160 results on '"John Bertin"'
Search Results
2. Ubiquitylation of MLKL at lysine 219 positively regulates necroptosis-induced tissue injury and pathogen clearance
- Author
-
Laura Ramos Garcia, Tencho Tenev, Richard Newman, Rachel O. Haich, Gianmaria Liccardi, Sidonie Wicky John, Alessandro Annibaldi, Lu Yu, Mercedes Pardo, Samuel N. Young, Cheree Fitzgibbon, Winnie Fernando, Naomi Guppy, Hyojin Kim, Lung-Yu Liang, Isabelle S. Lucet, Andrew Kueh, Ioannis Roxanis, Patrycja Gazinska, Martin Sims, Tomoko Smyth, George Ward, John Bertin, Allison M. Beal, Brad Geddes, Jyoti S. Choudhary, James M. Murphy, K. Aurelia Ball, Jason W. Upton, and Pascal Meier
- Subjects
Science - Abstract
Necroptosis is a form of cell death characterized by membrane rupture via MLKL oligomerization, although mechanistic details remain unclear. Here, the authors show that MLKL ubiquitylation of K219 facilitates high-order membrane assembly and subsequent rupture, promoting cytotoxicity.
- Published
- 2021
- Full Text
- View/download PDF
3. Inactivation of RIP3 kinase sensitizes to 15LOX/PEBP1-mediated ferroptotic death
- Author
-
Andrew M. Lamade, Limin Wu, Haider H. Dar, Heather L. Mentrup, Indira H. Shrivastava, Michael W. Epperly, Claudette M. St Croix, Yulia Y. Tyurina, Tamil S. Anthonymuthu, Qin Yang, Aleksandr A. Kapralov, Zhentai Huang, Gaowei Mao, Andrew A. Amoscato, Zachary E. Hier, Margarita A. Artyukhova, Galina Shurin, Joel C. Rosenbaum, Peter J. Gough, John Bertin, Andrew P. VanDemark, Simon C. Watkins, Kevin P. Mollen, Ivet Bahar, Joel S. Greenberger, Valerian E. Kagan, Michael J. Whalen, and Hülya Bayır
- Subjects
Lipid signaling ,Whole body irradiation ,Traumatic brain injury ,Regulated necrosis ,Necroptotic death ,Ferroptotic death ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Ferroptosis and necroptosis are two pro-inflammatory cell death programs contributing to major pathologies and their inhibition has gained attention to treat a wide range of disease states. Necroptosis relies on activation of RIP1 and RIP3 kinases. Ferroptosis is triggered by oxidation of polyunsaturated phosphatidylethanolamines (PUFA-PE) by complexes of 15-Lipoxygenase (15LOX) with phosphatidylethanolamine-binding protein 1 (PEBP1). The latter, also known as RAF kinase inhibitory protein, displays promiscuity towards multiple proteins. In this study we show that RIP3 K51A kinase inactive mice have increased ferroptotic burden and worse outcome after irradiation and brain trauma rescued by anti-ferroptotic compounds Liproxstatin-1 and Ferrostatin 16-86. Given structural homology between RAF and RIP3, we hypothesized that PEBP1 acts as a necroptosis-to-ferroptosis switch interacting with either RIP3 or 15LOX. Using genetic, biochemical, redox lipidomics and computational approaches, we uncovered that PEBP1 complexes with RIP3 and inhibits necroptosis. Elevated expression combined with higher affinity enables 15LOX to pilfer PEBP1 from RIP3, thereby promoting PUFA-PE oxidation and ferroptosis which sensitizes Rip3K51A/K51A kinase-deficient mice to total body irradiation and brain trauma. This newly unearthed PEBP1/15LOX-driven mechanism, along with previously established switch between necroptosis and apoptosis, can serve multiple and diverse cell death regulatory functions across various human disease states.
- Published
- 2022
- Full Text
- View/download PDF
4. RIPK1 Mediates TNF-Induced Intestinal Crypt Apoptosis During Chronic NF-κB ActivationSummary
- Author
-
Jerry Wong, Ricard Garcia-Carbonell, Matija Zelic, Samuel B. Ho, Brigid S. Boland, Shih-Jing Yao, Shalin A. Desai, Soumita Das, Núria Planell, Philip A. Harris, Joan Font-Burgada, Koji Taniguchi, John Bertin, Azucena Salas, Manolis Pasparakis, Pete J. Gough, Michelle Kelliher, Michael Karin, and Monica Guma
- Subjects
Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Background and Aims: Tumor necrosis factor (TNF) is a major pathogenic effector and a therapeutic target in inflammatory bowel disease (IBD), yet the basis for TNF-induced intestinal epithelial cell (IEC) death is unknown, because TNF does not kill normal IECs. Here, we investigated how chronic nuclear factor (NF)- κB activation, which occurs in human IBD, promotes TNF-dependent IEC death in mice. Methods: Human IBD specimens were stained for p65 and cleaved caspase-3. C57BL/6 mice with constitutively active IKKβ in IEC (Ikkβ(EE)IEC), Ripk1D138N/D138N knockin mice, and Ripk3-/- mice were injected with TNF or lipopolysaccharide. Enteroids were also isolated from these mice and challenged with TNF with or without RIPK1 and RIPK3 inhibitors or butylated hydroxyanisole. Ripoptosome-mediated caspase-8 activation was assessed by immunoprecipitation. Results: NF-κB activation in human IBD correlated with appearance of cleaved caspase-3. Congruently, unlike normal mouse IECs that are TNF-resistant, IECs in Ikkβ(EE)IEC mice and enteroids were susceptible to TNF-dependent apoptosis, which depended on the protein kinase function of RIPK1. Constitutively active IKKβ facilitated ripoptosome formation, a RIPK1 signaling complex that mediates caspase-8 activation by TNF. Butylated hydroxyanisole treatment and RIPK1 inhibitors attenuated TNF-induced and ripoptosome-mediated caspase-8 activation and IEC death in vitro and in vivo. Conclusions: Contrary to common expectations, chronic NF-κB activation induced intestinal crypt apoptosis after TNF stimulation, resulting in severe mucosal erosion. RIPK1 kinase inhibitors selectively inhibited TNF destructive properties while preserving its survival and proliferative properties, which do not require RIPK1 kinase activity. RIPK1 kinase inhibition could be a potential treatment for IBD. Keywords: IBD, RIPK1, Intestinal Epithelial Cell, Ripoptosome, Cell Death
- Published
- 2020
- Full Text
- View/download PDF
5. Stage-Dependent Impact of RIPK1 Inhibition on Atherogenesis: Dual Effects on Inflammation and Foam Cell Dynamics
- Author
-
Yuze Zhang, Huihui Li, Yonghu Huang, Hong Chen, Haojie Rao, Guoli Yang, Qing Wan, Zekun Peng, John Bertin, Brad Geddes, Michael Reilly, Jean-Luc Tran, and Miao Wang
- Subjects
atherosclerosis ,RIPK1 ,foam cells ,macrophages ,inflammation ,reverse cholesterol transport ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Objective: Atherosclerosis is an arterial occlusive disease with hypercholesterolemia and hypertension as common risk factors. Advanced-stage stenotic plaque, which features inflammation and necrotic core formation, is the major reason for clinical intervention. Receptor interacting serine/threonine-protein kinase 1 (RIPK1) mediates inflammation and cell death and is expressed in atherosclerotic lesions. The role of RIPK1 in advanced-stage atherosclerosis is unknown.Approach and Results: To investigate the effect of RIPK1 inhibition in advanced atherosclerotic plaque formation, we used ApoESA/SA mice, which exhibit hypercholesterolemia, and develop angiotensin-II mediated hypertension upon administration of doxycycline in drinking water. These mice readily develop severe atherosclerosis, including that in coronary arteries. Eight-week-old ApoESA/SA mice were randomized to orally receive a highly selective RIPK1 inhibitor (RIPK1i, GSK547) mixed with a western diet, or control diet. RIPK1i administration reduced atherosclerotic plaque lesion area at 2 weeks of treatment, consistent with suppressed inflammation (MCP-1, IL-1β, TNF-α) and reduced monocyte infiltration. However, administration of RIPK1i unexpectedly exacerbated atherosclerosis at 4 weeks of treatment, concomitant with increased macrophages and lipid deposition in the plaques. Incubation of isolated macrophages with oxidized LDL resulted in foam cell formation in vitro. RIPK1i treatment promoted such foam cell formation while suppressing the death of these cells. Accordingly, RIPK1i upregulated the expression of lipid metabolism-related genes (Cd36, Ppara, Lxrα, Lxrb, Srebp1c) in macrophage foam cells with ABCA1/ABCG1 unaltered. Furthermore, RIPK1i treatment inhibited ApoA1 synthesis in the liver and reduced plasma HDL levels.Conclusion: RIPK1 modulates the development of atherosclerosis in a stage-dependent manner, implicating both pro-atherosclerotic (monocyte infiltration and inflammation) and anti-atherosclerotic effects (suppressing foam cell accumulation and promoting ApoA1 synthesis). It is critical to identify an optimal therapeutic duration for potential clinical use of RIPK1 inhibitor in atherosclerosis or other related disease indications.
- Published
- 2021
- Full Text
- View/download PDF
6. LUBAC prevents lethal dermatitis by inhibiting cell death induced by TNF, TRAIL and CD95L
- Author
-
Lucia Taraborrelli, Nieves Peltzer, Antonella Montinaro, Sebastian Kupka, Eva Rieser, Torsten Hartwig, Aida Sarr, Maurice Darding, Peter Draber, Tobias L. Haas, Ayse Akarca, Teresa Marafioti, Manolis Pasparakis, John Bertin, Peter J. Gough, Philippe Bouillet, Andreas Strasser, Martin Leverkus, John Silke, and Henning Walczak
- Subjects
Science - Abstract
TNF mediated inflammation is critical in autoimmune mediated pathology, however many patients are refractory to current anti-TNF therapeutics. Here the authors show induction of several death ligands, in addition to TNF is sufficient to cause fatal dermatitis in a LUBAC deficient murine model of disease.
- Published
- 2018
- Full Text
- View/download PDF
7. Distinct Kinase-Independent Role of RIPK3 in CD11c+ Mononuclear Phagocytes in Cytokine-Induced Tissue Repair
- Author
-
Kenta Moriwaki, Sakthi Balaji, John Bertin, Peter J. Gough, and Francis Ka-Ming Chan
- Subjects
necroptosis ,IL-23 ,IL-1b ,injury ,tissue repair ,RIPK3 ,RHIM ,inflammation ,colitis ,dextran sodium sulfate ,Biology (General) ,QH301-705.5 - Abstract
Receptor interacting protein kinase 3 (RIPK3) induces necroptosis, a type of regulated necrosis, through its kinase domain and receptor interacting protein (RIP) homotypic interaction motif (RHIM). In addition, RIPK3 has been shown to regulate NLRP3 inflammasome and nuclear factor κB (NF-κB) activation. However, the relative contribution of these signaling pathways to RIPK3-dependent inflammation in distinct immune effectors is unknown. To investigate these questions, we generated RIPK3-GFP reporter mice. We found that colonic CD11c+CD11b+CD14+ mononuclear phagocytes (MNPs) expressed the highest level of RIPK3 in the lamina propria. Consequently, deletion of the RIPK3 RHIM in CD11c+ cells alone was sufficient to impair dextran sodium sulfate (DSS)-induced interleukin (IL)-23 and IL-1β expression, leading to severe intestinal inflammation. In contrast, mice expressing kinase inactive RIPK3 were not hypersensitive to DSS. Thus, a key physiological function of RIPK3 is to promote reparative cytokine expression through intestinal CD11c+ MNPs in a kinase- and necroptosis-independent manner.
- Published
- 2017
- Full Text
- View/download PDF
8. Protective Effects of Necrostatin-1 in Acute Pancreatitis: Partial Involvement of Receptor Interacting Protein Kinase 1
- Author
-
Yulin Ouyang, Li Wen, Jane A. Armstrong, Michael Chvanov, Diane Latawiec, Wenhao Cai, Mohammad Awais, Rajarshi Mukherjee, Wei Huang, Peter J. Gough, John Bertin, Alexei V. Tepikin, Robert Sutton, and David N. Criddle
- Subjects
acute pancreatitis ,receptor-interacting protein kinase 1 ,RIPK1 ,necrostatin-1 ,necroptosis ,cell death ,Cytology ,QH573-671 - Abstract
Acute pancreatitis (AP) is a severe and potentially fatal disease caused predominantly by alcohol excess and gallstones, which lacks a specific therapy. The role of Receptor-Interacting Protein Kinase 1 (RIPK1), a key component of programmed necrosis (Necroptosis), is unclear in AP. We assessed the effects of RIPK1 inhibitor Necrostatin-1 (Nec-1) and RIPK1 modification (RIPK1K45A: kinase dead) in bile acid (TLCS-AP), alcoholic (FAEE-AP) and caerulein hyperstimulation (CER-AP) mouse models. Involvement of collateral Nec-1 target indoleamine 2,3-dioxygenase (IDO) was probed with the inhibitor Epacadostat (EPA). Effects of Nec-1 and RIPK1K45A were also compared on pancreatic acinar cell (PAC) fate in vitro and underlying mechanisms explored. Nec-1 markedly ameliorated histological and biochemical changes in all models. However, these were only partially reduced or unchanged in RIPK1K45A mice. Inhibition of IDO with EPA was protective in TLCS-AP. Both Nec-1 and RIPK1K45A modification inhibited TLCS- and FAEE-induced PAC necrosis in vitro. Nec-1 did not affect TLCS-induced Ca2+ entry in PACs, however, it inhibited an associated ROS elevation. The results demonstrate protective actions of Nec-1 in multiple models. However, RIPK1-dependent necroptosis only partially contributed to beneficial effects, and actions on targets such as IDO are likely to be important.
- Published
- 2021
- Full Text
- View/download PDF
9. MALT1 Protease Activation Triggers Acute Disruption of Endothelial Barrier Integrity via CYLD Cleavage
- Author
-
Linda R. Klei, Dong Hu, Robert Panek, Danielle N. Alfano, Rachel E. Bridwell, Kelly M. Bailey, Katherine I. Oravecz-Wilson, Vincent J. Concel, Emily M. Hess, Matthew Van Beek, Phillip C. Delekta, Shufang Gu, Simon C. Watkins, Adrian T. Ting, Peter J. Gough, Kevin P. Foley, John Bertin, Linda M. McAllister-Lucas, and Peter C. Lucas
- Subjects
NF-κB ,thrombin ,Protease activated receptor-1 (PAR1) ,CARMA3 ,CARD10 ,Bcl10 ,G protein-coupled receptor (GPCR) ,endothelial permeability ,MALT1 protease ,CYLD ,Biology (General) ,QH301-705.5 - Abstract
Microvascular endothelial cells maintain a tight barrier to prevent passage of plasma and circulating immune cells into the extravascular tissue compartment, yet endothelial cells respond rapidly to vasoactive substances, including thrombin, allowing transient paracellular permeability. This response is a cornerstone of acute inflammation, but the mechanisms responsible are still incompletely understood. Here, we demonstrate that thrombin triggers MALT1 to proteolytically cleave cylindromatosis (CYLD). Fragmentation of CYLD results in microtubule disruption and a cascade of events leading to endothelial cell retraction and an acute permeability response. This finding reveals an unexpected role for the MALT1 protease, which previously has been viewed mostly as a driver of pro-inflammatory NF-κB signaling in lymphocytes. Thus, MALT1 not only promotes immune cell activation but also acutely regulates endothelial cell biology, actions that together facilitate tissue inflammation. Pharmacologic inhibition of MALT1 may therefore have synergistic impact by targeting multiple disparate steps in the overall inflammatory response.
- Published
- 2016
- Full Text
- View/download PDF
10. Structure Guided Design of Potent and Selective Ponatinib-Based Hybrid Inhibitors for RIPK1
- Author
-
Malek Najjar, Chalada Suebsuwong, Soumya S. Ray, Roshan J. Thapa, Jenny L. Maki, Shoko Nogusa, Saumil Shah, Danish Saleh, Peter J. Gough, John Bertin, Junying Yuan, Siddharth Balachandran, Gregory D. Cuny, and Alexei Degterev
- Subjects
Biology (General) ,QH301-705.5 - Abstract
RIPK1 and RIPK3, two closely related RIPK family members, have emerged as important regulators of pathologic cell death and inflammation. In the current work, we report that the Bcr-Abl inhibitor and anti-leukemia agent ponatinib is also a first-in-class dual inhibitor of RIPK1 and RIPK3. Ponatinib potently inhibited multiple paradigms of RIPK1- and RIPK3-dependent cell death and inflammatory tumor necrosis factor alpha (TNF-α) gene transcription. We further describe design strategies that utilize the ponatinib scaffold to develop two classes of inhibitors (CS and PN series), each with greatly improved selectivity for RIPK1. In particular, we detail the development of PN10, a highly potent and selective “hybrid” RIPK1 inhibitor, capturing the best properties of two different allosteric RIPK1 inhibitors, ponatinib and necrostatin-1. Finally, we show that RIPK1 inhibitors from both classes are powerful blockers of TNF-induced injury in vivo. Altogether, these findings outline promising candidate molecules and design approaches for targeting RIPK1- and RIPK3-driven inflammatory pathologies.
- Published
- 2015
- Full Text
- View/download PDF
11. MLKL Compromises Plasma Membrane Integrity by Binding to Phosphatidylinositol Phosphates
- Author
-
Yves Dondelinger, Wim Declercq, Sylvie Montessuit, Ria Roelandt, Amanda Goncalves, Inge Bruggeman, Paco Hulpiau, Kathrin Weber, Clark A. Sehon, Robert W. Marquis, John Bertin, Peter J. Gough, Savvas Savvides, Jean-Claude Martinou, Mathieu J.M. Bertrand, and Peter Vandenabeele
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Although mixed lineage kinase domain-like (MLKL) protein has emerged as a specific and crucial protein for necroptosis induction, how MLKL transduces the death signal remains poorly understood. Here, we demonstrate that the full four-helical bundle domain (4HBD) in the N-terminal region of MLKL is required and sufficient to induce its oligomerization and trigger cell death. Moreover, we found that a patch of positively charged amino acids on the surface of the 4HBD binds to phosphatidylinositol phosphates (PIPs) and allows recruitment of MLKL to the plasma membrane. Importantly, we found that recombinant MLKL, but not a mutant lacking these positive charges, induces leakage of PIP-containing liposomes as potently as BAX, supporting a model in which MLKL induces necroptosis by directly permeabilizing the plasma membrane. Accordingly, we found that inhibiting the formation of PI(5)P and PI(4,5)P2 specifically inhibits tumor necrosis factor (TNF)-mediated necroptosis but not apoptosis.
- Published
- 2014
- Full Text
- View/download PDF
12. MALT1 Protease Activity Is Required for Innate and Adaptive Immune Responses.
- Author
-
Jong W Yu, Sandy Hoffman, Allison M Beal, Angela Dykon, Michael A Ringenberg, Anna C Hughes, Lauren Dare, Amber D Anderson, Joshua Finger, Viera Kasparcova, David Rickard, Scott B Berger, Joshi Ramanjulu, John G Emery, Peter J Gough, John Bertin, and Kevin P Foley
- Subjects
Medicine ,Science - Abstract
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.
- Published
- 2015
- Full Text
- View/download PDF
13. Identification of selective small molecule inhibitors of the nucleotide-binding oligomerization domain 1 (NOD1) signaling pathway.
- Author
-
David J Rickard, Clark A Sehon, Viera Kasparcova, Lorena A Kallal, Pamela A Haile, Xin Zeng, Monica N Montoute, Derek D Poore, Hu Li, Zining Wu, Patrick M Eidam, John G Emery, Robert W Marquis, Peter J Gough, and John Bertin
- Subjects
Medicine ,Science - Abstract
NOD1 is an intracellular pattern recognition receptor that recognizes diaminopimelic acid (DAP), a peptidoglycan component in gram negative bacteria. Upon ligand binding, NOD1 assembles with receptor-interacting protein (RIP)-2 kinase and initiates a signaling cascade leading to the production of pro-inflammatory cytokines. Increased NOD1 signaling has been associated with a variety of inflammatory disorders suggesting that small-molecule inhibitors of this signaling complex may have therapeutic utility. We utilized a cell-based screening approach with extensive selectivity profiling to search for small molecule inhibitors of the NOD1 signaling pathway. Via this process we identified three distinct chemical series, xanthines (SB711), quinazolininones (GSK223) and aminobenzothiazoles (GSK966) that selectively inhibited iE-DAP-stimulated IL-8 release via the NOD1 signaling pathway. All three of the newly identified compound series failed to block IL-8 secretion in cells following stimulation with ligands for TNF receptor, TLR2 or NOD2 and, in addition, none of the compound series directly inhibited RIP2 kinase activity. Our initial exploration of the structure-activity relationship and physicochemical properties of the three series directed our focus to the quinazolininone biarylsulfonamides (GSK223). Further investigation allowed for the identification of significantly more potent analogs with the largest boost in activity achieved by fluoro to chloro replacement on the central aryl ring. These results indicate that the NOD1 signaling pathway, similarly to activation of NOD2, is amenable to modulation by small molecules that do not target RIP2 kinase. These compounds should prove useful tools to investigate the importance of NOD1 activation in various inflammatory processes and have potential clinical utility in diseases driven by hyperactive NOD1 signaling.
- Published
- 2014
- Full Text
- View/download PDF
14. Pathogen sensing pathways in human embryonic stem cell derived-endothelial cells: role of NOD1 receptors.
- Author
-
Daniel M Reed, Gabor Foldes, Timothy Gatheral, Koralia E Paschalaki, Zsuzsanna Lendvai, Zsolt Bagyura, Tamas Nemeth, Judit Skopal, Bela Merkely, Aurica G Telcian, Leila Gogsadze, Michael R Edwards, Peter J Gough, John Bertin, Sebastian L Johnston, Sian E Harding, and Jane A Mitchell
- Subjects
Medicine ,Science - Abstract
Human embryonic stem cell-derived endothelial cells (hESC-EC), as well as other stem cell derived endothelial cells, have a range of applications in cardiovascular research and disease treatment. Endothelial cells sense Gram-negative bacteria via the pattern recognition receptors (PRR) Toll-like receptor (TLR)-4 and nucleotide-binding oligomerisation domain-containing protein (NOD)-1. These pathways are important in terms of sensing infection, but TLR4 is also associated with vascular inflammation and atherosclerosis. Here, we have compared TLR4 and NOD1 responses in hESC-EC with those of endothelial cells derived from other stem cells and with human umbilical vein endothelial cells (HUVEC). HUVEC, endothelial cells derived from blood progenitors (blood outgrowth endothelial cells; BOEC), and from induced pluripotent stem cells all displayed both a TLR4 and NOD1 response. However, hESC-EC had no TLR4 function, but did have functional NOD1 receptors. In vivo conditioning in nude rats did not confer TLR4 expression in hESC-EC. Despite having no TLR4 function, hESC-EC sensed Gram-negative bacteria, a response that was found to be mediated by NOD1 and the associated RIP2 signalling pathways. Thus, hESC-EC are TLR4 deficient but respond to bacteria via NOD1. This data suggests that hESC-EC may be protected from unwanted TLR4-mediated vascular inflammation, thus offering a potential therapeutic advantage.
- Published
- 2014
- Full Text
- View/download PDF
15. Identification of benzimidazole diamides as selective inhibitors of the nucleotide-binding oligomerization domain 2 (NOD2) signaling pathway.
- Author
-
David J Rickard, Clark A Sehon, Viera Kasparcova, Lorena A Kallal, Xin Zeng, Monica N Montoute, Tushar Chordia, Derek D Poore, Hu Li, Zining Wu, Patrick M Eidam, Pamela A Haile, Jong Yu, John G Emery, Robert W Marquis, Peter J Gough, and John Bertin
- Subjects
Medicine ,Science - Abstract
NOD2 is an intracellular pattern recognition receptor that assembles with receptor-interacting protein (RIP)-2 kinase in response to the presence of bacterial muramyl dipeptide (MDP) in the host cell cytoplasm, thereby inducing signals leading to the production of pro-inflammatory cytokines. The dysregulation of NOD2 signaling has been associated with various inflammatory disorders suggesting that small-molecule inhibitors of this signaling complex may have therapeutic utility. To identify inhibitors of the NOD2 signaling pathway, we utilized a cell-based screening approach and identified a benzimidazole diamide compound designated GSK669 that selectively inhibited an MDP-stimulated, NOD2-mediated IL-8 response without directly inhibiting RIP2 kinase activity. Moreover, GSK669 failed to inhibit cytokine production in response to the activation of Toll-like receptor (TLR)-2, tumor necrosis factor receptor (TNFR)-1 and closely related NOD1, all of which share common downstream components with the NOD2 signaling pathway. While the inhibitors blocked MDP-induced NOD2 responses, they failed to block signaling induced by NOD2 over-expression or single stranded RNA, suggesting specificity for the MDP-induced signaling complex and activator-dependent differences in NOD2 signaling. Investigation of structure-activity relationship allowed the identification of more potent analogs that maintained NOD2 selectivity. The largest boost in activity was achieved by N-methylation of the C2-ethyl amide group. These findings demonstrate that the NOD2 signaling pathway is amenable to modulation by small molecules that do not target RIP2 kinase activity. The compounds we identified should prove useful tools to investigate the importance of NOD2 in various inflammatory processes and may have potential clinical utility.
- Published
- 2013
- Full Text
- View/download PDF
16. A key role for the endothelium in NOD1 mediated vascular inflammation: comparison to TLR4 responses.
- Author
-
Timothy Gatheral, Daniel M Reed, Laura Moreno, Peter J Gough, Bart J Votta, Clark A Sehon, David J Rickard, John Bertin, Eric Lim, Andrew G Nicholson, and Jane A Mitchell
- Subjects
Medicine ,Science - Abstract
Understanding the mechanisms by which pathogens induce vascular inflammation and dysfunction may reveal novel therapeutic targets in sepsis and related conditions. The intracellular receptor NOD1 recognises peptidoglycan which features in the cell wall of gram negative and some gram positive bacteria. NOD1 engagement generates an inflammatory response via activation of NFκB and MAPK pathways. We have previously shown that stimulation of NOD1 directly activates blood vessels and causes experimental shock in vivo. In this study we have used an ex vivo vessel-organ culture model to characterise the relative contribution of the endothelium in the response of blood vessels to NOD1 agonists. In addition we present the novel finding that NOD1 directly activates human blood vessels. Using human cultured cells we confirm that endothelial cells respond more avidly to NOD1 agonists than vascular smooth muscle cells. Accordingly we have sought to pharmacologically differentiate NOD1 and TLR4 mediated signalling pathways in human endothelial cells, focussing on TAK1, NFκB and p38 MAPK. In addition we profile novel inhibitors of RIP2 and NOD1 itself, which specifically inhibit NOD1 ligand induced inflammatory signalling in the vasculature. This paper is the first to demonstrate activation of whole human artery by NOD1 stimulation and the relative importance of the endothelium in the sensing of NOD1 ligands by vessels. This data supports the potential utility of NOD1 and RIP2 as therapeutic targets in human disease where vascular inflammation is a clinical feature, such as in sepsis and septic shock.
- Published
- 2012
- Full Text
- View/download PDF
17. Data from Elesclomol induces cancer cell apoptosis through oxidative stress
- Author
-
John Bertin, James Barsoum, Zhenjian Du, Mei Zhang, Chin-Yu Yang, Jane Kepros, Vishwasenani Balasubramanyam, Suqin He, and Jessica R. Kirshner
- Abstract
Elesclomol (formerly STA-4783) is a novel small molecule undergoing clinical evaluation in a pivotal phase III melanoma trial (SYMMETRY). In a phase II randomized, double-blinded, controlled, multi-center trial in 81 patients with stage IV metastatic melanoma, treatment with elesclomol plus paclitaxel showed a statistically significant doubling of progression-free survival time compared with treatment with paclitaxel alone. Although elesclomol displays significant therapeutic activity in the clinic, the mechanism underlying its anticancer activity has not been defined previously. Here, we show that elesclomol induces apoptosis in cancer cells through the induction of oxidative stress. Treatment of cancer cells in vitro with elesclomol resulted in the rapid generation of reactive oxygen species (ROS) and the induction of a transcriptional gene profile characteristic of an oxidative stress response. Inhibition of oxidative stress by the antioxidant N-acetylcysteine blocked the induction of gene transcription by elesclomol. In addition, N-acetylcysteine blocked drug-induced apoptosis, indicating that ROS generation is the primary mechanism responsible for the proapoptotic activity of elesclomol. Excessive ROS production and elevated levels of oxidative stress are critical biochemical alterations that contribute to cancer cell growth. Thus, the induction of oxidative stress by elesclomol exploits this unique characteristic of cancer cells by increasing ROS levels beyond a threshold that triggers cell death. [Mol Cancer Ther 2008;7(8):2319–27]
- Published
- 2023
18. Supplementary Table S1 from Elesclomol induces cancer cell apoptosis through oxidative stress
- Author
-
John Bertin, James Barsoum, Zhenjian Du, Mei Zhang, Chin-Yu Yang, Jane Kepros, Vishwasenani Balasubramanyam, Suqin He, and Jessica R. Kirshner
- Abstract
Supplementary Table S1 from Elesclomol induces cancer cell apoptosis through oxidative stress
- Published
- 2023
19. Ubiquitylation of MLKL at lysine 219 positively regulates necroptosis-induced tissue injury and pathogen clearance
- Author
-
Ioannis Roxanis, Laura Ramos Garcia, James M. Murphy, Sidonie Wicky John, Isabelle S Lucet, Andrew J. Kueh, Allison M. Beal, K. Aurelia Ball, Jyoti S. Choudhary, Cheree Fitzgibbon, Richard Newman, Jason W. Upton, Martin Sims, Lu Yu, George Ward, Gianmaria Liccardi, Winnie Fernando, Patrycja Gazinska, Naomi Guppy, Brad J. Geddes, Hyojin Kim, Samuel N. Young, Rachel O. Haich, Pascal Meier, Lung-Yu Liang, Mercedes Pardo, Tomoko Smyth, John Bertin, Tencho Tenev, and Alessandro Annibaldi
- Subjects
0301 basic medicine ,Muromegalovirus ,Programmed cell death ,Ubiquitylation ,Necroptosis ,Science ,General Physics and Astronomy ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Mice ,Necrosis ,03 medical and health sciences ,RIPK1 ,0302 clinical medicine ,Ubiquitin ,Animals ,Humans ,Cells, Cultured ,Skin ,Mice, Knockout ,Multidisciplinary ,biology ,Chemistry ,Lysine ,HEK 293 cells ,Ubiquitination ,Herpesviridae Infections ,General Chemistry ,Cell biology ,Mice, Inbred C57BL ,HEK293 Cells ,030104 developmental biology ,Lytic cycle ,Apoptosis ,NIH 3T3 Cells ,biology.protein ,Phosphorylation ,HT29 Cells ,Protein Kinases ,030217 neurology & neurosurgery - Abstract
Necroptosis is a lytic, inflammatory form of cell death that not only contributes to pathogen clearance but can also lead to disease pathogenesis. Necroptosis is triggered by RIPK3-mediated phosphorylation of MLKL, which is thought to initiate MLKL oligomerisation, membrane translocation and membrane rupture, although the precise mechanism is incompletely understood. Here, we show that K63-linked ubiquitin chains are attached to MLKL during necroptosis and that ubiquitylation of MLKL at K219 significantly contributes to the cytotoxic potential of phosphorylated MLKL. The K219R MLKL mutation protects animals from necroptosis-induced skin damage and renders cells resistant to pathogen-induced necroptosis. Mechanistically, we show that ubiquitylation of MLKL at K219 is required for higher-order assembly of MLKL at membranes, facilitating its rupture and necroptosis. We demonstrate that K219 ubiquitylation licenses MLKL activity to induce lytic cell death, suggesting that necroptotic clearance of pathogens as well as MLKL-dependent pathologies are influenced by the ubiquitin-signalling system., Necroptosis is a form of cell death characterized by membrane rupture via MLKL oligomerization, although mechanistic details remain unclear. Here, the authors show that MLKL ubiquitylation of K219 facilitates high-order membrane assembly and subsequent rupture, promoting cytotoxicity.
- Published
- 2021
20. RIPK1 kinase-dependent inflammation and cell death contribute to the pathogenesis of COPD
- Author
-
Hannelore P. Van Eeckhoutte, Chantal Donovan, Richard Y. Kim, Thomas M. Conlon, Meshal Ansari, Haroon Khan, Ranjith Jayaraman, Nicole G. Hansbro, Yves Dondelinger, Tom Delanghe, Allison M. Beal, Brad Geddes, John Bertin, Tom Vanden Berghe, Joyceline De Volder, Tania Maes, Peter Vandenabeele, Bart M. Vanaudenaerde, Dieter Deforce, Sonja Škevin, Filip Van Nieuwerburgh, Fien M. Verhamme, Guy F. Joos, Sobia Idrees, Herbert B. Schiller, Ali Önder Yildirim, Alen Faiz, Mathieu J.M. Bertrand, Guy G. Brusselle, Philip M. Hansbro, and Ken R. Bracke
- Subjects
Pulmonary and Respiratory Medicine ,11 Medical and Health Sciences, 1116 Medical Physiology ,Respiratory System - Abstract
BackgroundReceptor-interacting protein kinase 1 (RIPK1) is a key mediator of regulated cell death (including apoptosis and necroptosis) and inflammation, both drivers of COPD pathogenesis. We aimed to define the contribution of RIPK1 kinase-dependent cell death and inflammation in the pathogenesis of COPD.MethodsWe assessedRIPK1expression in single-cell RNA sequencing (RNA-seq) data from human and mouse lungs, and validated RIPK1 levels in lung tissue of COPD patientsviaimmunohistochemistry. Next, we assessed the consequences of genetic and pharmacological inhibition of RIPK1 kinase activity in experimental COPD, usingRipk1S25D/S25Dkinase-deficient mice and the RIPK1 kinase inhibitor GSK′547.ResultsRIPK1expression increased in alveolar type 1 (AT1), AT2, ciliated and neuroendocrine cells in human COPD. RIPK1 protein levels were significantly increased in airway epithelium of COPD patients compared with never-smokers and smokers without airflow limitation. In mice, exposure to cigarette smoke (CS) increasedRipk1expression similarly in AT2 cells, and further in alveolar macrophages and T-cells. Genetic and/or pharmacological inhibition of RIPK1 kinase activity significantly attenuated airway inflammation upon acute and subacute CS exposure, as well as airway remodelling, emphysema, and apoptotic and necroptotic cell death upon chronic CS exposure. Similarly, pharmacological RIPK1 kinase inhibition significantly attenuated elastase-induced emphysema and lung function decline. Finally, RNA-seq on lung tissue of CS-exposed mice revealed downregulation of cell death and inflammatory pathways upon pharmacological RIPK1 kinase inhibition.ConclusionsRIPK1 kinase inhibition is protective in experimental models of COPD and may represent a novel promising therapeutic approach.
- Published
- 2022
21. Inhibiting RIPK1 Kinase Activity Is Protective in Experimental Models of COPD
- Author
-
Hannelore P. Van Eeckhoutte, Chantal Donovan, Richard Kim, Haroon Khan, Ranjith Jayaraman, Yves Dondelinger, Tom Delanghe, Allison Beal, Brad Geddes, John Bertin, Tom Vanden Berghe, Peter Vandenabeele, Bart Vanaudenaerde, Dieter Deforce, Sonja Skevin, Filip Van Nieuwerburgh, Fien Verhamme, Guy Joos, Guy Brusselle, Mathieu Bertrand, Philip Hansbro, and Ken Bracke
- Published
- 2022
22. RIP1 Kinase Drives Macrophage-Mediated Adaptive Immune Tolerance in Pancreatic Cancer
- Author
-
Juan A. Kochen Rossi, Ankita Mishra, Berk Aykut, Mautin Hundeyin, Deirdre Jill Cohen, Ece Bagdatlioglu, Atsuo Ochi, Wenyu Su, Joshua Leinwand, Pamela Nassau, Narendra Verma, Mohammed Khan, Jacqueline I. Kim, Nan Wu, Michael Reilly, James H. Thorpe, Aarif Ahsan, Fei Li, Varshini Vasudevaraja, George Miller, Allison M. Beal, Ting Chen, Wei Wang, John D. Lich, Pardeep S. Taunk, Mukesh K. Mahajan, Inedouye Yaboh, Dennis Adeegbe, Jingjing Wu, Philip A. Harris, John Bertin, Johana Gutierrez, Kwok-Kin Wong, Brian Diskin, Emma Kurz, Yue Wu, Christina Almonte, J. Mosley, Michael Glacken, Jill M. Marinis, and Shivraj Savadkar
- Subjects
0301 basic medicine ,Cancer Research ,Macrophage polarization ,Biology ,Lymphocyte Activation ,Article ,Immune tolerance ,Mice ,03 medical and health sciences ,L Cells ,Downregulation and upregulation ,Cell Line, Tumor ,Pancreatic cancer ,Immune Tolerance ,medicine ,Animals ,Humans ,Macrophage ,Cytotoxic T cell ,Protein kinase A ,Mice, Knockout ,Kinase ,Macrophages ,Cell Biology ,Th1 Cells ,medicine.disease ,Mice, Inbred C57BL ,Pancreatic Neoplasms ,STAT1 Transcription Factor ,030104 developmental biology ,Oncology ,Receptor-Interacting Protein Serine-Threonine Kinases ,Cancer research ,Th17 Cells ,Carcinoma, Pancreatic Ductal ,T-Lymphocytes, Cytotoxic - Abstract
Summary Pancreatic ductal adenocarcinoma (PDA) is characterized by immune tolerance and immunotherapeutic resistance. We discovered upregulation of receptor-interacting serine/threonine protein kinase 1 (RIP1) in tumor-associated macrophages (TAMs) in PDA. To study its role in oncogenic progression, we developed a selective small-molecule RIP1 inhibitor with high in vivo exposure. Targeting RIP1 reprogrammed TAMs toward an MHCIIhiTNFα+IFNγ+ immunogenic phenotype in a STAT1-dependent manner. RIP1 inhibition in TAMs resulted in cytotoxic T cell activation and T helper cell differentiation toward a mixed Th1/Th17 phenotype, leading to tumor immunity in mice and in organotypic models of human PDA. Targeting RIP1 synergized with PD1-and inducible co-stimulator-based immunotherapies. Tumor-promoting effects of RIP1 were independent of its co-association with RIP3. Collectively, our work describes RIP1 as a checkpoint kinase governing tumor immunity.
- Published
- 2020
23. Understanding Pharmacokinetic Disconnect in Preclinical Species for 4-Aminoquinolines: Consequences of Low Permeability and High P-glycoprotein Efflux Ratio on Rat and Dog Oral Pharmacokinetics
- Author
-
Bartholomew J. Votta, Michael Reilly, Linda N. Casillas, Robert W. Marquis, Rakesh Nagilla, Mukesh K. Mahajan, Demartino Michael P, John Bertin, Helen H. Sun, Elizabeth J. Rivera, and Pamela A. Haile
- Subjects
Drug ,ATP Binding Cassette Transporter, Subfamily B ,media_common.quotation_subject ,Administration, Oral ,Pharmaceutical Science ,02 engineering and technology ,Pharmacology ,030226 pharmacology & pharmacy ,Permeability ,03 medical and health sciences ,Dogs ,0302 clinical medicine ,Pharmacokinetics ,Oral administration ,Animals ,Medicine ,media_common ,P-glycoprotein ,Gastrointestinal tract ,biology ,business.industry ,Biological Transport ,Transporter ,021001 nanoscience & nanotechnology ,Rats ,Bioavailability ,Aminoquinolines ,biology.protein ,Efflux ,0210 nano-technology ,business - Abstract
Receptor Interacting Protein 2 (RIP2) kinase inhibitors have been reported for therapeutic opportunities in inflammatory bowel diseases such as Ulcerative Colitis and Crohn's disease. During lead optimization, team identified 4-aminoquinoline series and several compounds from this series were investigated in rat and dog pharmacokinetic studies. While compounds such as GSKA and GSKB demonstrated acceptable pharmacokinetics in rat and dog, further progression of these compounds was halted due to adverse findings in advanced safety studies. Structurally similar analogues incorporating polarity at C-7 position of 4-aminoquinoline resulted in identification of GSKC - GSKF. Interestingly, following oral administration to rat at similar low dose, GSKC - GSKF demonstrated significantly low systemic drug exposure compared to GSKA and GSKB (3-17-fold difference). However, in dog, dose normalized oral systemic exposure for GSKC - GSKF was comparable to GSKA and GSKB (within 2-fold). A series of studies were conducted to understand the disconnect which highlighted that an intrinsic reduction in permeability and high P-glycoprotein (P-gp) efflux ratio for C-7 substituted analogues were driving pharmacokinetic disconnect between rat and dog. Oral absorption was minimally impacted in dog by P-gp mediated efflux compared to rat because the leakier gastrointestinal tract in dog likely overcomes this effect.
- Published
- 2020
24. An intestinal organoid–based platform that recreates susceptibility to T-cell–mediated tissue injury
- Author
-
Eugene Rudensky, Jennifer Tsai, Sandra J. Hoffman, Victor J. Torres, David Hudesman, Chen Liu, John Bertin, Ken Cadwell, Thomas Heaney, Brad J. Geddes, Marcel R.M. van den Brink, Jordan E. Axelrad, Yu Matsuzawa-Ishimoto, Xiaomin Yao, Allison M. Beal, Frank Yeung, P'ng Loke, Yusuke Shono, Ashley M. Hine, Jessica A Neil, Samantha L. Schuster, Michael Cammer, Ying-Han Chen, Amina Lazrak, Erin E Zwack, M. Zahidunnabi Dewan, and Katherine B Nichols
- Subjects
0301 basic medicine ,business.industry ,Necroptosis ,T cell ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Transplantation ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Graft-versus-host disease ,medicine.anatomical_structure ,Interferon ,030220 oncology & carcinogenesis ,medicine ,Organoid ,Cancer research ,business ,ATG16L1 ,Ex vivo ,medicine.drug - Abstract
A goal in precision medicine is to use patient-derived material to predict disease course and intervention outcomes. Here, we use mechanistic observations in a preclinical animal model to design an ex vivo platform that recreates genetic susceptibility to T-cell–mediated damage. Intestinal graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic hematopoietic cell transplantation. We found that intestinal GVHD in mice deficient in Atg16L1, an autophagy gene that is polymorphic in humans, is reversed by inhibiting necroptosis. We further show that cocultured allogeneic T cells kill Atg16L1-mutant intestinal organoids from mice, which was associated with an aberrant epithelial interferon signature. Using this information, we demonstrate that pharmacologically inhibiting necroptosis or interferon signaling protects human organoids derived from individuals harboring a common ATG16L1 variant from allogeneic T-cell attack. Our study provides a roadmap for applying findings in animal models to individualized therapy that targets affected tissues.
- Published
- 2020
25. Respiratory Syncytial Virus Infection Promotes Necroptosis and HMGB1 Release by Airway Epithelial Cells
- Author
-
Yves Dondelinger, Gunter Hartel, Mark L. Everard, John Bertin, Vivian Zhang, John W. Upham, Peter J. Gough, Natasha Collinson, Antoon J. M. van Oosterhout, Rhiannon B. Werder, Mathieu J.M. Bertrand, Jennifer Simpson, Ashik Ullah, Zhixuan Loh, Kirsten Spann, Simon Phipps, Christopher C Blyth, and Jason P. Lynch
- Subjects
GENETIC SUSCEPTIBILITY ,CHILDREN ,Critical Care and Intensive Care Medicine ,Mice ,0302 clinical medicine ,Medicine and Health Sciences ,Medicine ,Prospective Studies ,030212 general & internal medicine ,HMGB1 Protein ,biology ,Respiratory infection ,Pneumovirus ,respiratory system ,APOPTOSIS ,DEFICIENCY ,Child, Preschool ,Necroptosis ,Bronchiolitis ,bronchiolitis ,medicine.symptom ,Viral load ,MLKL ,Pulmonary and Respiratory Medicine ,PREDISPOSES ,necroptosis ,Inflammation ,Respiratory Mucosa ,Respiratory Syncytial Virus Infections ,HMGB1 ,03 medical and health sciences ,INFLAMMATION ,Animals ,Humans ,INNATE IMMUNE-RESPONSES ,business.industry ,Editorials ,Biology and Life Sciences ,Infant ,Epithelial Cells ,asthma ,medicine.disease ,030228 respiratory system ,Immunology ,biology.protein ,Respiratory epithelium ,MEMBRANE ,business - Abstract
Rationale: Respiratory syncytial virus (RSV) bronchiolitis causes significant infant mortality. Bronchiolitis is characterized by airway epithelial cell (AEC) death; however, the mode of death remains unknown. Objectives: To determine whether necroptosis contributes to RSV b r onchiolitis pathogenesis via HMGB1 (high mobility group box 1) release. Methods: Nasopharyngeal samples were collected from children presenting to the hospital with acute respiratory infection. Primary human AECs and neonatal mice were inoculated with RSV and murine Pneumovirus, respectively. Necroptosis was determined via viability assays and immunohistochemistry for RIPK1 (receptor-interacting protein kinase-1), MLKL (mixed lineage kinase domain-like pseudokinase) protein, and caspase-3. Necroptosis was blocked using pharmacological inhibitors and RIPK1 kinase-dead knockin mice. Measurements and Main Results: HMGB1 levels were elevated in nasopharyngeal samples of children with acute RSV infection. RSV-induced epithelial cell death was associated with increased phosphorylated RIPK1 and phosphorylated MLKL but not active caspase-3 expression. Inhibition of RIPK1 or MLKL attenuated RSV-induced HMGBI translocation and release, and lowered viral load. MLKL inhibition increased active caspase-3 expression in a caspase-8/9-dependent manner. In susceptible mice, Pneumovirus infection upregulated RIPK1 and MLKL expression in the airway epithelium at 8 to 10 days after infection, coinciding with AEC sloughing, HMGB1 release, and neutrophilic inflammation. Genetic or pharmacological inhibition of RIPK1 or MLKL attenuated these pathologies, lowered viral load, and prevented type 2 inflammation and airway remodeling. Necroptosis inhibition in early life ameliorated asthma progression induced by viral or allergen challenge in later life. Conclusions: Pneumovirus infection induces AEC necroptosis. Inhibition of necroptosis may be a viable strategy to limit the severity of viral bronchiolitis and break its nexus with asthma.
- Published
- 2020
26. MALT1 Protease Plays a Dual Role in the Allergic Response by Acting in Both Mast Cells and Endothelial Cells
- Author
-
Linda M. McAllister-Lucas, Tina L. Sumpter, Kevin Foley, Danielle N. Alfano, Peter J. Gough, John Bertin, Peter C. Lucas, Linda R. Klei, Matthew Trotta, and Hanna B. Klei
- Subjects
medicine.medical_treatment ,Immunology ,Lymphocyte Activation ,medicine.disease_cause ,Cell Line ,Proinflammatory cytokine ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Bone Marrow ,Hypersensitivity ,medicine ,Animals ,Humans ,Immunology and Allergy ,Mast Cells ,Mast cell cytokine production ,Inflammation ,Protease ,Receptors, IgE ,Chemistry ,FCER1 ,NF-kappa B ,Degranulation ,Endothelial Cells ,Immunoglobulin E ,Cell biology ,Mice, Inbred C57BL ,Cytokine ,Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein ,Allergic response ,Cytokines ,Female ,Histamine ,030215 immunology - Abstract
The signaling protein MALT1 plays a key role in promoting NF-κB activation in Ag-stimulated lymphocytes. In this capacity, MALT1 has two functions, acting as a scaffolding protein and as a substrate-specific protease. MALT1 is also required for NF-κB–dependent induction of proinflammatory cytokines after FcεR1 stimulation in mast cells, implicating a role in allergy. Because MALT1 remains understudied in this context, we sought to investigate how MALT1 proteolytic activity contributes to the overall allergic response. We compared bone marrow–derived mast cells from MALT1 knockout (MALT1−/−) and MALT1 protease-deficient (MALTPD/PD) mice to wild-type cells. We found that MALT1−/− and MALT1PD/PD mast cells are equally impaired in cytokine production following FcεRI stimulation, indicating that MALT1 scaffolding activity is insufficient to drive the cytokine response and that MALT1 protease activity is essential. In addition to cytokine production, acute mast cell degranulation is a critical component of allergic response. Intriguingly, whereas degranulation is MALT1-independent, MALT1PD/PD mice are protected from vascular edema induced by either passive cutaneous anaphylaxis or direct challenge with histamine, a major granule component. This suggests a role for MALT1 protease activity in endothelial cells targeted by mast cell–derived vasoactive substances. Indeed, we find that in human endothelial cells, MALT1 protease is activated following histamine treatment and is required for histamine-induced permeability. We thus propose a dual role for MALT1 protease in allergic response, mediating 1) IgE-dependent mast cell cytokine production, and 2) histamine-induced endothelial permeability. This dual role indicates that therapeutic inhibitors of MALT1 protease could work synergistically to control IgE-mediated allergic disease.
- Published
- 2020
27. Discovery of a First-in-Class Receptor Interacting Protein 2 (RIP2) Kinase Specific Clinical Candidate, 2-((4-(Benzo[d]thiazol-5-ylamino)-6-(tert-butylsulfonyl)quinazolin-7-yl)oxy)ethyl Dihydrogen Phosphate, for the Treatment of Inflammatory Diseases
- Author
-
Pamela A. Haile, Linda N. Casillas, Bartholomew J. Votta, Gren Z. Wang, Adam K. Charnley, Xiaoyang Dong, Michael J. Bury, Joseph J. Romano, John F. Mehlmann, Bryan W. King, Karl F. Erhard, Charles R. Hanning, David B. Lipshutz, Biva M. Desai, Carol A. Capriotti, Michelle C. Schaeffer, Scott B. Berger, Mukesh K. Mahajan, Michael A. Reilly, Rakesh Nagilla, Elizabeth J. Rivera, Helen H. Sun, John K. Kenna, Allison M. Beal, Michael T. Ouellette, Mike Kelly, Gillian Stemp, Máire A. Convery, Anna Vossenkämper, Thomas T. MacDonald, Peter J. Gough, John Bertin, and Robert W. Marquis
- Subjects
Drug Discovery ,Molecular Medicine - Published
- 2019
28. Discovery and Lead-Optimization of 4,5-Dihydropyrazoles as Mono-Kinase Selective, Orally Bioavailable and Efficacious Inhibitors of Receptor Interacting Protein 1 (RIP1) Kinase
- Author
-
Stéphane Sautet, Emilie Jigorel, Nicolas Faucher, Jamel Meslamani, Lara Kathryn Leister, Pascal Grondin, Julie A. Cox, Bryan W. King, Pamela Nassau, Gemma Victoria White, Brad J. Geddes, Alain Claude-Marie Daugan, Helen H. Sun, Michael T. Ouellette, Pauline Lamoureux, Elizabeth J. Rivera, Scott B. Berger, James H. Thorpe, Philip A. Harris, J. Mosley, Susan E. Hutchinson, Frédéric Donche, John Bertin, Sandra J. Hoffman, Natalie Wellaway, Patrick M. Eidam, Paris Ward, Florent Potvain, Clark A. Sehon, Sebastien Andre Campos, Robert W. Marquis, Peter J. Gough, Mukesh K. Mahajan, Veronique Beneton, Jae U. Jeong, Michelle C. Schaeffer, John D. Lich, Allison M. Beal, Rakesh Nagilla, James Michael Woolven, Bonnie L. Hoffman, Anderson Niall Andrew, Marie-Hélène Fouchet, Deepak Bandyopadhyay, Carol A. Capriotti, Rachel D. Totoritis, Joshua N. Finger, Kishore K. Pasikanti, David D. Wisnoski, Sze-Ling Ng, Nino Campobasso, Nicolas George, and Michael Reilly
- Subjects
Models, Molecular ,Encephalomyelitis, Autoimmune, Experimental ,Multiple Sclerosis ,Necroptosis ,Biological Availability ,Inflammation ,Pharmacology ,01 natural sciences ,Cell Line ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,In vivo ,Psoriasis ,Drug Discovery ,medicine ,Animals ,Humans ,Structure–activity relationship ,Enzyme Inhibitors ,030304 developmental biology ,0303 health sciences ,Kinase ,Chemistry ,RNA-Binding Proteins ,Haplorhini ,medicine.disease ,Ulcerative colitis ,High-Throughput Screening Assays ,Rats ,0104 chemical sciences ,Mice, Inbred C57BL ,Nuclear Pore Complex Proteins ,010404 medicinal & biomolecular chemistry ,Drug Design ,Rheumatoid arthritis ,Chronic Disease ,Pyrazoles ,Molecular Medicine ,medicine.symptom ,Retinitis Pigmentosa - Abstract
RIP1 kinase regulates necroptosis and inflammation and may play an important role in contributing to a variety of human pathologies, including inflammatory and neurological diseases. Currently, RIP1 kinase inhibitors have advanced into early clinical trials for evaluation in inflammatory diseases such as psoriasis, rheumatoid arthritis, and ulcerative colitis and neurological diseases such as amyotrophic lateral sclerosis and Alzheimer's disease. In this paper, we report on the design of potent and highly selective dihydropyrazole (DHP) RIP1 kinase inhibitors starting from a high-throughput screen and the lead-optimization of this series from a lead with minimal rat oral exposure to the identification of dihydropyrazole 77 with good pharmacokinetic profiles in multiple species. Additionally, we identified a potent murine RIP1 kinase inhibitor 76 as a valuable in vivo tool molecule suitable for evaluating the role of RIP1 kinase in chronic models of disease. DHP 76 showed efficacy in mouse models of both multiple sclerosis and human retinitis pigmentosa.
- Published
- 2019
29. Necroptosis inhibition as a therapy for Niemann-Pick disease, type C1: Inhibition of RIP kinases and combination therapy with 2-hydroxypropyl-β-cyclodextrin
- Author
-
John Bertin, Alexander Salman, Antony Cougnoux, Forbes D. Porter, S. Clifford, and S.-L. Ng
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Combination therapy ,Endocrinology, Diabetes and Metabolism ,Necroptosis ,Apoptosis ,Pharmacology ,Biochemistry ,Article ,Excipients ,Mice ,Necrosis ,03 medical and health sciences ,RIPK1 ,Endocrinology ,Niemann-Pick C1 Protein ,hemic and lymphatic diseases ,Genetics ,Animals ,Medicine ,Cognitive decline ,Kinase activity ,Molecular Biology ,Neuroinflammation ,Mice, Knockout ,Mice, Inbred BALB C ,Kinase ,business.industry ,Intracellular Signaling Peptides and Proteins ,Proteins ,nutritional and metabolic diseases ,Niemann-Pick Disease, Type C ,Combined Modality Therapy ,2-Hydroxypropyl-beta-cyclodextrin ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Receptor-Interacting Protein Serine-Threonine Kinases ,NPC1 ,business - Abstract
Niemann-Pick disease, type C1 (NPC1) is an inborn error of metabolism that results in endolysosomal accumulation of unesterified cholesterol. Clinically, NPC1 manifests as cholestatic liver disease in the newborn or as a progressive neurogenerative condition characterized by cerebellar ataxia and cognitive decline. Currently there are no FDA approved therapies for NPC1. Thus, understanding the pathological processes that contribute to neurodegeneration will be important in both developing and testing potential therapeutic interventions. Neuroinflammation and necroptosis contribute to the NPC1 pathological cascade. Receptor Interacting Protein Kinase 1 and 3 (RIPK1 and RIPK3), are protein kinases that play a central role in mediating neuronal necroptosis. Our prior work suggested that pharmacological inhibition of RIPK1 had a significant but modest beneficial effect; however, the inhibitors used in that study had suboptimal pharmacokinetic properties. In this work we evaluated both pharmacological and genetic inhibition of RIPK1 kinase activity. Lifespan in both Npc1(−/−)mice treated with GSK’547, a RIPK1 inhibitor with better pharmacokinetic properties, and Npc1(−/−) :Ripk1(kd/kd) double mutant mice was significantly increased. In both cases the increase in lifespan was modest, suggesting that the therapeutic potential of RIPK1 inhibition, as a monotherapy, is limited. We thus investigated the potential of combining RIPK1 inhibition with 2-hydroxypropyl-β-cyclodextrin (HPβCD) therapy HPβCD has been shown to slow neurological disease progression in NPC1 mice, cats and patients. HPpβCD appeared to have an additive positive effect on the pathology and survival of Npc1(−/−):Ripk1(kd/kd) mice. RIPK1 and RIPK3 are both critical components of the necrosome, thus we were surprised to observe no increase survival in Npc1(−/−);Ripk3(−/−) mice compared to Npc1(−/−) mice. These data suggest that although necroptosis is occurring in NPC1, the observed effects of RIPK1 inhibition may be related to its RIPK3-independent role in neuroinflammation and cytokine production.
- Published
- 2018
30. Stage-Dependent Impact of RIPK1 Inhibition on Atherogenesis: Dual Effects on Inflammation and Foam Cell Dynamics
- Author
-
Qing Wan, Hong Chen, John Bertin, Zekun Peng, Yonghu Huang, Brad J. Geddes, Yuze Zhang, Jean-Luc Tran, Michael Reilly, Guoli Yang, Miao Wang, Haojie Rao, and Huihui Li
- Subjects
Programmed cell death ,RIPK1 ,CD36 ,Inflammation ,Pharmacology ,Cardiovascular Medicine ,Macrophage ,Medicine ,Diseases of the circulatory (Cardiovascular) system ,Protein kinase A ,Receptor ,Foam cell ,Original Research ,biology ,business.industry ,Monocyte ,foam cells ,macrophages ,reverse cholesterol transport ,medicine.anatomical_structure ,cell death ,inflammation ,RC666-701 ,biology.protein ,lipids (amino acids, peptides, and proteins) ,medicine.symptom ,atherosclerosis ,Cardiology and Cardiovascular Medicine ,business - Abstract
Objective: Atherosclerosis is an arterial occlusive disease with hypercholesterolemia and hypertension as common risk factors. Advanced-stage stenotic plaque, which features inflammation and necrotic core formation, is the major reason for clinical intervention. Receptor interacting serine/threonine-protein kinase 1 (RIPK1) mediates inflammation and cell death and is expressed in atherosclerotic lesions. The role of RIPK1 in advanced-stage atherosclerosis is unknown.Approach and Results: To investigate the effect of RIPK1 inhibition in advanced atherosclerotic plaque formation, we used ApoESA/SA mice, which exhibit hypercholesterolemia, and develop angiotensin-II mediated hypertension upon administration of doxycycline in drinking water. These mice readily develop severe atherosclerosis, including that in coronary arteries. Eight-week-old ApoESA/SA mice were randomized to orally receive a highly selective RIPK1 inhibitor (RIPK1i, GSK547) mixed with a western diet, or control diet. RIPK1i administration reduced atherosclerotic plaque lesion area at 2 weeks of treatment, consistent with suppressed inflammation (MCP-1, IL-1β, TNF-α) and reduced monocyte infiltration. However, administration of RIPK1i unexpectedly exacerbated atherosclerosis at 4 weeks of treatment, concomitant with increased macrophages and lipid deposition in the plaques. Incubation of isolated macrophages with oxidized LDL resulted in foam cell formation in vitro. RIPK1i treatment promoted such foam cell formation while suppressing the death of these cells. Accordingly, RIPK1i upregulated the expression of lipid metabolism-related genes (Cd36, Ppara, Lxrα, Lxrb, Srebp1c) in macrophage foam cells with ABCA1/ABCG1 unaltered. Furthermore, RIPK1i treatment inhibited ApoA1 synthesis in the liver and reduced plasma HDL levels.Conclusion: RIPK1 modulates the development of atherosclerosis in a stage-dependent manner, implicating both pro-atherosclerotic (monocyte infiltration and inflammation) and anti-atherosclerotic effects (suppressing foam cell accumulation and promoting ApoA1 synthesis). It is critical to identify an optimal therapeutic duration for potential clinical use of RIPK1 inhibitor in atherosclerosis or other related disease indications.
- Published
- 2021
31. A diamidobenzimidazole STING agonist protects against SARS-CoV-2 infection
- Author
-
Sze-Ling Ng, René Maehr, Angel I. Morales-Ramos, Krishna-Mohan Parsi, Philip S. Landis, Liraz Shmuel-Galia, Zhaozhao Jiang, G. Scott Pesiridis, John Cruz, Ruth Wilson, Ramanjulu Joshi M, John Bertin, Katherine A. Fitzgerald, and Fiachra Humphries
- Subjects
0301 basic medicine ,viruses ,Immunology ,medicine.disease_cause ,Virus ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Medicine ,Research Articles ,Coronavirus ,Lung ,business.industry ,R-Articles ,Respiratory disease ,General Medicine ,medicine.disease ,Virology ,Sting ,030104 developmental biology ,medicine.anatomical_structure ,Viral replication ,business ,030217 neurology & neurosurgery ,Respiratory tract - Abstract
Coronaviruses are a family of RNA viruses that cause acute and chronic diseases of the upper and lower respiratory tract in humans and other animals. SARS-CoV-2 is a recently emerged coronavirus that has led to a global pandemic causing a severe respiratory disease known as COVID-19 with significant morbidity and mortality worldwide. The development of antiviral therapeutics are urgently needed while vaccine programs roll out worldwide. Here we describe a diamidobenzimidazole compound, diABZI-4, that activates STING and is highly effective in limiting SARS-CoV-2 replication in cells and animals. diABZI-4 inhibited SARS-CoV-2 replication in lung epithelial cells. Administration of diABZI-4 intranasally before or even after virus infection conferred complete protection from severe respiratory disease in K18-ACE2-transgenic mice infected with SARS-CoV-2. Intranasal delivery of diABZI-4 induced a rapid short-lived activation of STING, leading to transient proinflammatory cytokine production and lymphocyte activation in the lung associated with inhibition of viral replication. Our study supports the use of diABZI-4 as a host-directed therapy which mobilizes antiviral defenses for the treatment and prevention of COVID-19., Pharmacological activation of STING protects against SARS-CoV-2 infection.
- Published
- 2021
32. Protective Effects of Necrostatin-1 in Acute Pancreatitis: Partial Involvement of Receptor Interacting Protein Kinase 1
- Author
-
Mohammad Awais, Wei Huang, Wenhao Cai, John Bertin, Alexei V. Tepikin, Li Wen, Jane A Armstrong, Michael Chvanov, David N. Criddle, Rajarshi Mukherjee, Robert Sutton, Peter J. Gough, Diane Latawiec, and Yulin Ouyang
- Subjects
0301 basic medicine ,Male ,Programmed cell death ,Indoles ,RIPK1 ,acute pancreatitis ,indoleamine 2,3-dioxygenase ,QH301-705.5 ,Necroptosis ,necroptosis ,Acinar Cells ,Pharmacology ,Protective Agents ,Article ,Bile Acids and Salts ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Acinar cell ,Animals ,Indoleamine-Pyrrole 2,3,-Dioxygenase ,Biology (General) ,Protein kinase A ,Indoleamine 2,3-dioxygenase ,Pancreas ,Kinase ,Chemistry ,Imidazoles ,General Medicine ,medicine.disease ,epacadostat ,digestive system diseases ,Mice, Inbred C57BL ,receptor-interacting protein kinase 1 ,030104 developmental biology ,cell death ,Pancreatitis ,030220 oncology & carcinogenesis ,Alcohols ,Receptor-Interacting Protein Serine-Threonine Kinases ,Acute pancreatitis ,Calcium ,necrostatin-1 ,Reactive Oxygen Species ,Ceruletide - Abstract
Acute pancreatitis (AP) is a severe and potentially fatal disease caused predominantly by alcohol excess and gallstones, which lacks a specific therapy. The role of Receptor-Interacting Protein Kinase 1 (RIPK1), a key component of programmed necrosis (Necroptosis), is unclear in AP. We assessed the effects of RIPK1 inhibitor Necrostatin-1 (Nec-1) and RIPK1 modification (RIPK1K45A: kinase dead) in bile acid (TLCS-AP), alcoholic (FAEE-AP) and caerulein hyperstimulation (CER-AP) mouse models. Involvement of collateral Nec-1 target indoleamine 2,3-dioxygenase (IDO) was probed with the inhibitor Epacadostat (EPA). Effects of Nec-1 and RIPK1K45A were also compared on pancreatic acinar cell (PAC) fate in vitro and underlying mechanisms explored. Nec-1 markedly ameliorated histological and biochemical changes in all models. However, these were only partially reduced or unchanged in RIPK1K45A mice. Inhibition of IDO with EPA was protective in TLCS-AP. Both Nec-1 and RIPK1K45A modification inhibited TLCS- and FAEE-induced PAC necrosis in vitro. Nec-1 did not affect TLCS-induced Ca2+ entry in PACs, however, it inhibited an associated ROS elevation. The results demonstrate protective actions of Nec-1 in multiple models. However, RIPK1-dependent necroptosis only partially contributed to beneficial effects, and actions on targets such as IDO are likely to be important.
- Published
- 2021
33. Human rhinovirus promotes STING trafficking to replication organelles to promote viral replication
- Author
-
Martha Triantafilou, Joshi Ramanjulu, Lee M. Booty, Gisela Jimenez-Duran, Hakan Keles, Ken Saunders, Neysa Nevins, Emma Koppe, Louise K. Modis, G. Scott Pesiridis, John Bertin, and Kathy Triantafilou
- Subjects
Organelles ,Multidisciplinary ,Rhinovirus ,virus diseases ,General Physics and Astronomy ,Humans ,RNA Viruses ,General Chemistry ,Virus Replication ,eye diseases ,General Biochemistry, Genetics and Molecular Biology ,Enterovirus - Abstract
Human rhinovirus (HRV), like coronavirus (HCoV), are positive-strand RNA viruses that cause both upper and lower respiratory tract illness, with their replication facilitated by concentrating RNA-synthesizing machinery in intracellular compartments made of modified host membranes, referred to as replication organelles (ROs). Here we report a non-canonical, essential function for stimulator of interferon genes (STING) during HRV infections. While the canonical function of STING is to detect cytosolic DNA and activate inflammatory responses, HRV infection triggers the release of STIM1-bound STING in the ER by lowering Ca2+, thereby allowing STING to interact with phosphatidylinositol 4-phosphate (PI4P) and traffic to ROs to facilitates viral replication and transmission via autophagy. Our results thus hint a critical function of STING in HRV viral replication and transmission, with possible implications for other RO-mediated RNA viruses.
- Published
- 2020
34. Receptor-interacting protein 1 kinase inhibition therapeutically ameliorates experimental T cell-dependent colitis in mice
- Author
-
Gail Pearse, Isobel Harada, Tom Slocombe, Scott B. Berger, John Bertin, Allison M. Beal, Kathryn Fountain, Andrea C. Haynes, Alison Rowles, and Thomas Gobbetti
- Subjects
Cancer Research ,T cell ,T-Lymphocytes ,Immunology ,Mice, SCID ,Cellular and Molecular Neuroscience ,Mice ,medicine ,Animals ,Humans ,Colitis ,lcsh:QH573-671 ,Chemistry ,lcsh:Cytology ,Comment ,Cell Biology ,Chronic inflammation ,Kinase inhibition ,medicine.disease ,Disease Models, Animal ,medicine.anatomical_structure ,Pharmacodynamics ,Cancer research ,Female ,RECEPTOR-INTERACTING PROTEIN - Published
- 2020
35. RIPK1 gene variants associate with obesity in humans and can be therapeutically silenced to reduce obesity in mice
- Author
-
Anne-Claire Duchez, Markku Laakso, Kirsi H. Pietiläinen, Ludovic Boytard, Calvin Pan, David Smyth, Richard G. Lee, Marcus Alvarez, Joshua W. Kandiah, Peter J. Gough, Aldons J. Lusis, Peter Liu, Paulina Lau, Majid Nikpay, My-Anh Nguyen, Scott B. Berger, Denuja Karunakaran, Hailey Wyatt, Adil Rasheed, Barbara C. Vanderhyden, John Bertin, Michele Geoffrion, Sébastien Soubeyrand, Päivi Pajukanta, David P. Cook, Adam W. Turner, Katey J. Rayner, Bhama Ramkhelawon, Ruth McPherson, and Mary-Ellen Harper
- Subjects
Male ,Endocrinology, Diabetes and Metabolism ,Necroptosis ,Subcutaneous Fat ,Adipose tissue ,Mice, Obese ,Inflammation ,Genome-wide association study ,Biology ,FTO gene ,Article ,03 medical and health sciences ,RIPK1 ,Mice ,0302 clinical medicine ,Physiology (medical) ,Internal Medicine ,medicine ,Adipocytes ,Gene silencing ,Animals ,Humans ,Gene Silencing ,Obesity ,030304 developmental biology ,0303 health sciences ,Innate immune system ,Polymorphism, Genetic ,Cell Biology ,Glucose Tolerance Test ,3. Good health ,Mice, Inbred C57BL ,Basic-Leucine Zipper Transcription Factors ,Adipose Tissue ,Receptor-Interacting Protein Serine-Threonine Kinases ,Cancer research ,medicine.symptom ,Energy Metabolism ,030217 neurology & neurosurgery - Abstract
Obesity is a major public health burden worldwide and is characterized by chronic low-grade inflammation driven by the cooperation of the innate immune system and dysregulated metabolism in adipose tissue and other metabolic organs. Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is a central regulator of inflammatory cell function that coordinates inflammation, apoptosis and necroptosis in response to inflammatory stimuli. Here we show that genetic polymorphisms near the human RIPK1 locus associate with increased RIPK1 gene expression and obesity. We show that one of these single nucleotide polymorphisms is within a binding site for E4BP4 and increases RIPK1 promoter activity and RIPK1 gene expression in adipose tissue. Therapeutic silencing of RIPK1 in vivo in a mouse model of diet-induced obesity dramatically reduces fat mass, total body weight and improves insulin sensitivity, while simultaneously reducing macrophage and promoting invariant natural killer T cell accumulation in adipose tissue. These findings demonstrate that RIPK1 is genetically associated with obesity, and reducing RIPK1 expression is a potential therapeutic approach to target obesity and related diseases.
- Published
- 2020
36. RIPK1 Mediates TNF-Induced Intestinal Crypt Apoptosis During Chronic NF-κB Activation
- Author
-
P J Gough, Koji Taniguchi, Núria Planell, John Bertin, Soumita Das, Matija Zelic, Manolis Pasparakis, Philip A. Harris, Samuel B. Ho, Ricard Garcia-Carbonell, Shih Jing Yao, Joan Font-Burgada, Michelle A. Kelliher, Azucena Salas, Jerry Wong, Michael Karin, Monica Guma, Shalin A. Desai, and Brigid S. Boland
- Subjects
0301 basic medicine ,WT, wild type ,Indoles ,Ulcerative ,Apoptosis ,Crohn's Disease ,Oral and gastrointestinal ,IEC, intestinal epithelial cell ,Mice ,0302 clinical medicine ,Crohn Disease ,2.1 Biological and endogenous factors ,Gene Knock-In Techniques ,RNA-Seq ,Intestinal Mucosa ,Aetiology ,Intestinal Epithelial Cell ,TNF, tumor necrosis factor ,Cultured ,IBD, inflammatory bowel disease ,Cell Death ,Chemistry ,Kinase ,Caspase 3 ,Gastroenterology ,Imidazoles ,Colonoscopy ,Colitis ,3. Good health ,I-kappa B Kinase ,Cc-8, cleaved caspase-8 ,Organoids ,Receptor-Interacting Protein Serine-Threonine Kinases ,I.p., intraperitoneally ,LPS, lipopolysaccharide ,030211 gastroenterology & hepatology ,Tumor necrosis factor alpha ,Cc-3, cleaved caspase-3 ,NF, nuclear factor ,IHC, immunohistochemistry ,Adult ,Programmed cell death ,RIPK1 ,Colon ,Cells ,Knockout ,Primary Cell Culture ,IBD ,Ripoptosome ,PBS, phosphate-buffered saline ,Qpcr, quantitative polymerase chain reaction ,digestive system ,Autoimmune Disease ,03 medical and health sciences ,CHX, cycloheximide ,Ileum ,Animals ,Humans ,lcsh:RC799-869 ,Kinase activity ,IB, immunoblotting ,Protein kinase A ,Hepatology ,Animal ,Tumor Necrosis Factor-alpha ,Nec-1, necrostatin-1 ,Inflammatory Bowel Disease ,Transcription Factor RelA ,Epithelial Cells ,digestive system diseases ,UC, ulcerative colitis ,030104 developmental biology ,BHA, butylated hydroxyanisole ,DPI, phenyleneiodonium ,Disease Models ,Cancer research ,lcsh:Diseases of the digestive system. Gastroenterology ,CD, Crohn's disease ,Digestive Diseases - Abstract
Background and Aims: Tumor necrosis factor (TNF) is a major pathogenic effector and a therapeutic target in inflammatory bowel disease (IBD), yet the basis for TNF-induced intestinal epithelial cell (IEC) death is unknown, because TNF does not kill normal IECs. Here, we investigated how chronic nuclear factor (NF)- κB activation, which occurs in human IBD, promotes TNF-dependent IEC death in mice. Methods: Human IBD specimens were stained for p65 and cleaved caspase-3. C57BL/6 mice with constitutively active IKKβ in IEC (Ikkβ(EE)IEC), Ripk1D138N/D138N knockin mice, and Ripk3-/- mice were injected with TNF or lipopolysaccharide. Enteroids were also isolated from these mice and challenged with TNF with or without RIPK1 and RIPK3 inhibitors or butylated hydroxyanisole. Ripoptosome-mediated caspase-8 activation was assessed by immunoprecipitation. Results: NF-κB activation in human IBD correlated with appearance of cleaved caspase-3. Congruently, unlike normal mouse IECs that are TNF-resistant, IECs in Ikkβ(EE)IEC mice and enteroids were susceptible to TNF-dependent apoptosis, which depended on the protein kinase function of RIPK1. Constitutively active IKKβ facilitated ripoptosome formation, a RIPK1 signaling complex that mediates caspase-8 activation by TNF. Butylated hydroxyanisole treatment and RIPK1 inhibitors attenuated TNF-induced and ripoptosome-mediated caspase-8 activation and IEC death in vitro and in vivo. Conclusions: Contrary to common expectations, chronic NF-κB activation induced intestinal crypt apoptosis after TNF stimulation, resulting in severe mucosal erosion. RIPK1 kinase inhibitors selectively inhibited TNF destructive properties while preserving its survival and proliferative properties, which do not require RIPK1 kinase activity. RIPK1 kinase inhibition could be a potential treatment for IBD. Keywords: IBD, RIPK1, Intestinal Epithelial Cell, Ripoptosome, Cell Death
- Published
- 2020
37. t-BuOOH induces ferroptosis in human and murine cell lines
- Author
-
Peter J. Gough, Berenike Linz, Teodora Nikolova, Dagmar Faust, Peter Wipf, John Bertin, Christian Turmann, Anna Sophia Schröder, Christine Wenz, Stefan Krautwald, and Cornelia Dietrich
- Subjects
Keratinocytes ,0301 basic medicine ,Programmed cell death ,Cardiolipins ,Cell Survival ,Health, Toxicology and Mutagenesis ,Apoptosis ,Phenylenediamines ,Biology ,Toxicology ,medicine.disease_cause ,Cell Line ,Lipid peroxidation ,Mice ,03 medical and health sciences ,RIPK1 ,chemistry.chemical_compound ,tert-Butylhydroperoxide ,Quinoxalines ,medicine ,Animals ,Humans ,Spiro Compounds ,Protein kinase A ,Membrane Potential, Mitochondrial ,chemistry.chemical_classification ,Cyclohexylamines ,Reactive oxygen species ,Kinase ,Hydrogen Peroxide ,General Medicine ,Molecular biology ,Cell biology ,030104 developmental biology ,chemistry ,NIH 3T3 Cells ,Lipid Peroxidation ,Reactive Oxygen Species ,Oxidative stress - Abstract
Reactive oxygen species (ROS)-induced apoptosis has been extensively studied. Increasing evidence suggests that ROS, for instance, induced by hydrogen peroxide (H2O2), might also trigger regulated necrotic cell death pathways. Almost nothing is known about the cell death pathways triggered by tertiary-butyl hydroperoxide (t-BuOOH), a widely used inducer of oxidative stress. The lipid peroxidation products induced by t-BuOOH are involved in the pathophysiology of many diseases, such as cancer, cardiovascular diseases, or diabetes. In this study, we exposed murine fibroblasts (NIH3T3) or human keratinocytes (HaCaT) to t-BuOOH (50 or 200 μM, respectively) which induced a rapid necrotic cell death. Well-established regulators of cell death, i.e., p53, poly(ADP)ribose polymerase-1 (PARP-1), the stress kinases p38 and c-Jun N-terminal-kinases 1/2 (JNK1/2), or receptor-interacting serine/threonine protein kinase 1 (RIPK1) and 3 (RIPK3), were not required for t-BuOOH-mediated cell death. Using the selective inhibitors ferrostatin-1 (1 μM) and liproxstatin-1 (1 μM), we identified ferroptosis, a recently discovered cell death mechanism dependent on iron and lipid peroxidation, as the main cell death pathway. Accordingly, t-BuOOH exposure resulted in a ferrostatin-1- and liproxstatin-1-sensitive increase in lipid peroxidation and cytosolic ROS. Ferroptosis was executed independently from other t-BuOOH-mediated cellular damages, i.e., loss of mitochondrial membrane potential, DNA double-strand breaks, or replication block. H2O2 did not cause ferroptosis at equitoxic concentrations (300 μM) and induced a (1) lower and (2) ferrostatin-1- or liproxstatin-1-insensitive increase in lipid peroxidation. We identify that t-BuOOH and H2O2 produce a different pattern of lipid peroxidation, thereby leading to different cell death pathways and present t-BuOOH as a novel inducer of ferroptosis.
- Published
- 2017
38. Genetic Inhibition of Receptor Interacting Protein Kinase-1 Reduces Cell Death and Improves Functional Outcome After Intracerebral Hemorrhage in Mice
- Author
-
Lauren M. McAllister, Alexei Degterev, Yi Zheng, Joon Yong Chung, Limin Wu, Michael J. Whalen, Sevda Lule, John Bertin, Eng H. Lo, William J Edmiston, Emily Levy, and Peter J. Gough
- Subjects
0301 basic medicine ,Programmed cell death ,Necrosis ,Blotting, Western ,Morris water navigation task ,Apoptosis ,Pharmacology ,Article ,Mice ,03 medical and health sciences ,RIPK1 ,0302 clinical medicine ,medicine ,Animals ,Maze Learning ,Protein Kinase Inhibitors ,Cerebral Hemorrhage ,Neurons ,Advanced and Specialized Nursing ,Intracerebral hemorrhage ,Behavior, Animal ,Cell Death ,Kinase ,business.industry ,Point mutation ,Brain ,medicine.disease ,Immunohistochemistry ,030104 developmental biology ,Protein kinase domain ,Receptor-Interacting Protein Serine-Threonine Kinases ,Mutation ,Immunology ,Neurology (clinical) ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,030217 neurology & neurosurgery - Abstract
Background and Purpose— Recent studies using cultured cells and rodent intracerebral hemorrhage (ICH) models have implicated RIPK1 (receptor interacting protein kinase-1) as a driver of programmed necrosis and secondary injury based on use of chemical inhibitors. However, these inhibitors have off-target effects and cannot be used alone to prove a role for RIPK1. The aim of the current study was to examine the effect of genetic inhibition of the kinase domain of RIPK1 in a mouse ICH model. Methods— We subjected 2 lines of mice with RIPK1 point mutations of the kinase domain (K45A and D138N), rendering them kinase inactive, to autologous blood ICH and measured acute cell death and functional outcome. Results— Compared with wild-type controls, RIPK1 K45A/K45A and RIPK1 D138N/D138N had significantly less cells with plasmalemma permeability, less acute neuronal cell death, less weight loss and more rapid weight gain to baseline, and improved performance in a Morris water maze paradigm after autologous blood ICH. In addition, mice systemically administered GSK′963, a potent, specific, brain penetrant small molecule RIPK1 inhibitor, had reduced acute neuronal death at 24 hours after ICH. Conclusions— The data show that the kinase domain of RIPK1 is a disease driver of ICH, mediating both acute cell death and functional outcome, and support development of RIPK1 inhibitors as therapeutic agents for human ICH.
- Published
- 2017
39. RIPK1-dependent apoptosis bypasses pathogen blockade of innate signaling to promote immune defense
- Author
-
Baofeng Hu, Igor E. Brodsky, Ruth Choa, Kendra Asklof, Elisabet Bjanes, Elisabeth L. Buza, Alexandra DeLaney, Falon Gray, Lance W. Peterson, Meghan A. Wynosky-Dolfi, Christopher P. Dillon, Peter J. Gough, Douglas R. Green, Naomi H. Philip, Scott B. Berger, and John Bertin
- Subjects
0301 basic medicine ,Cell signaling ,Programmed cell death ,MAP Kinase Signaling System ,Immunology ,Yersinia pseudotuberculosis Infections ,Apoptosis ,Biology ,03 medical and health sciences ,RIPK1 ,Immunity ,Animals ,Immunology and Allergy ,Kinase activity ,Research Articles ,Disease Resistance ,Mice, Knockout ,Innate immune system ,Effector ,Macrophages ,Brief Definitive Report ,Models, Immunological ,NF-kappa B ,biochemical phenomena, metabolism, and nutrition ,Survival Analysis ,Immunity, Innate ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Yersinia pseudotuberculosis ,Receptor-Interacting Protein Serine-Threonine Kinases ,Host-Pathogen Interactions ,Cytokines ,Signal transduction ,Signal Transduction - Abstract
RIPK1 regulates cytokine signaling and cell death during infection and inflammation. Peterson et al. show that RIPK1 kinase activity triggers apoptosis in response to bacterial pathogen blockade of innate immune signaling and that this pathway of effector-triggered immunity is critical for a successful antibacterial response., Many pathogens deliver virulence factors or effectors into host cells in order to evade host defenses and establish infection. Although such effector proteins disrupt critical cellular signaling pathways, they also trigger specific antipathogen responses, a process termed “effector-triggered immunity.” The Gram-negative bacterial pathogen Yersinia inactivates critical proteins of the NF-κB and MAPK signaling cascade, thereby blocking inflammatory cytokine production but also inducing apoptosis. Yersinia-induced apoptosis requires the kinase activity of receptor-interacting protein kinase 1 (RIPK1), a key regulator of cell death, NF-κB, and MAPK signaling. Through the targeted disruption of RIPK1 kinase activity, which selectively disrupts RIPK1-dependent cell death, we now reveal that Yersinia-induced apoptosis is critical for host survival, containment of bacteria in granulomas, and control of bacterial burdens in vivo. We demonstrate that this apoptotic response provides a cell-extrinsic signal that promotes optimal innate immune cytokine production and antibacterial defense, demonstrating a novel role for RIPK1 kinase–induced apoptosis in mediating effector-triggered immunity to circumvent pathogen inhibition of immune signaling.
- Published
- 2017
40. Kinase Activities of RIPK1 and RIPK3 Can Direct IFN-β Synthesis Induced by Lipopolysaccharide
- Author
-
Alexei Degterev, Matija Zelic, Siddharth Balachandran, Michelle A. Kelliher, Nikolai Slavov, Danish Saleh, Saumil Shah, Katherine A. Fitzgerald, Apostolos Polykratis, Peter J. Gough, Malek Najjar, Manolis Pasparakis, Joan Mecsas, John Bertin, Michelle K. Paczosa, Shoko Nogusa, and Michael J. Whalen
- Subjects
Lipopolysaccharides ,0301 basic medicine ,Lipopolysaccharide ,Necroptosis ,Immunology ,Apoptosis ,Biology ,Article ,Mice ,Necrosis ,03 medical and health sciences ,RIPK1 ,chemistry.chemical_compound ,0302 clinical medicine ,Klebsiella ,Gram-Negative Bacteria ,Animals ,Immunology and Allergy ,Phosphorylation ,Central element ,Innate immune system ,Kinase ,Effector ,Macrophages ,Interferon-beta ,Yersinia ,Cell biology ,Toll-Like Receptor 4 ,030104 developmental biology ,chemistry ,Receptor-Interacting Protein Serine-Threonine Kinases ,030220 oncology & carcinogenesis ,TLR4 - Abstract
The innate immune response is a central element of the initial defense against bacterial and viral pathogens. Macrophages are key innate immune cells that upon encountering pathogen-associated molecular patterns respond by producing cytokines, including IFN-β. In this study, we identify a novel role for RIPK1 and RIPK3, a pair of homologous serine/threonine kinases previously implicated in the regulation of necroptosis and pathologic tissue injury, in directing IFN-β production in macrophages. Using genetic and pharmacologic tools, we show that catalytic activity of RIPK1 directs IFN-β synthesis induced by LPS in mice. Additionally, we report that RIPK1 kinase–dependent IFN-β production may be elicited in an analogous fashion using LPS in bone marrow–derived macrophages upon inhibition of caspases. Notably, this regulation requires kinase activities of both RIPK1 and RIPK3, but not the necroptosis effector protein, MLKL. Mechanistically, we provide evidence that necrosome-like RIPK1 and RIPK3 aggregates facilitate canonical TRIF–dependent IFN-β production downstream of the LPS receptor TLR4. Intriguingly, we also show that RIPK1 and RIPK3 kinase–dependent synthesis of IFN-β is markedly induced by avirulent strains of Gram-negative bacteria, Yersinia and Klebsiella, and less so by their wild-type counterparts. Overall, these observations identify unexpected roles for RIPK1 and RIPK3 kinases in the production of IFN-β during the host inflammatory responses to bacterial infection and suggest that the axis in which these kinases operate may represent a target for bacterial virulence factors.
- Published
- 2017
41. Discovery of a First-in-Class Receptor Interacting Protein 1 (RIP1) Kinase Specific Clinical Candidate (GSK2982772) for the Treatment of Inflammatory Diseases
- Author
-
Xiaoyang Dong, Anna Vossenkämper, Attiq Rahman, Viera Kasparcova, Robert Singhaus, Joshua N. Finger, Bryan W. King, Patrick M. Eidam, Michael Reilly, David D. Wisnoski, Scott B. Berger, John D. Lich, James Kang, Michael T. Ouellette, Lauren Dare, Philip A. Harris, Julie A. Cox, Nathan A. Miller, John Bertin, Michelle C. Schaeffer, Deepak Bandyopadhyay, Daohua Zhang, Alan R. Rendina, Clark A. Sehon, Sandra J. Hoffman, Peter J. Gough, Rachel D. Totoritis, Thomas T. MacDonald, Yunfeng Lan, Paris Ward, Barbara A. Swift, Robert W. Marquis, Lara Kathryn Leister, Rakesh Nagilla, Elizabeth J. Rivera, Helen H. Sun, Carol A. Capriotti, Nino Campobasso, Ruth Lehr, Christina S. Pao, and Jae U. Jeong
- Subjects
0301 basic medicine ,Swine ,Necroptosis ,Anti-Inflammatory Agents ,Inflammation ,Pharmacology ,Mice ,03 medical and health sciences ,Dogs ,0302 clinical medicine ,Psoriasis ,Drug Discovery ,medicine ,Animals ,Humans ,Potency ,Protein Kinase Inhibitors ,Tumor Necrosis Factor-alpha ,Chemistry ,Kinase ,Haplorhini ,Benzazepines ,medicine.disease ,Ulcerative colitis ,Rats ,Molecular Docking Simulation ,030104 developmental biology ,Receptor-Interacting Protein Serine-Threonine Kinases ,030220 oncology & carcinogenesis ,Rheumatoid arthritis ,Cytokines ,Swine, Miniature ,Molecular Medicine ,Colitis, Ulcerative ,Rabbits ,medicine.symptom ,RECEPTOR-INTERACTING PROTEIN - Abstract
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.
- Published
- 2017
42. Discovery of Pyrazolocarboxamides as Potent and Selective Receptor Interacting Protein 2 (RIP2) Kinase Inhibitors
- Author
-
Arthur Shu, Yiqian Lian, Huijie Li, Curt Dale Haffner, Terry Vincent Hughes, Ami S. Lakdawala, Julie A. Cox, Pamela A. Haile, Constantine Kreatsoulas, Beth Anne Knapp-Reed, Lisa M. Shewchuk, David B. Lipshutz, John F. Mehlmann, Michael T. Ouellette, Christopher J Aquino, Mark Elban, Nicole Cathleen Goodwin, Huiqiang Zhou, Adam Kenneth Charnley, Bartholomew J. Votta, Robert W. Marquis, Joseph J. Romano, Linda N. Casillas, Peter J. Gough, Máire A. Convery, and John Bertin
- Subjects
Bicyclic molecule ,010405 organic chemistry ,Kinase ,Chemistry ,Organic Chemistry ,Pyrazole ,01 natural sciences ,Biochemistry ,0104 chemical sciences ,RIPK2 ,010404 medicinal & biomolecular chemistry ,chemistry.chemical_compound ,NOD2 ,Drug Discovery ,NOD1 ,Transferase ,RECEPTOR-INTERACTING PROTEIN - Abstract
[Image: see text] Herein we report the discovery of pyrazolocarboxamides as novel, potent, and kinase selective inhibitors of receptor interacting protein 2 kinase (RIP2). Fragment based screening and design principles led to the identification of the inhibitor series, and X-ray crystallography was used to inform key structural changes. Through key substitutions about the N1 and C5 N positions on the pyrazole ring significant kinase selectivity and potency were achieved. Bridged bicyclic pyrazolocarboxamide 11 represents a selective and potent inhibitor of RIP2 and will allow for a more detailed investigation of RIP2 inhibition as a therapeutic target for autoinflammatory disorders.
- Published
- 2019
43. Identification of a RIP1 Kinase Inhibitor Clinical Candidate (GSK3145095) for the Treatment of Pancreatic Cancer
- Author
-
Joshua N. Finger, Michael T. Ouellette, Helen H. Sun, Robert W. Marquis, Viera Kasparcova, James H. Thorpe, Scott B. Berger, Philip A. Harris, Rakesh Nagilla, George Miller, Anirudh Chirala, James Kang, Elizabeth J. Rivera, John Bertin, Peter J. Gough, Mukesh K. Mahajan, Daohua Zhang, John D. Lich, Julie A. Cox, Wei Wang, Lara Kathryn Leister, Jill M. Marinis, Rachel D. Totoritis, Patrick M. Eidam, Michael Reilly, Jae U. Jeong, Dongling Wu, and Alan R. Rendina
- Subjects
Programmed cell death ,T cell ,type III kinase inhibitors ,pancreatic cancer ,Inflammation ,01 natural sciences ,Biochemistry ,Pancreatic cancer ,Drug Discovery ,medicine ,RIP1 ,010405 organic chemistry ,Kinase ,business.industry ,Organic Chemistry ,Cancer ,medicine.disease ,Phenotype ,Featured Letter ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,medicine.anatomical_structure ,Cancer research ,Adenocarcinoma ,medicine.symptom ,business - Abstract
RIP1 regulates cell death and inflammation and is believed to play an important role in contributing to a variety of human pathologies, including immune-mediated inflammatory diseases and cancer. While small-molecule inhibitors of RIP1 kinase have been advanced to the clinic for inflammatory diseases and CNS indications, RIP1 inhibitors for oncology indications have yet to be described. Herein we report on the discovery and profile of GSK3145095 (compound 6). Compound 6 potently binds to RIP1 with exquisite kinase specificity and has excellent activity in blocking RIP1 kinase-dependent cellular responses. Highlighting its potential as a novel cancer therapy, the inhibitor was also able to promote a tumor suppressive T cell phenotype in pancreatic adenocarcinoma organ cultures. Compound 6 is currently in phase 1 clinical studies for pancreatic adenocarcinoma and other selected solid tumors.
- Published
- 2019
44. RIPK1 and Caspase-8 Ensure Chromosome Stability Independently of Their Role in Cell Death and Inflammation
- Author
-
Peter J. Gough, David Robertson, Nieves Peltzer, Luca L. Fava, Holly Anderton, Manolis Pasparakis, Alexey I. Nesvizhskii, Katiuscia Bianchi, Marius Dannappel, Timo Glatter, Philip A. Harris, Tencho Tenev, Arnaud J. Legrand, Alexander Schmidt, Andreas Villunger, Gianmaria Liccardi, Pascal Meier, Manuel D. Haschka, John Silke, Alessandro Annibaldi, Maurice Darding, Hannah Schünke, Rebecca Feltham, John Bertin, and Laura Ramos Garcia
- Subjects
Fas-Associated Death Domain Protein ,CASP8 and FADD-Like Apoptosis Regulating Protein ,Apoptosis ,Cell Cycle Proteins ,Inbred C57BL ,caspase-8 ,Chromosome segregation ,Mice ,0302 clinical medicine ,Chromosome Segregation ,Phosphorylation ,Tissue homeostasis ,Mice, Knockout ,0303 health sciences ,Caspase 8 ,Protein-Serine-Threonine Kinases ,3. Good health ,Cell biology ,cell death ,Receptor-Interacting Protein Serine-Threonine Kinases ,Colonic Neoplasms ,cell cycle ,PLK1 ,HT29 Cells ,Ripoptosome assembly ,Signal Transduction ,RIPK1 ,chromosomal instability ,Necroptosis ,Knockout ,Ripoptosome ,Mitosis ,Protein Serine-Threonine Kinases ,Biology ,Article ,03 medical and health sciences ,BUBR1 ,cancer ,mitosis ,ripoptosome ,Aneuploidy ,Animals ,Fibroblasts ,Humans ,Inflammation ,Mice, Inbred C57BL ,Proto-Oncogene Proteins ,Chromosomal Instability ,Molecular Biology ,Metaphase ,030304 developmental biology ,Cell Biology ,030217 neurology & neurosurgery - Abstract
Summary Receptor-interacting protein kinase (RIPK) 1 functions as a key mediator of tissue homeostasis via formation of Caspase-8 activating ripoptosome complexes, positively and negatively regulating apoptosis, necroptosis, and inflammation. Here, we report an unanticipated cell-death- and inflammation-independent function of RIPK1 and Caspase-8, promoting faithful chromosome alignment in mitosis and thereby ensuring genome stability. We find that ripoptosome complexes progressively form as cells enter mitosis, peaking at metaphase and disassembling as cells exit mitosis. Genetic deletion and mitosis-specific inhibition of Ripk1 or Caspase-8 results in chromosome alignment defects independently of MLKL. We found that Polo-like kinase 1 (PLK1) is recruited into mitotic ripoptosomes, where PLK1’s activity is controlled via RIPK1-dependent recruitment and Caspase-8-mediated cleavage. A fine balance of ripoptosome assembly is required as deregulated ripoptosome activity modulates PLK1-dependent phosphorylation of downstream effectors, such as BUBR1. Our data suggest that ripoptosome-mediated regulation of PLK1 contributes to faithful chromosome segregation during mitosis., Graphical Abstract, Highlights • A RIPK1/FADD/Caspase-8/cFLIP complex (ripoptosome) forms during mitosis • The mitotic kinase PLK1 binds to RIPK1 and is recruited into mitotic ripoptosomes • PLK1 is cleaved by Caspase-8 in a RIPK1-dependent manner • Deletion and inhibition of RIPK1 or Caspase-8 lead to chromosomal instability, Liccardi et al. show that ripoptosome complexes assemble during physiological mitosis and regulate PLK1 activity via RIPK1-dependent recruitment and Caspase-8-mediated cleavage. These results reveal a direct role for RIPK1 and Caspase-8 in controlling chromosome segregation during mitosis.
- Published
- 2019
45. MALT1 Protease Activation Triggers Acute Disruption of Endothelial Barrier Integrity via CYLD Cleavage
- Author
-
Dong Hu, Peter C. Lucas, Emily M. Hess, Linda R. Klei, Katherine Oravecz-Wilson, Kevin Foley, Linda M. McAllister-Lucas, Vincent J. Concel, Robert L. Panek, Rachel E. Bridwell, Matthew Van Beek, Simon C. Watkins, Phillip C. Delekta, Peter J. Gough, Shufang Gu, Adrian T. Ting, Kelly M. Bailey, Danielle N. Alfano, and John Bertin
- Subjects
0301 basic medicine ,Microtubules ,NF-κB ,Mice ,chemistry.chemical_compound ,CARMA3 ,lcsh:QH301-705.5 ,NF-kappa B ,thrombin ,BCL10 ,Deubiquitinating Enzyme CYLD ,I-kappa B Kinase ,Neoplasm Proteins ,3. Good health ,Cell biology ,Endothelial stem cell ,Cysteine Endopeptidases ,Caspases ,Paracellular transport ,endothelial permeability ,medicine.symptom ,Protease activated receptor-1 (PAR1) ,Signal Transduction ,medicine.drug ,Primary Cell Culture ,CYLD ,G protein-coupled receptor (GPCR) ,Mice, Transgenic ,Inflammation ,Biology ,Article ,Permeability ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Thrombin ,Immune system ,medicine ,Animals ,Receptor, PAR-1 ,CARD10 ,Bcl10 ,Endothelial Cells ,Biological Transport ,CARD Signaling Adaptor Proteins ,MALT1 ,030104 developmental biology ,Gene Expression Regulation ,lcsh:Biology (General) ,chemistry ,Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein ,MALT1 protease - Abstract
SummaryMicrovascular endothelial cells maintain a tight barrier to prevent passage of plasma and circulating immune cells into the extravascular tissue compartment, yet endothelial cells respond rapidly to vasoactive substances, including thrombin, allowing transient paracellular permeability. This response is a cornerstone of acute inflammation, but the mechanisms responsible are still incompletely understood. Here, we demonstrate that thrombin triggers MALT1 to proteolytically cleave cylindromatosis (CYLD). Fragmentation of CYLD results in microtubule disruption and a cascade of events leading to endothelial cell retraction and an acute permeability response. This finding reveals an unexpected role for the MALT1 protease, which previously has been viewed mostly as a driver of pro-inflammatory NF-κB signaling in lymphocytes. Thus, MALT1 not only promotes immune cell activation but also acutely regulates endothelial cell biology, actions that together facilitate tissue inflammation. Pharmacologic inhibition of MALT1 may therefore have synergistic impact by targeting multiple disparate steps in the overall inflammatory response.
- Published
- 2016
46. RIPK3 Activates Parallel Pathways of MLKL-Driven Necroptosis and FADD-Mediated Apoptosis to Protect against Influenza A Virus
- Author
-
Stuart C. Sealfon, Christopher P. Dillon, Swantje Liedmann, Douglas R. Green, Shalini Sharma, Roshan J. Thapa, Justin P. Ingram, Andrew Oberst, Carolina B. López, Peter J. Gough, Rachelle Kosoff, Edward S. Mocarski, Siddharth Balachandran, Boris M. Hartmann, Shoko Nogusa, Paul G. Thomas, William J. Kaiser, Katherine Verbist, Thomas H. Oguin, John Bertin, Oliver E. Sturm, and Diego A. Rodriguez
- Subjects
0301 basic medicine ,Fas-Associated Death Domain Protein ,Necroptosis ,Apoptosis ,Biology ,Microbiology ,Article ,Cell Line ,Mice ,Necrosis ,03 medical and health sciences ,RIPK1 ,0302 clinical medicine ,Orthomyxoviridae Infections ,Virology ,Animals ,Humans ,FADD ,Kinase activity ,Mice, Knockout ,Innate immune system ,Kinase ,Signal transducing adaptor protein ,Epithelial Cells ,Fibroblasts ,Protein kinase R ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,Influenza A virus ,Receptor-Interacting Protein Serine-Threonine Kinases ,biology.protein ,Parasitology ,Protein Multimerization ,Protein Kinases ,030215 immunology - Abstract
Influenza A virus (IAV) is a lytic virus in primary cultures of many cell types and in vivo. We report that the kinase RIPK3 is essential for IAV-induced lysis of mammalian fibroblasts and lung epithelial cells. Replicating IAV drives assembly of a RIPK3-containing complex that includes the kinase RIPK1, the pseudokinase MLKL, and the adaptor protein FADD, and forms independently of signaling by RNA-sensing innate immune receptors (RLRs, TLRs, PKR), or the cytokines type I interferons and TNF-α. Downstream of RIPK3, IAV activates parallel pathways of MLKL-driven necroptosis and FADD-mediated apoptosis, with the former reliant on RIPK3 kinase activity and neither on RIPK1 activity. Mice deficient in RIPK3 or doubly deficient in MLKL and FADD, but not MLKL alone, are more susceptible to IAV than their wild-type counterparts, revealing an important role for RIPK3-mediated apoptosis in antiviral immunity. Collectively, these results outline RIPK3-activated cytolytic mechanisms essential for controlling respiratory IAV infection.
- Published
- 2016
47. RIPK1 and RIPK3 Kinases Promote Cell-Death-Independent Inflammation by Toll-like Receptor 4
- Author
-
Michael J. Whalen, Michelle A. Kelliher, Peter J. Gough, Danish Saleh, John Bertin, Saumil Shah, Apostolos Polykratis, Siddharth Balachandran, Alexander Poltorak, Malek Najjar, Manolis Pasparakis, Albert K. Tai, Alexei Degterev, Matija Zelic, Shoko Nogusa, and Joshua N. Finger
- Subjects
Lipopolysaccharides ,0301 basic medicine ,MAPK/ERK pathway ,Immunology ,Mice ,Necrosis ,03 medical and health sciences ,RIPK1 ,Animals ,Immunology and Allergy ,Cells, Cultured ,Caspase ,Inflammation ,Mice, Knockout ,Caspase 8 ,Mice, Inbred BALB C ,Toll-like receptor ,Innate immune system ,biology ,Kinase ,Macrophages ,Immunity, Innate ,Cell biology ,Mice, Inbred C57BL ,Adaptor Proteins, Vesicular Transport ,030104 developmental biology ,Infectious Diseases ,TRIF ,Receptor-Interacting Protein Serine-Threonine Kinases ,Cancer research ,biology.protein ,Female ,Signal transduction ,Transcriptome ,Signal Transduction - Abstract
Macrophages are a crucial component of the innate immune system in sensing pathogens and promoting local and systemic inflammation. RIPK1 and RIPK3 are homologous kinases, previously linked to activation of necroptotic death. In this study, we have described roles for these kinases as master regulators of pro-inflammatory gene expression induced by lipopolysaccharide, independent of their well-documented cell death functions. In primary macrophages, this regulation was elicited in the absence of caspase-8 activity, required the adaptor molecule TRIF, and proceeded in a cell autonomous manner. RIPK1 and RIPK3 kinases promoted sustained activation of Erk, cFos, and NF-κB, which were required for inflammatory changes. Utilizing genetic and pharmacologic tools, we showed that RIPK1 and RIPK3 account for acute inflammatory responses induced by lipopolysaccharide in vivo; notably, this regulation did not require exogenous manipulation of caspases. These findings identified a new pharmacologically accessible pathway that may be relevant to inflammatory pathologies.
- Published
- 2016
48. RIPK1 and PGAM5 Control Leishmania Replication through Distinct Mechanisms
- Author
-
Kendi Okuda, Valéria M. Borges, Sakthi Balaji, Aline Silva Barreto, Ricardo T. Gazzinelli, Francis Ka-Ming Chan, Roque P. Almeida, John Bertin, Marcelo T. Bozza, Nivea Farias Luz, and Peter J. Gough
- Subjects
0301 basic medicine ,Programmed cell death ,Necroptosis ,Interleukin-1beta ,Immunology ,Heme ,Nitric Oxide ,Article ,Host-Parasite Interactions ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,RIPK1 ,0302 clinical medicine ,Phosphoprotein Phosphatases ,Animals ,Humans ,Immunology and Allergy ,Kinase activity ,Leishmaniasis ,Leishmania ,Cell Death ,biology ,Kinase ,Effector ,Macrophages ,biology.organism_classification ,Cell biology ,030104 developmental biology ,chemistry ,Receptor-Interacting Protein Serine-Threonine Kinases ,Leishmaniasis, Visceral ,030215 immunology - Abstract
Leishmaniasis is an important parasitic disease found in the tropics and subtropics. Cutaneous and visceral leishmaniasis affect an estimated 1.5 million people worldwide. Despite its human health relevance, relatively little is known about the cell death pathways that control Leishmania replication in the host. Necroptosis is a recently identified form of cell death with potent antiviral effects. Receptor interacting protein kinase 1 (RIPK1) is a critical kinase that mediates necroptosis downstream of death receptors and TLRs. Heme, a product of hemoglobin catabolism during certain intracellular pathogen infections, is also a potent inducer of macrophage necroptosis. We found that human visceral leishmaniasis patients exhibit elevated serum levels of heme. Therefore, we examined the impact of heme and necroptosis on Leishmania replication. Indeed, heme potently inhibited Leishmania replication in bone marrow–derived macrophages. Moreover, we found that inhibition of RIPK1 kinase activity also enhanced parasite replication in the absence of heme. We further found that the mitochondrial phosphatase phosphoglycerate mutase family member 5 (PGAM5), a putative downstream effector of RIPK1, was also required for inhibition of Leishmania replication. In mouse infection, both PGAM5 and RIPK1 kinase activity are required for IL-1β expression in response to Leishmania. However, PGAM5, but not RIPK1 kinase activity, was directly responsible for Leishmania-induced IL-1β secretion and NO production in bone marrow–derived macrophages. Collectively, these results revealed that RIPK1 and PGAM5 function independently to exert optimal control of Leishmania replication in the host.
- Published
- 2016
49. K45A mutation of RIPK1 results in poor necroptosis and cytokine signaling in macrophages, which impacts inflammatory responses in vivo
- Author
-
John Bertin, Michael G. Schlossmacher, Subash Sad, Bojan Shutinoski, Dikchha Rijal, Norah A. Alturki, and P J Gough
- Subjects
Salmonella typhimurium ,0301 basic medicine ,Programmed cell death ,Necroptosis ,Apoptosis ,Caspase 8 ,Inhibitor of Apoptosis Proteins ,Mice ,Necrosis ,03 medical and health sciences ,RIPK1 ,Animals ,Phosphorylation ,Molecular Biology ,Caspase ,Inflammation ,Original Paper ,biology ,Lysine ,Macrophages ,JNK Mitogen-Activated Protein Kinases ,Cell Biology ,Shock, Septic ,Cell biology ,Endotoxins ,STAT1 Transcription Factor ,030104 developmental biology ,Receptors, Tumor Necrosis Factor, Type I ,Receptor-Interacting Protein Serine-Threonine Kinases ,Mutation ,Cancer research ,biology.protein ,Cytokines ,Tumor necrosis factor alpha ,Protein Multimerization ,Signal transduction ,Cell activation ,Protein Kinases ,Signal Transduction - Abstract
Receptor interacting protein kinase 1 (RIPK1) participates in several cell signaling complexes that promote cell activation and cell death. Stimulation of RIPK1 in the absence of caspase signaling induces regulated necrosis (necroptosis), which promotes an inflammatory response. Understanding of the mechanisms through which RIPK1 promotes inflammation has been unclear. Herein we have evaluated the impact of a K45A mutation of RIPK1 on necroptosis of macrophages and the activation of inflammatory response. We show that K45A mutation of RIPK1 results in attenuated necroptosis of macrophages in response to stimulation with LPS, TNFα and IFNβ in the absence of caspase signaling. Impairment in necroptosis correlated with poor phosphorylation of RIPK1, RIPK3 and reduced trimerization of MLKL. Furthermore, K45A mutation of RIPK1 resulted in poor STAT1 phosphorylation (at S727) and expression of RANTES and MIP-1α following TNF-R engagement in the absence of caspase activation. Our results further indicate that in the absence of stimulation by pathogen-associated molecular patterns (PAMPs), cellular inhibitors of apoptotic proteins (cIAPs) prevent the K45-dependent auto-phosphorylation of RIPK1, leading to resistance against necroptosis. Finally, RIPK1(K45A) mice displayed attenuated inflammatory response in vivo as they were significantly resistant against endotoxin shock, but highly susceptible against a challenge with Salmonella typhimurium. This correlated with reduced expression of IL-1β and ROS, and poor processing of caspase 8 by RIPK1(K45A) macrophages. Overall, these results indicate that K45 mediated kinase activity of RIPK1 is not only important for necroptosis but it also has a key role in promoting cytokine signaling and host response to inflammatory stimuli.
- Published
- 2016
50. The Mitochondrial Phosphatase PGAM5 Is Dispensable for Necroptosis but Promotes Inflammasome Activation in Macrophages
- Author
-
Carey L. O'Donnell, Raymond M. Welsh, Maria Jose de Rosa, Francis Ka-Ming Chan, Nivea Farias Luz, Peter J. Gough, Sakthi Balaji, Kenta Moriwaki, and John Bertin
- Subjects
Male ,0301 basic medicine ,Inflammasomes ,Fas-Associated Death Domain Protein ,Interleukin-1beta ,Apoptosis ,Mice ,Phosphoprotein Phosphatases ,Immunology and Allergy ,FADD ,MACROPHAGES ,Cells, Cultured ,Mice, Knockout ,Caspase 8 ,biology ,Caspase 1 ,Inflammasome ,Mitochondria ,Cell biology ,DNA-Binding Proteins ,Receptor-Interacting Protein Serine-Threonine Kinases ,CIENCIAS NATURALES Y EXACTAS ,PGAM5 ,Signal Transduction ,medicine.drug ,Programmed cell death ,Otras Ciencias Biológicas ,Necroptosis ,Immunology ,Article ,Vesicular stomatitis Indiana virus ,Ciencias Biológicas ,03 medical and health sciences ,RIPK1 ,AIM2 ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Animals ,NECROPTOSIS ,Inflammation ,Macrophages ,Dendritic Cells ,Phosphoric Monoester Hydrolases ,Mice, Inbred C57BL ,030104 developmental biology ,biology.protein ,Carrier Proteins ,Reactive Oxygen Species - Abstract
The cytokine IL-1β is intimately linked to many pathological inflammatory conditions. Mature IL-1β secretion requires cleavage by the inflammasome. Recent evidence indicates that many cell death signal adaptors have regulatory roles in inflammasome activity. These include the apoptosis inducers FADD and caspase 8, and the necroptosis kinases receptor interacting protein kinase 1 (RIPK1) and RIPK3. PGAM5 is a mitochondrial phosphatase that has been reported to function downstream of RIPK3 to promote necroptosis and IL-1β secretion. To interrogate the biological function of PGAM5, we generated Pgam5−/− mice. We found that Pgam5−/− mice were smaller compared with wild type littermates, and male Pgam5−/− mice were born at sub-Mendelian ratio. Despite these growth and survival defects, Pgam5−/− cells responded normally to multiple inducers of apoptosis and necroptosis. Rather, we found that PGAM5 is critical for IL-1β secretion in response to NLRP3 and AIM2 inflammasome agonists. Moreover, vesicular stomatosis virus–induced IL-1β secretion was impaired in Pgam5−/− bone marrow–derived macrophages, but not in Ripk3−/− bone marrow–derived dendritic cells, indicating that PGAM5 functions independent of RIPK3 to promote inflammasome activation. Mechanistically, PGAM5 promotes ASC polymerization, maintenance of mitochondrial integrity, and optimal reactive oxygen species production in response to inflammasome signals. Hence PGAM5 is a novel regulator of inflammasome and caspase 1 activity that functions independently of RIPK3. Fil: Moriwaki, Kenta. University of Massachussets; Estados Unidos Fil: Farias Luz, Nivea. Universidade Federal da Bahia; Brasil. University of Massachussets; Estados Unidos Fil: Balaji, Sakthi. University of Massachussets; Estados Unidos Fil: de Rosa, Maria Jose. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro Científico Tecnológico Conicet - Bahía Blanca. Instituto de Investigaciones Bioquímicas de Bahía Blanca. Universidad Nacional del Sur. Instituto de Investigaciones Bioquímicas de Bahía Blanca; Argentina. Universidad Nacional del Sur. Departamento de Biología, Bioquímica y Farmacia; Argentina Fil: O'Donnell, Carey L.. University of Massachussets; Estados Unidos Fil: Gough, Peter J.. Glaxo Smith Kline; Estados Unidos Fil: Bertin, John. Glaxo Smith Kline; Estados Unidos Fil: Welsh, Raymond M.. University of Massachussets; Estados Unidos Fil: Chan, Francis Ka Ming. University of Massachussets; Estados Unidos
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.