Network


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

Hotspot


Dive into the research topics where Joshua N. Finger is active.

Publication


Featured researches published by Joshua N. Finger.


Molecular Cell | 2014

RIP3 Induces Apoptosis Independent of Pronecrotic Kinase Activity

Pratyusha Mandal; Scott B. Berger; Sirika Pillay; Kenta Moriwaki; Chunzi Huang; Hongyan Guo; John D. Lich; Joshua N. Finger; Viera Kasparcova; Bart Votta; Michael T. Ouellette; Bryan W. King; David D. Wisnoski; Ami S. Lakdawala; Michael P. DeMartino; Linda N. Casillas; Pamela A. Haile; Clark A. Sehon; Robert W. Marquis; Jason W. Upton; Lisa P. Daley-Bauer; Linda Roback; Nancy Ramia; Cole M. Dovey; Jan E. Carette; Francis Ka-Ming Chan; John Bertin; Peter J. Gough; Edward S. Mocarski; William J. Kaiser

Receptor-interacting protein kinase 3 (RIP3 or RIPK3) has emerged as a central player in necroptosis and a potential target to control inflammatory disease. Here, three selective small-molecule compounds are shown to inhibit RIP3 kinase-dependent necroptosis, although their therapeutic value is undermined by a surprising, concentration-dependent induction of apoptosis. These compounds interact with RIP3 to activate caspase 8 (Casp8) via RHIM-driven recruitment of RIP1 (RIPK1) to assemble a Casp8-FADD-cFLIP complex completely independent of pronecrotic kinase activities and MLKL. RIP3 kinase-dead D161N mutant induces spontaneous apoptosis independent of compound, whereas D161G, D143N, and K51A mutants, like wild-type, only trigger apoptosis when compound is present. Accordingly, RIP3-K51A mutant mice (Rip3(K51A/K51A)) are viable and fertile, in stark contrast to the perinatal lethality of Rip3(D161N/D161N) mice. RIP3 therefore holds both necroptosis and apoptosis in balance through a Ripoptosome-like platform. This work highlights a common mechanism unveiling RHIM-driven apoptosis by therapeutic or genetic perturbation of RIP3.


Journal of Immunology | 2014

Cutting Edge: RIP1 Kinase Activity Is Dispensable for Normal Development but Is a Key Regulator of Inflammation in SHARPIN-Deficient Mice

Scott B. Berger; Viera Kasparcova; Sandy Hoffman; Barb Swift; Lauren Dare; Michelle Schaeffer; Carol Capriotti; Michael N. Cook; Joshua N. Finger; Angela Hughes-Earle; Philip A. Harris; William J. Kaiser; Edward S. Mocarski; John Bertin; Peter J. Gough

RIP1 (RIPK1) kinase is a key regulator of TNF-induced NF-κB activation, apoptosis, and necroptosis through its kinase and scaffolding activities. Dissecting the balance of RIP1 kinase activity and scaffolding function in vivo during development and TNF-dependent inflammation has been hampered by the perinatal lethality of RIP1-deficient mice. In this study, we generated RIP1 kinase-dead (Ripk1(K45A)) mice and showed they are viable and healthy, indicating that the kinase activity of RIP1, but not its scaffolding function, is dispensable for viability and homeostasis. After validating that the Ripk1(K45A) mice were specifically protected against necroptotic stimuli in vitro and in vivo, we crossed them with SHARPIN-deficient cpdm mice, which develop severe skin and multiorgan inflammation that has been hypothesized to be mediated by TNF-dependent apoptosis and/or necroptosis. Remarkably, crossing Ripk1(K45A) mice with the cpdm strain protected against all cpdm-related pathology. Together, these data suggest that RIP1 kinase represents an attractive therapeutic target for TNF-driven inflammatory diseases.


Journal of Biological Chemistry | 2012

Autolytic Proteolysis within the Function to Find Domain (FIIND) Is Required for NLRP1 Inflammasome Activity

Joshua N. Finger; John D. Lich; Lauren Dare; Michael N. Cook; Kristin K. Brown; Chaya Duraiswami; John Bertin; Peter J. Gough

Background: NLRP1 mediates the release of the inflammatory cytokine IL-1β and is linked to several human inflammatory diseases. Results: Autolytic proteolysis occurs within the C terminus of NLRP1 and is modulated by polymorphisms and alternative mRNA splicing. Conclusion: Autolytic cleavage is a key regulator of the NLRP1 inflammasome and downstream IL-1β production. Significance: Understanding the mechanisms underlying NLRP1 activation is required to develop effective therapeutics. Nucleotide-binding domain leucine-rich repeat proteins (NLRs) play a key role in immunity and disease through their ability to modulate inflammation in response to pathogen-derived and endogenous danger signals. Here, we identify the requirements for activation of NLRP1, an NLR protein associated with a number of human pathologies, including vitiligo, rheumatoid arthritis, and Crohn disease. We demonstrate that NLRP1 activity is dependent upon ASC, which associates with the C-terminal CARD domain of NLRP1. In addition, we show that NLRP1 activity is dependent upon autolytic cleavage at Ser1213 within the FIIND. Importantly, this post translational event is dependent upon the highly conserved distal residue His1186. A disease-associated single nucleotide polymorphism near His1186 and a naturally occurring mRNA splice variant lacking exon 14 differentially affect this autolytic processing and subsequent NLRP1 activity. These results describe key molecular pathways that regulate NLRP1 activity and offer insight on how small sequence variations in NLR genes may influence human disease pathogenesis.


ACS Medicinal Chemistry Letters | 2013

Discovery of Small Molecule RIP1 Kinase Inhibitors for the Treatment of Pathologies Associated with Necroptosis.

Philip A. Harris; Deepak Bandyopadhyay; Scott B. Berger; Nino Campobasso; Carol Capriotti; Julie A. Cox; Lauren Dare; Joshua N. Finger; Sandra J. Hoffman; Kirsten M. Kahler; Ruth Lehr; John D. Lich; Rakesh Nagilla; Robert T. Nolte; Michael T. Ouellette; Christina S. Pao; Michelle Schaeffer; Angela Smallwood; Helen H. Sun; Barbara A. Swift; Rachel Totoritis; Paris Ward; Robert W. Marquis; John Bertin; Peter J. Gough

Potent inhibitors of RIP1 kinase from three distinct series, 1-aminoisoquinolines, pyrrolo[2,3-b]pyridines, and furo[2,3-d]pyrimidines, all of the type II class recognizing a DLG-out inactive conformation, were identified from screening of our in-house kinase focused sets. An exemplar from the furo[2,3-d]pyrimidine series showed a dose proportional response in protection from hypothermia in a mouse model of TNFα induced lethal shock.


Journal of Medicinal Chemistry | 2017

Discovery of a First-in-Class Receptor Interacting Protein 1 (RIP1) Kinase Specific Clinical Candidate (GSK2982772) for the Treatment of Inflammatory Diseases

Philip A. Harris; Scott B. Berger; Jae U. Jeong; Rakesh Nagilla; Deepak Bandyopadhyay; Nino Campobasso; Carol Capriotti; Julie A. Cox; Lauren Dare; Xiaoyang Dong; Patrick M. Eidam; Joshua N. Finger; Sandra J. Hoffman; James Kang; Viera Kasparcova; Bryan W. King; Ruth Lehr; Yunfeng Lan; Lara Kathryn Leister; John D. Lich; Thomas T. MacDonald; Nathan A. Miller; Michael T. Ouellette; Christina S. Pao; Attiq Rahman; Michael Reilly; Alan R. Rendina; Elizabeth J. Rivera; Michelle Schaeffer; Clark A. Sehon

RIP1 regulates necroptosis and inflammation and may play an important role in contributing to a variety of human pathologies, including immune-mediated inflammatory diseases. Small-molecule inhibitors of RIP1 kinase that are suitable for advancement into the clinic have yet to be described. Herein, we report our lead optimization of a benzoxazepinone hit from a DNA-encoded library and the discovery and profile of clinical candidate GSK2982772 (compound 5), currently in phase 2a clinical studies for psoriasis, rheumatoid arthritis, and ulcerative colitis. Compound 5 potently binds to RIP1 with exquisite kinase specificity and has excellent activity in blocking many TNF-dependent cellular responses. Highlighting its potential as a novel anti-inflammatory agent, the inhibitor was also able to reduce spontaneous production of cytokines from human ulcerative colitis explants. The highly favorable physicochemical and ADMET properties of 5, combined with high potency, led to a predicted low oral dose in humans.


Stroke | 2017

Neuronal Death After Hemorrhagic Stroke In Vitro and In Vivo Shares Features of Ferroptosis and Necroptosis

Marietta Zille; Saravanan S. Karuppagounder; Yingxin Chen; Peter J. Gough; John Bertin; Joshua N. Finger; Teresa A. Milner; Elizabeth A. Jonas; Rajiv R. Ratan

Background and Purpose— Intracerebral hemorrhage leads to disability or death with few established treatments. Adverse outcomes after intracerebral hemorrhage result from irreversible damage to neurons resulting from primary and secondary injury. Secondary injury has been attributed to hemoglobin and its oxidized product hemin from lysed red blood cells. The aim of this study was to identify the underlying cell death mechanisms attributable to secondary injury by hemoglobin and hemin to broaden treatment options. Methods— We investigated cell death mechanisms in cultured neurons exposed to hemoglobin or hemin. Chemical inhibitors implicated in all known cell death pathways were used. Identified cell death mechanisms were confirmed using molecular markers and electron microscopy. Results— Chemical inhibitors of ferroptosis and necroptosis protected against hemoglobin- and hemin-induced toxicity. By contrast, inhibitors of caspase-dependent apoptosis, protein or mRNA synthesis, autophagy, mitophagy, or parthanatos had no effect. Accordingly, molecular markers of ferroptosis and necroptosis were increased after intracerebral hemorrhage in vitro and in vivo. Electron microscopy showed that hemin induced a necrotic phenotype. Necroptosis and ferroptosis inhibitors each abrogated death by >80% and had similar therapeutic windows in vitro. Conclusions— Experimental intracerebral hemorrhage shares features of ferroptotic and necroptotic cell death, but not caspase-dependent apoptosis or autophagy. We propose that ferroptosis or necroptotic signaling induced by lysed blood is sufficient to reach a threshold of death that leads to neuronal necrosis and that inhibition of either of these pathways can bring cells below that threshold to survival.


PLOS ONE | 2015

MALT1 Protease Activity Is Required for Innate and Adaptive Immune Responses.

Jong W. Yu; Sandy Hoffman; Allison M. Beal; Angela Dykon; Michael A. Ringenberg; Anna C. Hughes; Lauren Dare; Amber D. Anderson; Joshua N. Finger; Viera Kasparcova; David J. Rickard; Scott B. Berger; Joshi M. Ramanjulu; John G. Emery; Peter J. Gough; John Bertin; Kevin Foley

CARMA-BCL10-MALT1 signalosomes play important roles in antigen receptor signaling and other pathways. Previous studies have suggested that as part of this complex, MALT1 functions as both a scaffolding protein to activate NF-κB through recruitment of ubiquitin ligases, and as a protease to cleave and inactivate downstream inhibitory signaling proteins. However, our understanding of the relative importance of these two distinct MALT1 activities has been hampered by a lack of selective MALT1 protease inhibitors with suitable pharmacologic properties. To fully investigate the role of MALT1 protease activity, we generated mice homozygous for a protease-dead mutation in MALT1. We found that some, but not all, MALT1 functions in immune cells were dependent upon its protease activity. Protease-dead mice had defects in the generation of splenic marginal zone and peritoneal B1 B cells. CD4+ and CD8+ T cells displayed decreased T cell receptor-stimulated proliferation and IL-2 production while B cell receptor-stimulated proliferation was partially dependent on protease activity. In dendritic cells, stimulation of cytokine production through the Dectin-1, Dectin-2, and Mincle C-type lectin receptors was also found to be partially dependent upon protease activity. In vivo, protease-dead mice had reduced basal immunoglobulin levels, and showed defective responses to immunization with T-dependent and T-independent antigens. Surprisingly, despite these decreased responses, MALT1 protease-dead mice, but not MALT1 null mice, developed mixed inflammatory cell infiltrates in multiple organs, suggesting MALT1 protease activity plays a role in immune homeostasis. These findings highlight the importance of MALT1 protease activity in multiple immune cell types, and in integrating immune responses in vivo.


Journal of Medicinal Chemistry | 2016

The Identification and Pharmacological Characterization of 6-(tert-Butylsulfonyl)-N-(5-fluoro-1H-indazol-3-yl)quinolin-4-amine (GSK583), a Highly Potent and Selective Inhibitor of RIP2 Kinase

Pamela A. Haile; Bartholomew J. Votta; Robert W. Marquis; Michael Jonathan Bury; John F. Mehlmann; Robert R. Singhaus; Adam K. Charnley; Ami S. Lakdawala; David B. Lipshutz; Biva Desai; Barbara Swift; Carol Capriotti; Scott B. Berger; Mukesh K. Mahajan; Michael Reilly; Elizabeth J. Rivera; Helen H. Sun; Rakesh Nagilla; Allison M. Beal; Joshua N. Finger; Michael N. Cook; Bryan W. King; Michael T. Ouellette; Rachel Totoritis; Maria Pierdomenico; Anna Negroni; Laura Stronati; Salvatore Cucchiara; Bartłomiej Ziółkowski; Anna Vossenkämper

RIP2 kinase is a central component of the innate immune system and enables downstream signaling following activation of the pattern recognition receptors NOD1 and NOD2, leading to the production of inflammatory cytokines. Recently, several inhibitors of RIP2 kinase have been disclosed that have contributed to the fundamental understanding of the role of RIP2 in this pathway. However, because they lack either broad kinase selectivity or strong affinity for RIP2, these tools have only limited utility to assess the role of RIP2 in complex environments. We present, herein, the discovery and pharmacological characterization of GSK583, a next-generation RIP2 inhibitor possessing exquisite selectivity and potency. Having demonstrated the pharmacological precision of this tool compound, we report its use in elucidating the role of RIP2 kinase in a variety of in vitro, in vivo, and ex vivo experiments, further clarifying our understanding of the role of RIP2 in NOD1 and NOD2 mediated disease pathogenesis.


Pharmacology Research & Perspectives | 2017

Randomized clinical study of safety, pharmacokinetics, and pharmacodynamics of RIPK1 inhibitor GSK2982772 in healthy volunteers

Kathleen Weisel; Nicola Scott; Debra J. Tompson; Bartholomew J. Votta; Sujith Madhavan; Kat Povey; Allen Wolstenholme; Monica Simeoni; Todd Rudo; Lauren E. Richards-Peterson; Tarjinder Sahota; J. Gene Wang; John D. Lich; Joshua N. Finger; Adeline Verticelli; Michael Reilly; Peter J. Gough; Philip A. Harris; John Bertin; Mei‐Lun Wang

GSK2982772 is a highly selective inhibitor of receptor‐interacting protein kinase 1 (RIPK1) being developed to treat chronic inflammatory diseases. This first‐in‐human study evaluated safety, tolerability, pharmacokinetics (PK), and exploratory pharmacodynamics (PD) of GSK2982772 administered orally to healthy male volunteers. This was a Phase I, randomized, placebo‐controlled, double‐blind study. In Part A, subjects received single ascending doses of GSK2982772 (0.1‐120 mg) or placebo in a crossover design during each of 4 treatment periods. In Part B, subjects received repeat doses of GSK2982772 (20 mg once daily [QD] to up to 120 mg twice daily [BID]) or placebo for 14 days. Part C was an open‐label relative bioavailability study comparing 20‐mg tablets vs capsules. Safety, tolerability, pharmacokinetics (PK), RIPK1 target engagement (TE), and pharmacodynamics (PD) were assessed. The most common adverse events (AEs) were contact dermatitis and headache. Most AEs were mild in intensity, and there were no deaths or serious AEs. The PK of GSK2982772 was approximately linear over the dose range studied (up to 120 mg BID). There was no evidence of drug accumulation upon repeat dosing. Greater than 90% RIPK1 TE was achieved over a 24‐hour period for the 60‐mg and 120‐mg BID dosing regimens. Single and repeat doses of GSK2982772 were safe and well tolerated. PK profiles showed dose linearity. The high levels of RIPK1 TE support progression into Phase II clinical trials for further clinical development.


Pharmacology Research & Perspectives | 2017

Identification of an antibody‐based immunoassay for measuring direct target binding of RIPK1 inhibitors in cells and tissues

Joshua N. Finger; Jean-Marie Brusq; Nino Campobasso; Michael N. Cook; Jennifer Deutsch; Heather Haag; Philip A. Harris; Earl L. Jenkins; Devika Joglekar; John D. Lich; Sean Maguire; Rakesh Nagilla; Elizabeth J. Rivera; Helen H. Sun; Bartholomew J. Votta; John Bertin; Peter J. Gough

Therapies that suppress RIPK1 kinase activity are emerging as promising therapeutic agents for the treatment of multiple inflammatory disorders. The ability to directly measure drug binding of a RIPK1 inhibitor to its target is critical for providing insight into pharmacokinetics, pharmacodynamics, safety and clinical efficacy, especially for a first‐in‐class small‐molecule inhibitor where the mechanism has yet to be explored. Here, we report a novel method for measuring drug binding to RIPK1 protein in cells and tissues. This TEAR1 (Target Engagement Assessment for RIPK1) assay is a pair of immunoassays developed on the principle of competition, whereby a first molecule (ie, drug) prevents the binding of a second molecule (ie, antibody) to the target protein. Using the TEAR1 assay, we have validated the direct binding of specific RIPK1 inhibitors in cells, blood and tissues following treatment with benzoxazepinone (BOAz) RIPK1 inhibitors. The TEAR1 assay is a valuable tool for facilitating the clinical development of the lead RIPK1 clinical candidate compound, GSK2982772, as a first‐in‐class RIPK1 inhibitor for the treatment of inflammatory disease.

Collaboration


Dive into the Joshua N. Finger's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Scott B. Berger

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John D. Lich

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge