Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Cell Death Dis ; 14(11): 755, 2023 11 18.
Article in English | MEDLINE | ID: mdl-37980412

ABSTRACT

Plasma membrane permeabilization (PMP) is a defining feature of regulated necrosis. It allows the extracellular release of damage-associated molecular patterns (DAMPs) that trigger sterile inflammation. The pore forming molecules MLKL and GSDMs drive PMP in necroptosis and pyroptosis, respectively, but the process of PMP remains unclear in many other forms of regulated necrosis. Here, we identified NINJ1 as a crucial regulator of PMP and consequent DAMP release during ferroptosis, parthanatos, H2O2-induced necrosis and secondary necrosis. Importantly, the membrane-permeabilizing function of NINJ1 takes place after the metabolic death of the cells and is independent of the pore-forming molecules MLKL, GSDMD and GSDME. During ferroptosis, NINJ1 acts downstream of lipid peroxidation, which suggested a role for reactive oxygen species (ROS) in NINJ1 activation. Reactive oxygen species were however neither sufficient nor required to trigger NINJ1-dependent PMP. Instead, we found that NINJ1 oligomerization is induced by the swelling of the cell and that its permeabilizing potential still requires an addition, and yet to be discovered, activation mechanism.


Subject(s)
Apoptosis , Hydrogen Peroxide , Humans , Reactive Oxygen Species/metabolism , Hydrogen Peroxide/metabolism , Apoptosis/physiology , Necrosis/metabolism , Cell Membrane/metabolism , Nerve Growth Factors/metabolism , Cell Adhesion Molecules, Neuronal/metabolism
2.
Trends Immunol ; 44(8): 628-643, 2023 08.
Article in English | MEDLINE | ID: mdl-37357102

ABSTRACT

Tumor necrosis factor (TNF) plays a central role in orchestrating mammalian inflammatory responses. It promotes inflammation either directly by inducing inflammatory gene expression or indirectly by triggering cell death. TNF-mediated cell death-driven inflammation can be beneficial during infection by providing cell-extrinsic signals that help to mount proper immune responses. Uncontrolled cell death caused by TNF is instead highly detrimental and is believed to cause several human autoimmune diseases. Death is not the default response to TNF sensing. Molecular brakes, or cell death checkpoints, actively repress TNF cytotoxicity to protect the organism from its detrimental consequences. These checkpoints therefore constitute essential safeguards against inflammatory diseases. Recent advances in the field have revealed the existence of several new and unexpected brakes against TNF cytotoxicity and pathogenicity.


Subject(s)
Apoptosis , Signal Transduction , Animals , Humans , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases , Cell Death , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Inflammation/pathology , Mammals
3.
Front Chem ; 11: 1160164, 2023.
Article in English | MEDLINE | ID: mdl-37090247

ABSTRACT

Receptor-Interacting serine/threonine-Protein Kinase 1 (RIPK1) emerged as an important driver of inflammation and, consequently, inflammatory pathologies. The enzymatic activity of RIPK1 is known to indirectly promote inflammation by triggering cell death, in the form of apoptosis, necroptosis and pyroptosis. Small molecule Receptor-Interacting serine/threonine-Protein Kinase 1 inhibitors have therefore recently entered clinical trials for the treatment of a subset of inflammatory pathologies. We previously identified GSK2656157 (GSK'157), a supposedly specific inhibitor of protein kinase R (PKR)-like ER kinase (PERK), as a much more potent type II Receptor-Interacting serine/threonine-Protein Kinase 1 inhibitor. We now performed further structural optimisation on the GSK'157 scaffold in order to develop a novel class of more selective Receptor-Interacting serine/threonine-Protein Kinase 1 inhibitors. Based on a structure-activity relationship (SAR) reported in the literature, we anticipated that introducing a substituent on the para-position of the pyridinyl ring would decrease the interaction with PERK. Herein, we report a series of novel GSK'157 analogues with different para-substituents with increased selectivity for Receptor-Interacting serine/threonine-Protein Kinase 1. The optimisation led to UAMC-3861 as the best compound of this series in terms of activity and selectivity for Receptor-Interacting serine/threonine-Protein Kinase 1 over PERK. The most selective compounds were screened in vitro for their ability to inhibit RIPK1-dependent apoptosis and necroptosis. With this work, we successfully synthesised a novel series of potent and selective type II Receptor-Interacting serine/threonine-Protein Kinase 1 inhibitors based on the GSK'157 scaffold.

4.
Cell Death Differ ; 30(5): 1097-1154, 2023 05.
Article in English | MEDLINE | ID: mdl-37100955

ABSTRACT

Apoptosis is a form of regulated cell death (RCD) that involves proteases of the caspase family. Pharmacological and genetic strategies that experimentally inhibit or delay apoptosis in mammalian systems have elucidated the key contribution of this process not only to (post-)embryonic development and adult tissue homeostasis, but also to the etiology of multiple human disorders. Consistent with this notion, while defects in the molecular machinery for apoptotic cell death impair organismal development and promote oncogenesis, the unwarranted activation of apoptosis promotes cell loss and tissue damage in the context of various neurological, cardiovascular, renal, hepatic, infectious, neoplastic and inflammatory conditions. Here, the Nomenclature Committee on Cell Death (NCCD) gathered to critically summarize an abundant pre-clinical literature mechanistically linking the core apoptotic apparatus to organismal homeostasis in the context of disease.


Subject(s)
Apoptosis , Caspases , Animals , Humans , Apoptosis/genetics , Cell Death , Caspases/genetics , Caspases/metabolism , Carcinogenesis , Mammals/metabolism
5.
Eur Respir J ; 61(4)2023 04.
Article in English | MEDLINE | ID: mdl-36549711

ABSTRACT

BACKGROUND: Receptor-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. METHODS: We assessed RIPK1 expression in single-cell RNA sequencing (RNA-seq) data from human and mouse lungs, and validated RIPK1 levels in lung tissue of COPD patients via immunohistochemistry. Next, we assessed the consequences of genetic and pharmacological inhibition of RIPK1 kinase activity in experimental COPD, using Ripk1 S25D/S25D kinase-deficient mice and the RIPK1 kinase inhibitor GSK'547. RESULTS: RIPK1 expression 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) increased Ripk1 expression 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. CONCLUSIONS: RIPK1 kinase inhibition is protective in experimental models of COPD and may represent a novel promising therapeutic approach.


Subject(s)
Emphysema , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Humans , Mice , Animals , Lung , Cell Death , Inflammation/metabolism , Mice, Inbred C57BL , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
6.
Nat Rev Immunol ; 23(5): 289-303, 2023 05.
Article in English | MEDLINE | ID: mdl-36380021

ABSTRACT

Tumour necrosis factor (TNF) is a central cytokine in inflammatory reactions, and biologics that neutralize TNF are among the most successful drugs for the treatment of chronic inflammatory and autoimmune pathologies. In recent years, it became clear that TNF drives inflammatory responses not only directly by inducing inflammatory gene expression but also indirectly by inducing cell death, instigating inflammatory immune reactions and disease development. Hence, inhibitors of cell death are being considered as a new therapy for TNF-dependent inflammatory diseases.


Subject(s)
Inflammation , Tumor Necrosis Factor-alpha , Humans , Inflammation/drug therapy , Cytokines/metabolism , Cell Death
7.
Science ; 378(6625): 1201-1207, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36520901

ABSTRACT

Cell death induced by tumor necrosis factor (TNF) can be beneficial during infection by helping to mount proper immune responses. However, TNF-induced death can also drive a variety of inflammatory pathologies. Protectives brakes, or cell-death checkpoints, normally repress TNF cytotoxicity to protect the organism from its potential detrimental consequences. Thus, although TNF can kill, this only occurs when one of the checkpoints is inactivated. Here, we describe a checkpoint that prevents apoptosis through the detoxification of the cytotoxic complex IIa that forms upon TNF sensing. We found that autophagy-related 9A (ATG9A) and 200kD FAK family kinase-interacting protein (FIP200) promote the degradation of this complex through a light chain 3 (LC3)-independent lysosomal targeting pathway. This detoxification mechanism was found to counteract TNF receptor 1 (TNFR1)-mediated embryonic lethality and inflammatory skin disease in mouse models.


Subject(s)
Apoptosis , Autophagy-Related Proteins , Membrane Proteins , Tumor Necrosis Factor-alpha , Vesicular Transport Proteins , Animals , Mice , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Dermatitis/genetics , Dermatitis/metabolism , Dermatitis/pathology , Disease Models, Animal , Embryo Loss/genetics , Embryo Loss/metabolism , Embryo Loss/pathology , Lysosomes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
8.
Front Immunol ; 13: 1067164, 2022.
Article in English | MEDLINE | ID: mdl-36532075

ABSTRACT

The Inhibitor of Kappa B Kinase (IKK) complex is a critical regulator of NF-κB activation. More recently, IKK has also been shown to repress RIPK1 dependent extrinsic cell death pathways by directly phosphorylating RIPK1 at serine 25. In T cells, IKK expression is essential for normal development in the thymus, by promoting survival of thymocytes independently of NF-κB activation. RIPK1 undergoes extensive phosphorylation following TNF stimulation in T cells, though which targets are required to repress RIPK1 has not been defined. Here, we show that TNF induced phosphorylation of RIPK1 at S25 is IKK dependent. We test the relevance of this phosphorylation event in T cells using mice with a RIPK1S25D phosphomimetic point mutation to endogenous RIPK1. We find that this mutation protects T cells from TNF induced cell death when IKK activity is inhibited in vitro, and can rescues development of IKK deficient thymocytes in vivo to a degree comparable with kinase dead RIPK1D138N. Together, these data show that phosphorylation of RIPK1S25 by IKK represents a key regulatory event promoting survival of T cells by IKK.


Subject(s)
NF-kappa B , Serine , Mice , Animals , Phosphorylation , NF-kappa B/metabolism , Serine/metabolism , Apoptosis , Tumor Necrosis Factor-alpha/metabolism , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Cell Death , Thymocytes/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
9.
EMBO Rep ; 23(12): e55233, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36194667

ABSTRACT

The anti-inflammatory protein A20 serves as a critical brake on NF-κB signaling and NF-κB-dependent inflammation. In humans, polymorphisms in or near the TNFAIP3/A20 gene have been associated with several inflammatory disorders, including rheumatoid arthritis (RA), and experimental studies in mice have demonstrated that myeloid-specific A20 deficiency causes the development of a severe polyarthritis resembling human RA. Myeloid A20 deficiency also promotes osteoclastogenesis in mice, suggesting a role for A20 in the regulation of osteoclast differentiation and bone formation. We show here that osteoclast-specific A20 knockout mice develop severe osteoporosis, but not inflammatory arthritis. In vitro, osteoclast precursor cells from A20 deficient mice are hyper-responsive to RANKL-induced osteoclastogenesis. Mechanistically, we show that A20 is recruited to the RANK receptor complex within minutes of ligand binding, where it restrains NF-κB activation independently of its deubiquitinating activity but through its zinc finger (ZnF) 4 and 7 ubiquitin-binding functions. Together, these data demonstrate that A20 acts as a regulator of RANK-induced NF-κB signaling to control osteoclast differentiation, assuring proper bone development and turnover.


Subject(s)
NF-kappa B , Humans , Animals , Mice
10.
Cell Rep ; 40(7): 111205, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35977488

ABSTRACT

Despite its crucial role in initiation of cytotoxic immune responses, the molecular pathways underlying antigen cross-presentation remain incompletely understood. The mechanism of antigen exit from endocytic compartments into the cytosol is a long-standing matter of controversy, confronting two main models: transfer through specific channels/transporters or rupture of endocytic membranes and leakage of luminal content. By monitoring the occurrence of intracellular damage in conventional dendritic cells (cDCs), we show that cross-presenting cDC1s display more frequent endomembrane injuries and increased recruitment of endosomal sorting complex required for transport (ESCRT)-III, the main repair system for intracellular membranes, relative to cDC2s. Silencing of CHMP2a or CHMP4b, two effector subunits of ESCRT-III, enhances cytosolic antigen export and cross-presentation. This phenotype is partially reversed by chemical inhibition of RIPK3, suggesting that endocytic damage is related to basal activation of the necroptosis pathway. Membrane repair therefore proves crucial in containing antigen export to the cytosol and cross-presentation in cDCs.


Subject(s)
Cross-Priming , Endosomal Sorting Complexes Required for Transport , Antigen Presentation , Antigens/metabolism , Cytosol/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism
11.
Biomedicines ; 10(5)2022 Apr 28.
Article in English | MEDLINE | ID: mdl-35625752

ABSTRACT

RIPK1 (receptor-interacting serine/threonine-protein kinase 1) enzymatic activity drives both apoptosis and necroptosis, a regulated form of necrosis. Because necroptosis is involved in necrotic core development in atherosclerotic plaques, we investigated the effects of a RIPK1S25D/S25D mutation, which prevents activation of RIPK1 kinase, on atherogenesis in ApoE-/- mice. After 16 weeks of western-type diet (WD), atherosclerotic plaques from ApoE-/- RIPK1S25D/S25D mice were significantly larger compared to ApoE-/- RIPK1+/+ mice (167 ± 34 vs. 78 ± 18 × 103 µm2, p = 0.01). Cell numbers (350 ± 34 vs. 154 ± 33 nuclei) and deposition of glycosaminoglycans (Alcian blue: 31 ± 6 vs. 14 ± 4%, p = 0.023) were increased in plaques from ApoE-/- RIPK1S25D/S25D mice while macrophage content (Mac3: 2.3 ± 0.4 vs. 9.8 ± 2.4%, p = 0.012) was decreased. Plaque apoptosis was not different between both groups. In contrast, pharmacological inhibition of RIPK1 kinase with GSK'547 (10 mg/kg BW/day) in ApoE-/- Fbn1C1039G+/- mice, a model of advanced atherosclerosis, did not alter plaque size after 20 weeks WD, but induced apoptosis (TUNEL: 136 ± 20 vs. 62 ± 9 cells/mm2, p = 0.004). In conclusion, inhibition of RIPK1 kinase activity accelerated plaque progression in ApoE-/- RIPK1S25D/S25D mice and induced apoptosis in GSK'547-treated ApoE-/- Fbn1C1039G+/- mice. Thus, without directly comparing the genetic and pharmacological studies, it can be concluded that targeting RIPK1 kinase activity does not limit atherogenesis.

12.
Cell Death Dis ; 12(7): 699, 2021 07 14.
Article in English | MEDLINE | ID: mdl-34262020

ABSTRACT

Butylate hydroxyanisole (BHA) is a synthetic phenol that is widely utilized as a preservative by the food and cosmetic industries. The antioxidant properties of BHA are also frequently used by scientists to claim the implication of reactive oxygen species (ROS) in various cellular processes, including cell death. We report on the surprising finding that BHA functions as a direct inhibitor of RIPK1, a major signaling hub downstream of several immune receptors. Our in silico analysis predicts binding of 3-BHA, but not 2-BHA, to RIPK1 in an inactive DLG-out/Glu-out conformation, similar to the binding of the type III inhibitor Nec-1s to RIPK1. This predicted superior inhibitory capacity of 3-BHA over 2-BHA was confirmed in cells and using in vitro kinase assays. We demonstrate that the reported protective effect of BHA against tumor necrosis factor (TNF)-induced necroptotic death does not originate from ROS scavenging but instead from direct RIPK1 enzymatic inhibition, a finding that most probably extends to other reported effects of BHA. Accordingly, we show that BHA not only protects cells against RIPK1-mediated necroptosis but also against RIPK1 kinase-dependent apoptosis. We found that BHA treatment completely inhibits basal and induced RIPK1 enzymatic activity in cells, monitored at the level of TNFR1 complex I under apoptotic conditions or in the cytosol under necroptosis. Finally, we show that oral administration of BHA protects mice from RIPK1 kinase-dependent lethality caused by TNF injection, a model of systemic inflammatory response syndrome. In conclusion, our results demonstrate that BHA can no longer be used as a strict antioxidant and that new functions of RIPK1 may emerge from previously reported effects of BHA.


Subject(s)
Apoptosis/drug effects , Butylated Hydroxyanisole/pharmacology , Fibroblasts/drug effects , Food Additives/pharmacology , Necroptosis/drug effects , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Systemic Inflammatory Response Syndrome/prevention & control , Animals , Antioxidants/pharmacology , Disease Models, Animal , Female , Fibroblasts/enzymology , Fibroblasts/pathology , HT29 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Docking Simulation , Protein Binding , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Systemic Inflammatory Response Syndrome/chemically induced , Systemic Inflammatory Response Syndrome/enzymology , Systemic Inflammatory Response Syndrome/pathology , Tumor Necrosis Factor-alpha
13.
Nat Commun ; 11(1): 1747, 2020 04 08.
Article in English | MEDLINE | ID: mdl-32269263

ABSTRACT

Receptor interacting protein kinase 1 (RIPK1) regulates cell death and inflammatory responses downstream of TNFR1 and other receptors, and has been implicated in the pathogenesis of inflammatory and degenerative diseases. RIPK1 kinase activity induces apoptosis and necroptosis, however the mechanisms and phosphorylation events regulating RIPK1-dependent cell death signaling remain poorly understood. Here we show that RIPK1 autophosphorylation at serine 166 plays a critical role for the activation of RIPK1 kinase-dependent apoptosis and necroptosis. Moreover, we show that S166 phosphorylation is required for RIPK1 kinase-dependent pathogenesis of inflammatory pathologies in vivo in four relevant mouse models. Mechanistically, we provide evidence that trans autophosphorylation at S166 modulates RIPK1 kinase activation but is not by itself sufficient to induce cell death. These results show that S166 autophosphorylation licenses RIPK1 kinase activity to induce downstream cell death signaling and inflammation, suggesting that S166 phosphorylation can serve as a reliable biomarker for RIPK1 kinase-dependent pathologies.


Subject(s)
Apoptosis , Inflammation/metabolism , Inflammation/pathology , Phosphoserine/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Alanine Transaminase/metabolism , Animals , Bone Marrow Cells/cytology , Colitis/pathology , Genotype , Hepatitis/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mutation/genetics , Neoplasms/pathology , Phosphorylation , Sepsis/pathology , Skin/pathology , Tumor Necrosis Factor-alpha
14.
Trends Immunol ; 41(5): 421-435, 2020 05.
Article in English | MEDLINE | ID: mdl-32241683

ABSTRACT

A20 is a potent anti-inflammatory molecule, and mutations in TNFAIP3, the gene encoding A20, are associated with a wide panel of inflammatory pathologies, both in human and mouse. The anti-inflammatory properties of A20 are commonly attributed to its ability to suppress inflammatory NF-κB signaling by functioning as a ubiquitin-editing enzyme. However, A20 also protects cells from death, independently of NF-κB regulation, and recent work has demonstrated that cell death may drive some of the inflammatory conditions caused by A20 deficiency. Adding to the fact that the protective role of A20 does not primarily rely on its catalytic activities, these findings shed new light on A20 biology.


Subject(s)
Inflammation , NF-kappa B , Humans , Animals , Mice , NF-kappa B/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Inflammation/metabolism , Signal Transduction , Cell Death , Anti-Inflammatory Agents
15.
Nat Immunol ; 21(4): 381-387, 2020 04.
Article in English | MEDLINE | ID: mdl-32205881

ABSTRACT

Protein ubiquitination regulates protein stability and modulates the composition of signaling complexes. A20 is a negative regulator of inflammatory signaling, but the molecular mechanisms involved are ill understood. Here, we generated Tnfaip3 gene-targeted A20 mutant mice bearing inactivating mutations in the zinc finger 7 (ZnF7) and ZnF4 ubiquitin-binding domains, revealing that binding to polyubiquitin is essential for A20 to suppress inflammatory disease. We demonstrate that a functional ZnF7 domain was required for recruiting A20 to the tumor necrosis factor receptor 1 (TNFR1) signaling complex and to suppress inflammatory signaling and cell death. The combined inactivation of ZnF4 and ZnF7 phenocopied the postnatal lethality and severe multiorgan inflammation of A20-deficient mice. Conditional tissue-specific expression of mutant A20 further revealed the key role of ubiquitin-binding in myeloid and intestinal epithelial cells. Collectively, these results demonstrate that the anti-inflammatory and cytoprotective functions of A20 are largely dependent on its ubiquitin-binding properties.


Subject(s)
Inflammation/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Animals , Epithelial Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Polyubiquitin/metabolism , Protein Binding/physiology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin/metabolism , Ubiquitination/physiology , Zinc Fingers/physiology
16.
Cell Rep ; 30(7): 2237-2247.e6, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32075762

ABSTRACT

Inflammatory signaling pathways are tightly regulated to avoid chronic inflammation and the development of disease. OTULIN is a deubiquitinating enzyme that controls inflammation by cleaving linear ubiquitin chains generated by the linear ubiquitin chain assembly complex. Here, we show that ablation of OTULIN in liver parenchymal cells in mice causes severe liver disease which is characterized by liver inflammation, hepatocyte apoptosis, and compensatory hepatocyte proliferation, leading to steatohepatitis, fibrosis, and hepatocellular carcinoma (HCC). Genetic ablation of Fas-associated death domain (FADD) completely rescues and knockin expression of kinase inactive receptor-interacting protein kinase 1 (RIPK1) significantly protects mice from developing liver disease, demonstrating that apoptosis of OTULIN-deficient hepatocytes triggers disease pathogenesis in this model. Finally, we demonstrate that type I interferons contribute to disease in hepatocyte-specific OTULIN-deficient mice. Our study reveals the critical importance of OTULIN in protecting hepatocytes from death, thereby preventing the development of chronic liver inflammation and HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Endopeptidases/metabolism , Fas-Associated Death Domain Protein/antagonists & inhibitors , Hepatitis/metabolism , Liver Neoplasms/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/physiology , CHO Cells , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cricetulus , Fas-Associated Death Domain Protein/metabolism , Hepatitis/genetics , Hepatitis/pathology , Humans , Interferon Type I/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction
17.
Am J Respir Crit Care Med ; 201(11): 1358-1371, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32105156

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 bronchiolitis 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 HMGB1 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.


Subject(s)
Bronchiolitis/virology , Epithelial Cells/metabolism , Epithelial Cells/pathology , HMGB1 Protein/metabolism , Necroptosis , Respiratory Mucosa/cytology , Respiratory Syncytial Virus Infections/metabolism , Animals , Child, Preschool , Humans , Infant , Mice , Prospective Studies
18.
Trends Cell Biol ; 30(3): 189-200, 2020 03.
Article in English | MEDLINE | ID: mdl-31959328

ABSTRACT

The serine/threonine kinase RIPK1 has emerged as a crucial component of the inflammatory response activated downstream of several immune receptors, where it paradoxically functions as a scaffold to protect the cell from death or instead as an active kinase to promote the killing of the cell. While RIPK1 kinase-dependent cell death has revealed its physiological importance in the context of microbial infection, aberrant activation of RIPK1 is also demonstrated to promote cell death-driven inflammatory pathologies, highlighting the importance of fundamentally understanding proper RIPK1 regulation. Recent advances in the field demonstrated the crucial role of phosphorylation in the fine-tuning of RIPK1 activation and, additionally, question the exact mechanism by which RIPK1 enzymatic activity transmits the death signal.


Subject(s)
Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Cell Death , Humans , Models, Biological , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Ubiquitination
19.
Cell Death Dis ; 11(1): 60, 2020 Jan 23.
Article in English | MEDLINE | ID: mdl-31974345

ABSTRACT

The original version of this article contained an error in the name of one of the co-authors (Wim Declercq). This has been corrected in the PDF and HTML versions.

20.
J Immunol ; 204(4): 775-787, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31900335

ABSTRACT

Immunogenic cell death (ICD) occurs when a dying cell releases cytokines and damage-associated molecular patterns, acting as adjuvants, and expresses Ags that induce a specific antitumor immune response. ICD is studied mainly in the context of regulated cell death pathways, especially caspase-mediated apoptosis marked by endoplasmic reticulum stress and calreticulin exposure and, more recently, also in relation to receptor-interacting protein kinase-driven necroptosis, whereas unregulated cell death like accidental necrosis is nonimmunogenic. Importantly, the murine cancer cell lines used in ICD studies often express virally derived peptides that are recognized by the immune system as tumor-associated Ags. However, it is unknown how different cell death pathways may affect neoepitope cross-presentation and Ag recognition of cancer cells. We used a prophylactic tumor vaccination model and observed that both apoptotic and necroptotic colon carcinoma CT26 cells efficiently immunized mice against challenge with a breast cancer cell line that expresses the same immunodominant tumor Ag, AH1, but only necroptotic CT26 cells would mount an immune response against CT26-specific neoepitopes. By CRISPR/Cas9 genome editing, we knocked out AH1 and saw that only necroptotic CT26 cells were still able to protect mice against tumor challenge. Hence, in this study, we show that endogenous AH1 tumor Ag expression can mask the strength of immunogenicity induced by different cell death pathways and that upon knockout of AH1, necroptosis was more immunogenic than apoptosis in a prophylactic tumor vaccination model. This work highlights necroptosis as a possible preferred ICD form over apoptosis in the treatment of cancer.


Subject(s)
Antigens, Neoplasm/immunology , Apoptosis/immunology , Immunodominant Epitopes/immunology , Necroptosis/immunology , Neoplasms, Experimental/immunology , Animals , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...