNA decapping scavenger enzyme (DcpS), but the mechanism whereby DcpS inhibition leads to therapeutic benefit is unclear. Compound 1 is a dibasic lipophilic molecule that is predicted to accumulate in lysosomes. To understand if the in vivo efficacy is due to DcpS inhibition or other effects resulting from the physicochemical properties of the chemotype, we undertook structure based molecular design to identify DcpS inhibitors with improved physicochemical properties. Herein we describe the design, synthesis, and in vitro pharmacological characterization of these DcpS inhibitors along with the in vivo mouse CNS PK profile of PF-DcpSi (compound 24), one of the analogs found to be efficacious in SMA mouse model.


ChemBioChem | 2016

Chemoselective Preparation of Clickable Aryl Sulfonyl Fluoride Monomers: A Toolbox of Highly Functionalized Intermediates for Chemical Biology Probe Synthesis.

Olugbeminiyi O. Fadeyi; Mihir D. Parikh; Ming Z. Chen; Robert E. Kyne; Alexandria P. Taylor; Inish O'Doherty; Stephen E. Kaiser; Sabrina Barbas; Sherry Niessen; Manli Shi; Scott Weinrich; John Charles Kath; Lyn H. Jones; Ralph P. Robinson

Sulfonyl fluoride (SF)‐based activity probes have become important tools in chemical biology. Herein, exploiting the relative chemical stability of SF to carry out a number of unprecedented SF‐sparing functional group manipulations, we report the chemoselective synthesis of a toolbox of highly functionalized aryl SF monomers that we used to quickly prepare SF chemical biology probes. In addition to SF, the monomers bear an embedded click handle (a terminal alkyne that can perform copper(I)‐mediated azide–alkyne cycloaddition). The monomers can be used either as fragments to prepare clickable SF analogues of drugs (biologically active compounds) bearing an aryl ring or, alternatively, attached to drugs as minimalist clickable aryl SF substituents.


Molecular BioSystems | 2015

A library approach to rapidly discover photoaffinity probes of the mRNA decapping scavenger enzyme DcpS

Hua Xu; Erik C. Hett; Ariamala Gopalsamy; Mihir D. Parikh; Kieran F. Geoghegan; Robert E. Kyne; Carol A. Menard; Arjun Narayanan; Ralph P. Robinson; Douglas S. Johnson; Michael A. Tones; Lyn H. Jones


Bioorganic & Medicinal Chemistry | 2007

Discovery of +(2-{4-[2-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)ethoxy]phenyl}-cyclopropyl)acetic acid as potent and selective αvβ3 inhibitor: Design, synthesis, and optimization

Srinivasan R. Nagarajan; Hwang-Fun Lu; Alan F. Gasiecki; Ish Kurmar Khanna; Mihir D. Parikh; Bipinchandra Nanubhai Desai; Thomas E. Rogers; Michael Clare; Barbara B. Chen; Mark A. Russell; Jeffery L. Keene; Tiffany Duffin; V. Wayne Engleman; Mary Beth Finn; Sandra K. Freeman; Jon A. Klover; G. Alan Nickols; Maureen A. Nickols; Kristen E. Shannon; Christina A. Steininger; William F. Westlin; Marisa M. Westlin; Melanie L. Williams

Researchain Logo
Decentralizing Knowledge