Network


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

Hotspot


Dive into the research topics where Kelli Kuhen is active.

Publication


Featured researches published by Kelli Kuhen.


Cell | 2008

Global analysis of host-pathogen interactions that regulate early stage HIV-1 replication

Ronny König; Yingyao Zhou; Daniel Elleder; Tracy L. Diamond; Ghislain M. C. Bonamy; Jeffrey T. Irelan; Chih-yuan Chiang; Buu P. Tu; Paul D. De Jesus; Caroline E. Lilley; Shannon Seidel; Amanda M. Opaluch; Jeremy S. Caldwell; Matthew D. Weitzman; Kelli Kuhen; Sourav Bandyopadhyay; Trey Ideker; Anthony P. Orth; Loren Miraglia; Frederic D. Bushman; John A. T. Young; Sumit K. Chanda

Human Immunodeficiency Viruses (HIV-1 and HIV-2) rely upon host-encoded proteins to facilitate their replication. Here, we combined genome-wide siRNA analyses with interrogation of human interactome databases to assemble a host-pathogen biochemical network containing 213 confirmed host cellular factors and 11 HIV-1-encoded proteins. Protein complexes that regulate ubiquitin conjugation, proteolysis, DNA-damage response, and RNA splicing were identified as important modulators of early-stage HIV-1 infection. Additionally, over 40 new factors were shown to specifically influence the initiation and/or kinetics of HIV-1 DNA synthesis, including cytoskeletal regulatory proteins, modulators of posttranslational modification, and nucleic acid-binding proteins. Finally, 15 proteins with diverse functional roles, including nuclear transport, prostaglandin synthesis, ubiquitination, and transcription, were found to influence nuclear import or viral DNA integration. Taken together, the multiscale approach described here has uncovered multiprotein virus-host interactions that likely act in concert to facilitate the early steps of HIV-1 infection.


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.


Journal of Medicinal Chemistry | 2010

Spirotetrahydro β-carbolines (spiroindolones): a new class of potent and orally efficacious compounds for the treatment of malaria.

Bryan K. S. Yeung; Bin Zou; Matthias Rottmann; Suresh B. Lakshminarayana; Shi Hua Ang; Seh Yong Leong; Jocelyn Tan; Josephine Wong; Sonja Keller-Maerki; Christoph Fischli; Anne Goh; Esther K. Schmitt; Philipp Krastel; Eric Francotte; Kelli Kuhen; David Plouffe; Kerstin Henson; Trixie Wagner; Elizabeth A. Winzeler; Frank Petersen; Reto Brun; Véronique Dartois; Thierry T. Diagana; Thomas H. Keller

The antiplasmodial activity of a series of spirotetrahydro β-carbolines is described. Racemic spiroazepineindole (1) was identified from a phenotypic screen on wild type Plasmodium falciparum with an in vitro IC50 of 90 nM. Structure−activity relationships for the optimization of 1 to compound 20a (IC50 = 0.2 nM) including the identification of the active 1R,3S enantiomer and elimination of metabolic liabilities is presented. Improvement of the pharmacokinetic profile of the series translated to exceptional oral efficacy in the P. berghei infected malaria mouse model where full cure was achieved in four of five mice with three daily doses of 30 mg/kg.


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.


Bioorganic & Medicinal Chemistry Letters | 2006

Design, synthesis, and biological evaluations of novel oxindoles as HIV-1 non-nucleoside reverse transcriptase inhibitors. Part 2.

Tao Jiang; Kelli Kuhen; Karen Wolff; Hong Yin; Kimberly Bieza; Jeremy S. Caldwell; Badry Bursulaya; Tove Tuntland; Kanyin Zhang; Donald S. Karanewsky; Yun He

A series of heterocycle-containing oxindoles was synthesized and their HIV antiviral activities were assessed. Some of these analogs exhibited potent inhibitory activities against both wild-type virus and a number of drug-resistant mutant viruses. In addition, oxindole 9z also showed promising pharmacokinetics.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Whole-genome sequencing and microarray analysis of ex vivo Plasmodium vivax reveal selective pressure on putative drug resistance genes

Neekesh V. Dharia; A. Taylor Bright; Scott J. Westenberger; S. Whitney Barnes; Serge Batalov; Kelli Kuhen; Rachel Borboa; Colleen M. McClean; Joseph M. Vinetz; Victor Neyra; Alejandro Llanos-Cuentas; John W. Barnwell; John R. Walker; Elizabeth A. Winzeler

Plasmodium vivax causes 25–40% of malaria cases worldwide, yet research on this human malaria parasite has been neglected. Nevertheless, the recent publication of the P. vivax reference genome now allows genomics and systems biology approaches to be applied to this pathogen. We show here that whole-genome analysis of the parasite can be achieved directly from ex vivo-isolated parasites, without the need for in vitro propagation. A single isolate of P. vivax obtained from a febrile patient with clinical malaria from Peru was subjected to whole-genome sequencing (30× coverage). This analysis revealed over 18,261 single-nucleotide polymorphisms (SNPs), 6,257 of which were further validated using a tiling microarray. Within core chromosomal genes we find that one SNP per every 985 bases of coding sequence distinguishes this recent Peruvian isolate, designated IQ07, from the reference Salvador I strain obtained in 1972. This full-genome sequence of an uncultured P. vivax isolate shows that the same regions with low numbers of aligned sequencing reads are also highly variable by genomic microarray analysis. Finally, we show that the genes containing the largest ratio of nonsynonymous-to-synonymous SNPs include two AP2 transcription factors and the P. vivax multidrug resistance-associated protein (PvMRP1), an ABC transporter shown to be associated with quinoline and antifolate tolerance in Plasmodium falciparum. This analysis provides a data set for comparative analysis with important potential for identifying markers for global parasite diversity and drug resistance mapping studies.


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.


Science Translational Medicine | 2013

Indolcarboxamide Is a Preclinical Candidate for Treating Multidrug-Resistant Tuberculosis

Srinivasa P. S. Rao; Suresh B. Lakshminarayana; Ravinder Reddy Kondreddi; Maxime Herve; Luis R. Camacho; sarath Kalapala; Jan Jiricek; Ng L. Ma; Bee Huat Tan; Seow H. Ng; Mahesh Nanjundappa; Sindhu Ravindran; Peck Gee Seah; Pamela Thayalan; Siao H. Lim; Boon Heng Lee; Anne Goh; Whitney Barnes; Zhong Chen; Kerstin Gagaring; Arnab K. Chatterjee; Kevin Pethe; Kelli Kuhen; John R. Walker; Gu Feng; Sreehari Babu; Lijun Zhang; Francesca Blasco; David Beer; Margaret Weaver

The small-molecule indolcarboxamide is a potential drug candidate for treating multidrug-resistant tuberculosis. Combating the Scourge of TB Tuberculosis (TB) caused by the bacterium Mycobacterium tuberculosis (Mtb) continues to be an epidemic in many parts of the world. Resistance to multiple drugs and the emergence of the HIV epidemic have created new challenges in TB treatment. Drugs with new mechanisms of action and improved safety profiles are urgently needed to manage TB. To achieve this goal, Rao et al. screened a chemical library of nearly 2 million compounds for inhibitors of mycobacterial growth. Using phenotypic high-throughput screening, they identified a group of molecules called indolcarboxamides as a new class of antitubercular bactericidal agents. Several indolcarboxamide analogs were evaluated to optimize their activity against Mtb and improve their properties. Two lead candidates, NITD-304 and NITD-349, with promising in vivo pharmacokinetic profiles showed potent activity against both drug-sensitive and multidrug-resistant Mtb clinical isolates. Investigating the mechanism of action, the authors found that the molecular target of the indolcarboxamides was MmpL3, a protein that is essential for mycobacterial cell wall biosynthesis and growth. NITD-304 and NITD-349 were efficacious in treating Mtb infections in mouse models of acute and chronic TB with a favorable safety margin. NITD-304 and NITD-349 are promising new drug candidates for treating TB with the potential to help fill the gap in the global TB drug discovery portfolio. New chemotherapeutic compounds against multidrug-resistant Mycobacterium tuberculosis (Mtb) are urgently needed to combat drug resistance in tuberculosis (TB). We have identified and characterized the indolcarboxamides as a new class of antitubercular bactericidal agent. Genetic and lipid profiling studies identified the likely molecular target of indolcarboxamides as MmpL3, a transporter of trehalose monomycolate that is essential for mycobacterial cell wall biosynthesis. Two lead candidates, NITD-304 and NITD-349, showed potent activity against both drug-sensitive and multidrug-resistant clinical isolates of Mtb. Promising pharmacokinetic profiles of both compounds after oral dosing in several species enabled further evaluation for efficacy and safety. NITD-304 and NITD-349 were efficacious in treating both acute and chronic Mtb infections in mouse efficacy models. Furthermore, dosing of NITD-304 and NITD-349 for 2 weeks in exploratory rat toxicology studies revealed a promising safety margin. Finally, neither compound inhibited the activity of major cytochrome P-450 enzymes or the hERG (human ether-a-go-go related gene) channel. These results suggest that NITD-304 and NITD-349 should undergo further development as a potential treatment for multidrug-resistant TB.

Collaboration


Dive into the Kelli Kuhen's collaboration.

Top Co-Authors

Avatar

Arnab K. Chatterjee

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David C. Tully

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kerstin Gagaring

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Rachel Borboa

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Richard Glynne

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Yun He

Chongqing University

View shared research outputs
Researchain Logo
Decentralizing Knowledge