Network


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

Hotspot


Dive into the research topics where Arnab K. Chatterjee is active.

Publication


Featured researches published by Arnab K. Chatterjee.


Proceedings of the National Academy of Sciences of the United States of America | 2008

In silico activity profiling reveals the mechanism of action of antimalarials discovered in a high-throughput screen

David Plouffe; Achim Brinker; Case W. McNamara; Kerstin Henson; Nobutaka Kato; Kelli Kuhen; Advait Nagle; Francisco Adrian; Jason Matzen; Paul Anderson; Tae-gyu Nam; Nathanael S. Gray; Arnab K. Chatterjee; Jeff Janes; S. Frank Yan; Richard Trager; Jeremy S. Caldwell; Peter G. Schultz; Yingyao Zhou; Elizabeth A. Winzeler

The growing resistance to current first-line antimalarial drugs represents a major health challenge. To facilitate the discovery of new antimalarials, we have implemented an efficient and robust high-throughput cell-based screen (1,536-well format) based on proliferation of Plasmodium falciparum (Pf) in erythrocytes. From a screen of ≈1.7 million compounds, we identified a diverse collection of ≈6,000 small molecules comprised of >530 distinct scaffolds, all of which show potent antimalarial activity (<1.25 μM). Most known antimalarials were identified in this screen, thus validating our approach. In addition, we identified many novel chemical scaffolds, which likely act through both known and novel pathways. We further show that in some cases the mechanism of action of these antimalarials can be determined by in silico compound activity profiling. This method uses large datasets from unrelated cellular and biochemical screens and the guilt-by-association principle to predict which cellular pathway and/or protein target is being inhibited by select compounds. In addition, the screening method has the potential to provide the malaria community with many new starting points for the development of biological probes and drugs with novel antiparasitic activities.


Science | 2011

Imaging of Plasmodium liver stages to drive next-generation antimalarial drug discovery

Stephan Meister; David Plouffe; Kelli Kuhen; Ghislain M. C. Bonamy; Tao Wu; S. Whitney Barnes; Selina Bopp; Rachel Borboa; A. Taylor Bright; Jianwei Che; Steve Cohen; Neekesh V. Dharia; Kerstin Gagaring; Montip Gettayacamin; Perry Gordon; Todd Groessl; Nobutaka Kato; Marcus C. S. Lee; Case W. McNamara; David A. Fidock; Advait Nagle; Tae-gyu Nam; Wendy Richmond; Jason Roland; Matthias Rottmann; Bin Zhou; Patrick Froissard; Richard Glynne; Dominique Mazier; Jetsumon Sattabongkot

Imidazolopiperazine compounds inhibit liver-stage malaria parasites with one oral dose in mice. Most malaria drug development focuses on parasite stages detected in red blood cells, even though, to achieve eradication, next-generation drugs active against both erythrocytic and exo-erythrocytic forms would be preferable. We applied a multifactorial approach to a set of >4000 commercially available compounds with previously demonstrated blood-stage activity (median inhibitory concentration < 1 micromolar) and identified chemical scaffolds with potent activity against both forms. From this screen, we identified an imidazolopiperazine scaffold series that was highly enriched among compounds active against Plasmodium liver stages. The orally bioavailable lead imidazolopiperazine confers complete causal prophylactic protection (15 milligrams/kilogram) in rodent models of malaria and shows potent in vivo blood-stage therapeutic activity. The open-source chemical tools resulting from our effort provide starting points for future drug discovery programs, as well as opportunities for researchers to investigate the biology of exo-erythrocytic forms.


Nature | 2013

Targeting Plasmodium PI(4)K to eliminate malaria

Case W. McNamara; Marcus C. S. Lee; Chek Shik Lim; Siau Hoi Lim; Jason Roland; Advait Nagle; Oliver Simon; Bryan K. S. Yeung; Arnab K. Chatterjee; Susan McCormack; Micah J. Manary; Anne-Marie Zeeman; Koen J. Dechering; T. R. Santha Kumar; Philipp P. Henrich; Kerstin Gagaring; Maureen Ibanez; Nobutaka Kato; Kelli Kuhen; Christoph Fischli; Matthias Rottmann; David Plouffe; Badry Bursulaya; Stephan Meister; Lucia E. Rameh; Joerg Trappe; Dorothea Haasen; Martijn Timmerman; Robert W. Sauerwein; Rossarin Suwanarusk

Achieving the goal of malaria elimination will depend on targeting Plasmodium pathways essential across all life stages. Here we identify a lipid kinase, phosphatidylinositol-4-OH kinase (PI(4)K), as the target of imidazopyrazines, a new antimalarial compound class that inhibits the intracellular development of multiple Plasmodium species at each stage of infection in the vertebrate host. Imidazopyrazines demonstrate potent preventive, therapeutic, and transmission-blocking activity in rodent malaria models, are active against blood-stage field isolates of the major human pathogens P. falciparum and P. vivax, and inhibit liver-stage hypnozoites in the simian parasite P. cynomolgi. We show that imidazopyrazines exert their effect through inhibitory interaction with the ATP-binding pocket of PI(4)K, altering the intracellular distribution of phosphatidylinositol-4-phosphate. Collectively, our data define PI(4)K as a key Plasmodium vulnerability, opening up new avenues of target-based discovery to identify drugs with an ideal activity profile for the prevention, treatment and elimination of malaria.Summary Achieving the goal of malaria elimination will depend on targeting Plasmodium pathways essential across all life stages. Here, we identify a lipid kinase, phosphatidylinositol 4-kinase (PI4K), as the target of imidazopyrazines, a novel antimalarial compound class that inhibits the intracellular development of multiple Plasmodium species at each stage of infection in the vertebrate host. Imidazopyrazines demonstrate potent preventive, therapeutic, and transmission-blocking activity in rodent malaria models, are active against blood-stage field isolates of the major human pathogens, P. falciparum and P. vivax, and inhibit liver stage hypnozoites in the simian parasite P. cynomolgi. We show that imidazopyrazines exert their effect through inhibitory interaction with the ATP-binding pocket of PI4K, altering the intracellular distribution of phosphatidylinositol 4-phosphate. Collectively, our data define PI4K as a key Plasmodium vulnerability, opening up new avenues of target-based discovery to identify drugs with an ideal activity profile for the prevention, treatment and elimination of malaria.


Nature Chemical Biology | 2008

Gene expression signatures and small-molecule compounds link a protein kinase to Plasmodium falciparum motility.

Nobutaka Kato; Tomoyo Sakata; Ghislain Breton; Karine G. Le Roch; Advait Nagle; Carsten B Andersen; Badry Bursulaya; Kerstin Henson; Jeffrey R. Johnson; Kota Arun Kumar; Felix Marr; Daniel E. Mason; Case W. McNamara; David Plouffe; Muriel Spooner; Tove Tuntland; Yingyao Zhou; Eric C. Peters; Arnab K. Chatterjee; Peter G. Schultz; Gary E. Ward; Nathanael S. Gray; Jeffrey F. Harper; Elizabeth A. Winzeler

Calcium-dependent protein kinases play a crucial role in intracellular calcium signaling in plants, some algae and protozoa. In Plasmodium falciparum, calcium-dependent protein kinase 1 (PfCDPK1) is expressed during schizogony in the erythrocytic stage as well as in the sporozoite stage. It is coexpressed with genes that encode the parasite motor complex, a cellular component required for parasite invasion of host cells, parasite motility and potentially cytokinesis. A targeted gene-disruption approach demonstrated that pfcdpk1 seems to be essential for parasite viability. An in vitro biochemical screen using recombinant PfCDPK1 against a library of 20,000 compounds resulted in the identification of a series of structurally related 2,6,9-trisubstituted purines. Compound treatment caused sudden developmental arrest at the late schizont stage in P. falciparum and a large reduction in intracellular parasites in Toxoplasma gondii, which suggests a possible role for PfCDPK1 in regulation of parasite motility during egress and invasion.


Nature Reviews Microbiology | 2013

Antimalarial drug discovery — approaches and progress towards new medicines

Erika L. Flannery; Arnab K. Chatterjee; Elizabeth A. Winzeler

Malaria elimination has recently been reinstated as a global health priority but current therapies seem to be insufficient for the task. Elimination efforts require new drug classes that alleviate symptoms, prevent transmission and provide a radical cure. To develop these next-generation medicines, public–private partnerships are funding innovative approaches to identify compounds that target multiple parasite species at multiple stages of the parasite life cycle. In this Review, we discuss the cell-, chemistry- and target-based approaches used to discover new drug candidates that are currently in clinical trials or undergoing preclinical testing.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Identification of a small molecule with activity against drug-resistant and persistent tuberculosis

Feng Wang; Dhinakaran Sambandan; Rajkumar Halder; Jianing Wang; Sarah M. Batt; Brian Weinrick; Insha Ahmad; Peng-Yu Yang; Yong Zhang; John Kim; Morad Hassani; Stanislav Huszár; Claudia Trefzer; Zhenkun Ma; Takushi Kaneko; Khisi E. Mdluli; Scott G. Franzblau; Arnab K. Chatterjee; Kai Johnson; Katarína Mikušová; Gurdyal S. Besra; Klaus Fütterer; William R. Jacobs; Peter G. Schultz

Significance The global problem of TB has worsened in recent years with the emergence of drug-resistant organisms, and new drugs are clearly needed. In a cell-based high-throughput screen, a small molecule, TCA1, was discovered that has activity against replicating and nonreplicating Mycobacterium tuberculosis. It is also efficacious in acute and chronic rodent models of TB alone or combined with frontline TB drugs. TCA1 functions by a unique mechanism, inhibiting enzymes involved in cell wall and molybdenum cofactor biosynthesis. This discovery represents a significant advance in the search for new agents to treat persistent and drug-resistant TB. A cell-based phenotypic screen for inhibitors of biofilm formation in mycobacteria identified the small molecule TCA1, which has bactericidal activity against both drug-susceptible and -resistant Mycobacterium tuberculosis (Mtb) and sterilizes Mtb in vitro combined with rifampicin or isoniazid. In addition, TCA1 has bactericidal activity against nonreplicating Mtb in vitro and is efficacious in acute and chronic Mtb infection mouse models both alone and combined with rifampicin or isoniazid. Transcriptional analysis revealed that TCA1 down-regulates genes known to be involved in Mtb persistence. Genetic and affinity-based methods identified decaprenyl-phosphoryl-β-D-ribofuranose oxidoreductase DprE1 and MoeW, enzymes involved in cell wall and molybdenum cofactor biosynthesis, respectively, as targets responsible for the activity of TCA1. These in vitro and in vivo results indicate that this compound functions by a unique mechanism and suggest that TCA1 may lead to the development of a class of antituberculosis agents.


Current Medicinal Chemistry | 2006

Inhibitors of cathepsin s

Hong Liu; David C. Tully; Robert Epple; Badry Bursulaya; Jennifer A. Williams; Arnab K. Chatterjee; Jennifer L. Harris; Jun Li

Cathepsin B is an abundant and ubiquitously expressed cysteine peptidase of the papain family. It is involved in many physiological processes, such as remodeling of the extracellular matrix (wound healing), apoptosis, and activation of thyroxine and renin. In addition to its physiological roles, cathepsin B is important in many pathological processes, such as inflammation, parasite infection and cancer, where it is highly up-regulated. In cancer patients, elevated cathepsin B activity correlates to poor therapy outcome. Therefore, it is not surprising that the use of cathepsin B inhibitors reduces both tumor cell motility and invasiveness in vitro. This review summarizes recent developments in cathepsin B inhibition. To date, numerous protein inhibitors of cathepsin B have been described, some of which are of endogenous origin and function as regulators of cathepsin B activity in the cell, such as the cystatins. In addition, some exogenous protein inhibitors of cathepsin B have been isolated from various natural sources, and the use of X-ray crystal structures of cathepsin B complexed with such protein inhibitors has resulted in the design and synthesis of many new small-molecular-weight compounds as inhibitors of cathepsin B. These synthetic compounds generally contain an electrophilic functionality that reacts with cathepsin B. In the present review, these inhibitors are divided according to their mechanisms of action, as reversible and irreversible, and then further subdivided into groups for their full descriptions.


Nature | 2016

Allosteric nanobodies reveal the dynamic range and diverse mechanisms of G-protein-coupled receptor activation

Dean P. Staus; Ryan T. Strachan; Aashish Manglik; Biswaranjan Pani; Alem W. Kahsai; Tae Hun Kim; Laura M. Wingler; Seungkirl Ahn; Arnab K. Chatterjee; Ali Masoudi; Andrew C. Kruse; Els Pardon; Jan Steyaert; William I. Weis; R. Scott Prosser; Brian K. Kobilka; Tommaso Costa; Robert J. Lefkowitz

G-protein-coupled receptors (GPCRs) modulate many physiological processes by transducing a variety of extracellular cues into intracellular responses. Ligand binding to an extracellular orthosteric pocket propagates conformational change to the receptor cytosolic region to promote binding and activation of downstream signalling effectors such as G proteins and β-arrestins. It is well known that different agonists can share the same binding pocket but evoke unique receptor conformations leading to a wide range of downstream responses (‘efficacy’). Furthermore, increasing biophysical evidence, primarily using the β2-adrenergic receptor (β2AR) as a model system, supports the existence of multiple active and inactive conformational states. However, how agonists with varying efficacy modulate these receptor states to initiate cellular responses is not well understood. Here we report stabilization of two distinct β2AR conformations using single domain camelid antibodies (nanobodies)—a previously described positive allosteric nanobody (Nb80) and a newly identified negative allosteric nanobody (Nb60). We show that Nb60 stabilizes a previously unappreciated low-affinity receptor state which corresponds to one of two inactive receptor conformations as delineated by X-ray crystallography and NMR spectroscopy. We find that the agonist isoprenaline has a 15,000-fold higher affinity for β2AR in the presence of Nb80 compared to the affinity of isoprenaline for β2AR in the presence of Nb60, highlighting the full allosteric range of a GPCR. Assessing the binding of 17 ligands of varying efficacy to the β2AR in the absence and presence of Nb60 or Nb80 reveals large ligand-specific effects that can only be explained using an allosteric model which assumes equilibrium amongst at least three receptor states. Agonists generally exert efficacy by stabilizing the active Nb80-stabilized receptor state (R80). In contrast, for a number of partial agonists, both stabilization of R80 and destabilization of the inactive, Nb60-bound state (R60) contribute to their ability to modulate receptor activation. These data demonstrate that ligands can initiate a wide range of cellular responses by differentially stabilizing multiple receptor states.


ACS Chemical Biology | 2012

A High-Throughput Screen To Identify Inhibitors of ATP Homeostasis in Non-replicating Mycobacterium tuberculosis

Puiying A. Mak; Srinivasa P. S. Rao; Mai Ping Tan; Xiuhua Lin; Jason Chyba; Joann Tay; Seow Hwee Ng; Bee Huat Tan; Joseph Cherian; Jeyaraj Duraiswamy; Pablo Bifani; Vivian Lim; Boon Heng Lee; Ngai Ling Ma; David Beer; Pamela Thayalan; Kelli Kuhen; Arnab K. Chatterjee; Frantisek Supek; Richard Glynne; Jun Zheng; Helena I. Boshoff; rd Clifton E. Barry; Thomas Dick; Kevin Pethe; Luis R. Camacho

Growing evidence suggests that the presence of a subpopulation of hypoxic non-replicating, phenotypically drug-tolerant mycobacteria is responsible for the prolonged duration of tuberculosis treatment. The discovery of new antitubercular agents active against this subpopulation may help in developing new strategies to shorten the time of tuberculosis therapy. Recently, the maintenance of a low level of bacterial respiration was shown to be a point of metabolic vulnerability in Mycobacterium tuberculosis. Here, we describe the development of a hypoxic model to identify compounds targeting mycobacterial respiratory functions and ATP homeostasis in whole mycobacteria. The model was adapted to 1,536-well plate format and successfully used to screen over 600,000 compounds. Approximately 800 compounds were confirmed to reduce intracellular ATP levels in a dose-dependent manner in Mycobacterium bovis BCG. One hundred and forty non-cytotoxic compounds with activity against hypoxic non-replicating M. tuberculosis were further validated. The resulting collection of compounds that disrupt ATP homeostasis in M. tuberculosis represents a valuable resource to decipher the biology of persistent mycobacteria.


Journal of Medicinal Chemistry | 2011

Imidazolopiperazines: hit to lead optimization of new antimalarial agents.

Tao Wu; Advait Nagle; Kelli Kuhen; Kerstin Gagaring; Rachel Borboa; Caroline Francek; Zhong Chen; David Plouffe; Anne Goh; Suresh B. Lakshminarayana; Jeanette Wu; Hui Qing Ang; Peiting Zeng; Min Low Kang; William Tan; Maria Tan; Nicole Ye; Xuena Lin; Christopher Caldwell; Jared Ek; Suzanne Skolnik; Fenghua Liu; Jianling Wang; Jonathan Chang; Chun Li; Thomas Hollenbeck; Tove Tuntland; John Isbell; Christoph Fischli; Reto Brun

Starting from a hit series from a GNF compound library collection and based on a cell-based proliferation assay of Plasmodium falciparum, a novel imidazolopiperazine scaffold was optimized. SAR for this series of compounds is discussed, focusing on optimization of cellular potency against wild-type and drug resistant parasites and improvement of physiochemical and pharmacokinetic properties. The lead compounds in this series showed good potencies in vitro and decent oral exposure levels in vivo. In a Plasmodium berghei mouse infection model, one lead compound lowered the parasitemia level by 99.4% after administration of 100 mg/kg single oral dose and prolonged mice survival by an average of 17.0 days. The lead compounds were also well-tolerated in the preliminary in vitro toxicity studies and represents an interesting lead for drug development.

Collaboration


Dive into the Arnab K. Chatterjee's collaboration.

Top Co-Authors

Avatar

David C. Tully

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Kelli Kuhen

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter G. Schultz

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert H. Grubbs

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tove Tuntland

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Case W. McNamara

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

David Plouffe

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Researchain Logo
Decentralizing Knowledge