Network


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

Hotspot


Dive into the research topics where Rajan Pragani is active.

Publication


Featured researches published by Rajan Pragani.


Journal of Biological Chemistry | 2014

Biochemical, Cellular and Biophysical Characterization of a Potent Inhibitor of Mutant Isocitrate Dehydrogenase IDH1

Mindy I. Davis; Stefan Gross; Min Shen; Kimberly Straley; Rajan Pragani; Wendy A. Lea; Janeta Popovici-Muller; Byron DeLaBarre; Erin Artin; Natasha Thorne; Douglas S. Auld; Zhuyin Li; Lenny Dang; Matthew B. Boxer; Anton Simeonov

Background: IDH1 R132H, implicated in glioblastoma and AML, produces the oncometabolite 2-HG. Results: A detailed binding mechanism of a small molecule inhibitor (ML309) is proposed. Conclusion: ML309 competes with α-KG but is uncompetitive with NADPH and rapidly and reversibly affects cellular 2-HG levels. Significance: Understanding IDH1 R132H inhibition sets the stage for targeting IDH1 R132H for the treatment of cancer. Two mutant forms (R132H and R132C) of isocitrate dehydrogenase 1 (IDH1) have been associated with a number of cancers including glioblastoma and acute myeloid leukemia. These mutations confer a neomorphic activity of 2-hydroxyglutarate (2-HG) production, and 2-HG has previously been implicated as an oncometabolite. Inhibitors of mutant IDH1 can potentially be used to treat these diseases. In this study, we investigated the mechanism of action of a newly discovered inhibitor, ML309, using biochemical, cellular, and biophysical approaches. Substrate binding and product inhibition studies helped to further elucidate the IDH1 R132H catalytic cycle. This rapidly equilibrating inhibitor is active in both biochemical and cellular assays. The (+) isomer is active (IC50 = 68 nm), whereas the (−) isomer is over 400-fold less active (IC50 = 29 μm) for IDH1 R132H inhibition. IDH1 R132C was similarly inhibited by (+)-ML309. WT IDH1 was largely unaffected by (+)-ML309 (IC50 >36 μm). Kinetic analyses combined with microscale thermophoresis and surface plasmon resonance indicate that this reversible inhibitor binds to IDH1 R132H competitively with respect to α-ketoglutarate and uncompetitively with respect to NADPH. A reaction scheme for IDH1 R132H inhibition by ML309 is proposed in which ML309 binds to IDH1 R132H after formation of the IDH1 R132H NADPH complex. ML309 was also able to inhibit 2-HG production in a glioblastoma cell line (IC50 = 250 nm) and had minimal cytotoxicity. In the presence of racemic ML309, 2-HG levels drop rapidly. This drop was sustained until 48 h, at which point the compound was washed out and 2-HG levels recovered.


PLOS ONE | 2013

A Homogeneous, High-Throughput Assay for Phosphatidylinositol 5-Phosphate 4-Kinase with a Novel, Rapid Substrate Preparation

Mindy I. Davis; Atsuo T. Sasaki; Min Shen; Brooke M. Emerling; Natasha Thorne; Sam Michael; Rajan Pragani; Matthew B. Boxer; Kazutaka Sumita; Koh Takeuchi; Douglas S. Auld; Zhuyin Li; Lewis C. Cantley; Anton Simeonov

Phosphoinositide kinases regulate diverse cellular functions and are important targets for therapeutic development for diseases, such as diabetes and cancer. Preparation of the lipid substrate is crucial for the development of a robust and miniaturizable lipid kinase assay. Enzymatic assays for phosphoinositide kinases often use lipid substrates prepared from lyophilized lipid preparations by sonication, which result in variability in the liposome size from preparation to preparation. Herein, we report a homogeneous 1536-well luciferase-coupled bioluminescence assay for PI5P4Kα. The substrate preparation is novel and allows the rapid production of a DMSO-containing substrate solution without the need for lengthy liposome preparation protocols, thus enabling the scale-up of this traditionally difficult type of assay. The Z’-factor value was greater than 0.7 for the PI5P4Kα assay, indicating its suitability for high-throughput screening applications. Tyrphostin AG-82 had been identified as an inhibitor of PI5P4Kα by assessing the degree of phospho transfer of γ-32P-ATP to PI5P; its inhibitory activity against PI5P4Kα was confirmed in the present miniaturized assay. From a pilot screen of a library of bioactive compounds, another tyrphostin, I-OMe tyrphostin AG-538 (I-OMe-AG-538), was identified as an ATP-competitive inhibitor of PI5P4Kα with an IC50 of 1 µM, affirming the suitability of the assay for inhibitor discovery campaigns. This homogeneous assay may apply to other lipid kinases and should help in the identification of leads for this class of enzymes by enabling high-throughput screening efforts.


Journal of Biological Chemistry | 2016

Small Molecule Inhibition of the Ubiquitin-specific Protease USP2 Accelerates cyclin D1 Degradation and Leads to Cell Cycle Arrest in Colorectal Cancer and Mantle Cell Lymphoma Models

Mindy I. Davis; Rajan Pragani; Jennifer T. Fox; Min Shen; Kalindi Parmar; Emily Gaudiano; Li Liu; Cordelle Tanega; Lauren McGee; Matthew D. Hall; Crystal McKnight; Paul Shinn; Henrike Nelson; Debasish Chattopadhyay; Alan D. D'Andrea; Douglas S. Auld; Larry DeLucas; Zhuyin Li; Matthew B. Boxer; Anton Simeonov

Deubiquitinases are important components of the protein degradation regulatory network. We report the discovery of ML364, a small molecule inhibitor of the deubiquitinase USP2 and its use to interrogate the biology of USP2 and its putative substrate cyclin D1. ML364 has an IC50 of 1.1 μm in a biochemical assay using an internally quenched fluorescent di-ubiquitin substrate. Direct binding of ML364 to USP2 was demonstrated using microscale thermophoresis. ML364 induced an increase in cellular cyclin D1 degradation and caused cell cycle arrest as shown in Western blottings and flow cytometry assays utilizing both Mino and HCT116 cancer cell lines. ML364, and not the inactive analog 2, was antiproliferative in cancer cell lines. Consistent with the role of cyclin D1 in DNA damage response, ML364 also caused a decrease in homologous recombination-mediated DNA repair. These effects by a small molecule inhibitor support a key role for USP2 as a regulator of cell cycle, DNA repair, and tumor cell growth.


Bioorganic & Medicinal Chemistry Letters | 2015

Structure activity relationships of human galactokinase inhibitors

Li Liu; Manshu Tang; Martin J. Walsh; Kyle R. Brimacombe; Rajan Pragani; Cordelle Tanega; Jason M. Rohde; Heather L. Baker; Elizabeth Fernandez; Burchelle Blackman; James M. Bougie; William Leister; Douglas S. Auld; Min Shen; Kent Lai; Matthew B. Boxer

Classic Galactosemia is a rare inborn error of metabolism that is caused by deficiency of galactose-1-phosphate uridyltransferase (GALT), an enzyme within the Leloir pathway that is responsible for the conversion of galactose-1-phosphate (gal-1-p) and UDP-glucose to glucose-1-phosphate and UDP-galactose. This deficiency results in elevated intracellular concentrations of its substrate, gal-1-p, and this increased concentration is believed to be the major pathogenic mechanism in Classic Galactosemia. Galactokinase (GALK) is an upstream enzyme of GALT in the Leloir pathway and is responsible for conversion of galactose and ATP to gal-1-p and ADP. Therefore, it was hypothesized that the identification of a small-molecule inhibitor of human GALK would act to prevent the accumulation of gal-1-p and offer a novel entry therapy for this disorder. Herein we describe a quantitative high-throughput screening campaign that identified a single chemotype that was optimized and validated as a GALK inhibitor.


Scientific Reports | 2017

Assessing inhibitors of mutant isocitrate dehydrogenase using a suite of pre-clinical discovery assays

Daniel J. Urban; Natalia Martínez; Mindy I. Davis; Kyle R. Brimacombe; Dorian M. Cheff; Tobie D. Lee; Mark J. Henderson; Steven A. Titus; Rajan Pragani; Jason M. Rohde; Li Liu; Yuhong Fang; Surendra Karavadhi; Pranav Shah; Olivia W. Lee; Amy Wang; Andrew L. McIver; Hongchao Zheng; Xiaodong Wang; Xin Xu; Ajit Jadhav; Anton Simeonov; Min Shen; Matthew B. Boxer; Matthew D. Hall

Isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) are key metabolic enzymes that are mutated in a variety of cancers to confer a gain-of-function activity resulting in the accumulation of an oncometabolite, D-2-hydroxyglutarate (2-HG). Accumulation of 2-HG can result in epigenetic dysregulation and a block in cellular differentiation, suggesting these mutations play a role in neoplasia. Based on its potential as a cancer target, a number of small molecule inhibitors have been developed to specifically inhibit mutant forms of IDH (mIDH1 and mIDH2). We present a comprehensive suite of in vitro preclinical drug development assays that can be used as a tool-box to identify lead compounds for mIDH drug discovery programs, as well as what we believe is the most comprehensive publically available dataset on the top mIDH inhibitors. This involved biochemical, cell-based, and tier-one ADME techniques.


Cancer Research | 2015

Abstract 4714: Targeting p53 mutant cancers through inhibition of the phosphatidylinositol-5-phosphate 4-kinases

Brooke M. Emerling; Zhiwei Yang; Ryan Loughran; T.Jonathan Yang; Jared L. Johnson; Rajan Pragani; Mindy I. Davis; Min Shen; Matthew B. Boxer; Anton Simeonov; Lewis C. Cantley

The bulk of cellular phosphatidylinositol-4,5-bisphosphate (PI-4,5-P2) is generated by the canonical pathway in which a 5-kinase converts phosphatidylinositol-4-phosphate (PI-4-P) to PI-4,5-P2. However, several years ago we discovered that PI-4, 5-P2 can also be generated at intracellular sites by a family of kinases that phosphorylate the 4 position of phosphatidylinositol-5-phosphate (PI-5-P), a lipid that was not previously known to exist in nature. These enzymes are called the type 2 phosphatidylinositol-5-phosphate 4-kinases (PI5P4Ks). To date, we have shown that a subset of breast cancers express high levels of PI5P4Kα and/or β, and have provided evidence that these kinases are essential for growth in the absence of p53 (Emerling et al., 2013). Furthermore, we showed that PI5P4Kα and β play critical roles in mediating changes in metabolism in response to cellular stress, in particular, stress that occurs in the absence of p53. Here, we disclose the discovery of potent and selective inhibitors of PI5P4K (Kis in the range of 10 to 200 nM). These novel PI5P4K inhibitors have provided us with powerful tools to further investigate the role of the PI5P4K enzymes in cancer metabolism. Through pharmacological inhibition of PI5P4K, we now have support that PI5P4K provides an alternative pathway to p53 in regard to mediating responses to metabolic and oxidative stress, thereby suggesting that the PI5P4K enzymes are essential for survival mechanisms when p53 function is lost. Above all, these inhibitors may be effective therapies not only for breast cancers with genetic aberrations in TP53, but in all cancer types with loss of p53 function. Supported by DOD Breast Research Program Breakthrough Award to B.M.E. References Emerling BM, Hurov JB, Poulogiannis G, Tsukazawa KS, Wulf G, Bell EL, Shim H, Choo-Wing R, Bellinger G, Lamia KA, Rameh LE, Sasaki A, Asara JM, Yuan X, Bullock A, Brown V, Signoretti S, and Cantley LC. (2013) Depletion of a Putatively Druggable Class of Phosphatidylinositol Kinases Inhibits Growth of p53 Null Tumors. Cell. Nov 7; 155 (4): 844-57. Citation Format: Brooke M. Emerling, Zhiwei Yang, Ryan Loughran, T.Jonathan Yang, Jared Johnson, Rajan Pragani, Mindy Davis, Min Shen, Matthew Boxer, Anton Simeonov, Lewis C. Cantley. Targeting p53 mutant cancers through inhibition of the phosphatidylinositol-5-phosphate 4-kinases. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4714. doi:10.1158/1538-7445.AM2015-4714


Cell | 2016

Breaking Cryo-EM Resolution Barriers to Facilitate Drug Discovery.

Alan Merk; Alberto Bartesaghi; Soojay Banerjee; Veronica Falconieri; Prashant Rao; Mindy I. Davis; Rajan Pragani; Matthew B. Boxer; Lesley A. Earl; Jacqueline L. S. Milne; Sriram Subramaniam


Archive | 2013

ML309: A potent inhibitor of R132H mutant IDH1 capable of reducing 2-hydroxyglutarate production in U87 MG glioblastoma cells

Mindy I. Davis; Rajan Pragani; Janeta Popovici-Muller; Stefan Gross; Natasha Thorne; Frank Salituro; Valeria Fantin; Kimberly Straley; Michael Su; Lenny Dang; Anton Simeonov; Min Shen; Matthew B. Boxer


Archive | 2017

UTILISATIONS DE DÉRIVÉS DE THIAZOLE EN TANT QU'INHIBITEURS MUTANTS D'IDH1 POUR LE TRAITEMENT DU CANCER

Matthew B. Boxer; Xiaodong Wang; Kyle R. Brimacombe; Mindy I. Davis; Yuhong Fang; Matthew D. Hall; Ajit Jadhav; Surendra Karavadhi; Li Liu; Natalia Martínez; Andrew L. McIver; Rajan Pragani; Jason M. Rohde; Anton Simeonov; Wei Zhao; Min Shen


Archive | 2016

MUTANT IDH1 INHIBITORS USEFUL FOR TREATING CANCER

Matthew B. Boxer; Jason M. Rohde; Rajan Pragani; Li Liu; Mindy I. Davis; Kyle R. Brimacombe; Min Shen; Anton Simeonov; Surendra Karavadhi; Daniel J. Urban; Ajit Jadhav; Xiaodong Wang; Andrew L. McIver

Collaboration


Dive into the Rajan Pragani's collaboration.

Top Co-Authors

Avatar

Matthew B. Boxer

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Min Shen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Mindy I. Davis

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Anton Simeonov

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Natasha Thorne

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stefan Gross

Anschutz Medical Campus

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge