Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
Add more filters










Publication year range
1.
Eur J Clin Invest ; 52(3): e13648, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34219227

ABSTRACT

BACKGROUND: Receptor-interacting protein kinase 3 (RIPK3) is a key player of regulated necrosis or necroptosis, an inflammatory form of cell death possibly governing outcomes in chronic liver diseases, such as nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. METHODS: This narrative review is based on literature search using PubMed. RESULTS: RIPK3 activation depends on post-transcriptional modifications, including phosphorylation, hence coordinating the assembly of macromolecular death complex named 'necrosome', which may also involve diverse mitochondrial components. Curiously, recent studies suggested a potential link between RIPK3 and mitochondrial bioenergetics. RIPK3 can modulate mitochondrial function and quality through the regulation of mitochondrial reactive oxygen species production, sequestration of metabolic enzymes and resident mitochondrial proteins, activity of mitochondrial respiratory chain complexes, mitochondrial biogenesis and fatty acid oxidation. CONCLUSIONS: Since mitochondrial dysfunction and RIPK3-mediated necroptosis are intimately involved in chronic liver disease pathogenesis, understanding the role of RIPK3 in mitochondrial bioenergetics and its potential translational application are of great interest.


Subject(s)
Energy Metabolism , Mitochondria, Liver/physiology , Non-alcoholic Fatty Liver Disease/etiology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Humans , Mitochondria, Liver/metabolism
2.
Sci Rep ; 11(1): 18679, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34548542

ABSTRACT

Programmed necrosis, such as necroptosis and pyroptosis, is a highly pro-inflammatory cellular event that is associated with chronic inflammation. Although there are various triggers of pyroptosis and necroptosis in autoimmune tissue inflammation and subsequent lytic forms of cell death release abundant inflammatory mediators, including damage-associated molecular patterns and IL-1ß, capable of amplifying autoimmune Th17 effector functions, it remains largely unclear whether the programs play a crucial role in the pathogenesis of autoimmune arthritis. We herein report that Gasdermin D (Gsdmd) and receptor interacting serine/threonine kinase 3 (Ripk3)-key molecules of pyroptosis and necroptosis, respectively-are upregulated in inflamed synovial tissues, but dispensable for IL-1ß production and the development of IL-17-producing T helper (Th17) cell-mediated autoimmune arthritis in SKG mice. Gsdmd-/-, Ripk3-/-, or Gsdmd-/- Ripk3-/- SKG mice showed severe arthritis with expansion of arthritogenic Th17 cells in the draining LNs and inflamed joints, which was comparable to that in wild-type SKG mice. Despite the marked reduction of IL-1ß secretion from Gsdmd-/- or Ripk3-/- bone marrow-derived DCs by canonical stimuli, IL-1ß levels in the inflamed synovium were not affected in the absence of Gsdmd or Ripk3. Our results revealed that T cell-mediated autoimmune arthritis proceeds independently of the pyroptosis and necroptosis pathways.


Subject(s)
Arthritis/immunology , Autoimmune Diseases/immunology , Inflammation/immunology , Intracellular Signaling Peptides and Proteins/physiology , Phosphate-Binding Proteins/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Th17 Cells/immunology , Animals , Chronic Disease , Mice
3.
Cells ; 10(9)2021 09 12.
Article in English | MEDLINE | ID: mdl-34572045

ABSTRACT

Receptor interacting protein kinase 3 (RIPK3)-mediated smooth muscle cell (SMC) necroptosis has been shown to contribute to the pathogenesis of abdominal aortic aneurysms (AAAs). However, the signaling steps downstream from RIPK3 during SMC necroptosis remain unknown. In this study, the roles of mixed lineage kinase domain-like pseudokinase (MLKL) and calcium/calmodulin-dependent protein kinase II (CaMKII) in SMC necroptosis were investigated. We found that both MLKL and CaMKII were phosphorylated in SMCs in a murine CaCl2-driven model of AAA and that Ripk3 deficiency reduced the phosphorylation of MLKL and CaMKII. In vitro, mouse aortic SMCs were treated with tumor necrosis factor α (TNFα) plus Z-VAD-FMK (zVAD) to induce necroptosis. Our data showed that both MLKL and CaMKII were phosphorylated after TNFα plus zVAD treatment in a time-dependent manner. SiRNA silencing of Mlkl-diminished cell death and administration of the CaMKII inhibitor myristoylated autocamtide-2-related inhibitory peptide (Myr-AIP) or siRNAs against Camk2d partially inhibited necroptosis. Moreover, knocking down Mlkl decreased CaMKII phosphorylation, but silencing Camk2d did not affect phosphorylation, oligomerization, or trafficking of MLKL. Together, our results indicate that both MLKL and CaMKII are involved in RIPK3-mediated SMC necroptosis, and that MLKL is likely upstream of CaMKII in this process.


Subject(s)
Aortic Aneurysm, Abdominal/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Myocytes, Smooth Muscle/pathology , Necrosis , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/metabolism , Calcium Chloride/toxicity , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Phosphorylation , Protein Kinases/chemistry , Protein Kinases/genetics , RNA, Small Interfering/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
4.
PLoS Biol ; 19(8): e3001304, 2021 08.
Article in English | MEDLINE | ID: mdl-34437534

ABSTRACT

Tumor necrosis factor receptor-1 (TNFR1) signaling, apart from its pleiotropic functions in inflammation, plays a role in embryogenesis as deficiency of varieties of its downstream molecules leads to embryonic lethality in mice. Caspase-8 noncleavable receptor interacting serine/threonine kinase 1 (RIPK1) mutations occur naturally in humans, and the corresponding D325A mutation in murine RIPK1 leads to death at early midgestation. It is known that both the demise of Ripk1D325A/D325A embryos and the death of Casp8-/- mice are initiated by TNFR1, but they are mediated by apoptosis and necroptosis, respectively. Here, we show that the defects in Ripk1D325A/D325A embryos occur at embryonic day 10.5 (E10.5), earlier than that caused by Casp8 knockout. By analyzing a series of genetically mutated mice, we elucidated a mechanism that leads to the lethality of Ripk1D325A/D325A embryos and compared it with that underlies Casp8 deletion-mediated lethality. We revealed that the apoptosis in Ripk1D325A/D325A embryos requires a scaffold function of RIPK3 and enzymatically active caspase-8. Unexpectedly, caspase-1 and caspase-11 are downstream of activated caspase-8, and concurrent depletion of Casp1 and Casp11 postpones the E10.5 lethality to embryonic day 13.5 (E13.5). Moreover, caspase-3 is an executioner of apoptosis at E10.5 in Ripk1D325A/D325A mice as its deletion extends life of Ripk1D325A/D325A mice to embryonic day 11.5 (E11.5). Hence, an unexpected death pathway of TNFR1 controls RIPK1 D325A mutation-induced lethality at E10.5.


Subject(s)
Caspase 8/physiology , Embryonic Development , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Animals , Caspases/metabolism , Cell Death , Mice , Primary Cell Culture , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
5.
Cell Death Dis ; 12(6): 593, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34103472

ABSTRACT

Autophagy is an important renal-protective mechanism in septic acute kidney injury (AKI). Receptor interacting protein kinase 3 (RIP3) has been implicated in the renal tubular injury and renal dysfunction during septic AKI. Here we investigated the role and mechanism of RIP3 on autophagy in septic AKI. We showed an activation of RIP3, accompanied by an accumulation of the autophagosome marker LC3II and the autophagic substrate p62, in the kidneys of lipopolysaccharide (LPS)-induced septic AKI mice and LPS-treated cultured renal proximal tubular epithelial cells (PTECs). The lysosome inhibitor did not further increase the levels of LCII or p62 in LPS-treated PTECs. Moreover, inhibition of RIP3 attenuated the aberrant accumulation of LC3II and p62 under LPS treatment in vivo and in vitro. By utilizing mCherry-GFP-LC3 autophagy reporter mice in vivo and PTECs overexpression mRFP-GFP-LC3 in vitro, we observed that inhibition of RIP3 restored the formation of autolysosomes and eliminated the accumulated autophagosomes under LPS treatment. These results indicated that RIP3 impaired autophagic degradation, contributing to the accumulation of autophagosomes. Mechanistically, the nuclear translocation of transcription factor EB (TFEB), a master regulator of the lysosome and autophagy pathway, was inhibited in LPS-induced mice and LPS-treated PTECs. Inhibition of RIP3 restored the nuclear translocation of TFEB in vivo and in vitro. Co-immunoprecipitation further showed an interaction of RIP3 and TFEB in LPS-treated PTECs. Also, the expression of LAMP1 and cathepsin B, two potential target genes of TFEB involved in lysosome function, were decreased under LPS treatment in vivo and in vitro, and this decrease was rescued by inhibiting RIP3. Finally, overexpression of TFEB restored the autophagic degradation in LPS-treated PTECs. Together, the present study has identified a pivotal role of RIP3 in suppressing autophagic degradation through impeding the TFEB-lysosome pathway in septic AKI, providing potential therapeutic targets for the prevention and treatment of septic AKI.


Subject(s)
Acute Kidney Injury/etiology , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Sepsis/complications , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Autophagosomes/genetics , Autophagosomes/metabolism , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Sepsis/genetics , Sepsis/metabolism , Sepsis/pathology , Signal Transduction/genetics
6.
Cell Death Dis ; 12(6): 601, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112763

ABSTRACT

The sensing of various extrinsic stimuli triggers the receptor-interacting protein kinase-3 (RIPK3)-mediated signaling pathway, which leads to mixed-lineage kinase-like (MLKL) phosphorylation followed by necroptosis. Although necroptosis is a form of cell death and is involved in inflammatory conditions, the roles of necroptosis in acute pancreatitis (AP) remain unclear. In the current study, we administered caerulein to Ripk3- or Mlkl-deficient mice (Ripk3-/- or Mlkl-/- mice, respectively) and assessed the roles of necroptosis in AP. We found that Ripk3-/- mice had significantly more severe pancreatic edema and inflammation associated with macrophage and neutrophil infiltration than control mice. Consistently, Mlkl-/- mice were more susceptible to caerulein-induced AP, which occurred in a time- and dose-dependent manner, than control mice. Mlkl-/- mice exhibit weight loss, edematous pancreatitis, necrotizing pancreatitis, and acinar cell dedifferentiation in response to tissue damage. Genetic deletion of Mlkl resulted in downregulation of the antiapoptotic genes Bclxl and Cflar in association with increases in the numbers of apoptotic cells, as detected by TUNEL assay. These findings suggest that RIPK3 and MLKL-mediated necroptosis exerts protective effects in AP and caution against the use of necroptosis inhibitors for AP treatment.


Subject(s)
Necroptosis/physiology , Pancreatitis/pathology , Protein Kinases/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Cell Dedifferentiation/genetics , Cytoprotection/genetics , Disease Progression , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Necroptosis/genetics , Neutrophil Activation/genetics , Pancreas/metabolism , Pancreas/pathology , Pancreatitis/genetics , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
7.
J Invest Dermatol ; 141(12): 2921-2931.e6, 2021 12.
Article in English | MEDLINE | ID: mdl-34102211

ABSTRACT

Vitiligo is a common depigmentation disease characterized by melanocyte death, which is attributed to various mechanisms such as apoptosis and autoimmune destruction. However, whether necroptosis, a newly discovered way of cell death, plays a key role in the pathogenesis of vitiligo is still elusive and has not been well-studied. In this study, we found that necroptosis markers, including phosphorylated RIP3 and phosphorylated-MLKL, were positive in melanocytes from vitiligo perilesional skin, which supported the existence of necroptosis in vitiligo. Furthermore, the expression of RIP1 was remarkably upregulated in melanocytes treated with hydrogen peroxide. Then, RIP1 intervention suppression and MLKL deficiency could significantly enhance the resistance of melanocytes to hydrogen peroxide‒induced necroptosis. Mechanistically, we confirmed that RIP1 and RIP3 could form necrosomes under oxidative stress and further trigger phosphorylated MLKL translocation to the cell membrane, which led to the destruction of melanocytes. Finally, we showed that RIP1-mediated generation of mitochondrial ROS contributed to necrosome formation in melanocytes. Collectively, our study confirms that necroptosis significantly facilitates oxidative stress‒induced melanocyte death through the RIP1 signaling pathway, offering insight into vitiligo.


Subject(s)
Melanocytes/pathology , Necroptosis/physiology , Nuclear Pore Complex Proteins/physiology , Oxidative Stress/physiology , RNA-Binding Proteins/physiology , Vitiligo/etiology , Humans , MAP Kinase Signaling System/physiology , Protein Kinases/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Vitiligo/pathology
8.
Biomolecules ; 11(5)2021 04 28.
Article in English | MEDLINE | ID: mdl-33924766

ABSTRACT

Chronic inflammatory disorders are characterised by aberrant and exaggerated inflammatory immune cell responses. Modes of extrinsic cell death, apoptosis and necroptosis, have now been shown to be potent drivers of deleterious inflammation, and mutations in core repressors of these pathways underlie many autoinflammatory disorders. The receptor-interacting protein (RIP) kinases, RIPK1 and RIPK3, are integral players in extrinsic cell death signalling by regulating the production of pro-inflammatory cytokines, such as tumour necrosis factor (TNF), and coordinating the activation of the NOD-like receptor protein 3 (NLRP3) inflammasome, which underpin pathological inflammation in numerous chronic inflammatory disorders. In this review, we firstly give an overview of the inflammatory cell death pathways regulated by RIPK1 and RIPK3. We then discuss how dysregulated signalling along these pathways can contribute to chronic inflammatory disorders of the joints, skin, and gastrointestinal tract, and discuss the emerging evidence for targeting these RIP kinases in the clinic.


Subject(s)
Inflammation/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis/physiology , Cell Death/immunology , Chronic Disease , Cytokines/metabolism , Humans , Inflammasomes/metabolism , Inflammation/physiopathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Necrosis/metabolism , Phosphorylation , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
9.
FASEB J ; 35(5): e21561, 2021 05.
Article in English | MEDLINE | ID: mdl-33864423

ABSTRACT

Methamphetamine (METH) is a synthetic drug with severe neurotoxicity, however, the regulation of METH-induced neuronal programmed necrosis remains poorly understood. The aim of this study was to identify the molecular mechanisms of METH-induced neuronal programmed necrosis. We found that neuronal programmed necrosis occurred in the striatum of brain samples from human and mice that were exposed to METH. The receptor-interacting protein kinase 3 (RIP3) was highly expressed in the neurons of human and mice exposed to METH, and RIP3-silenced or RIP1-inhibited protected neurons developed neuronal programmed necrosis in vitro and in vivo following METH exposure. Moreover, the RIP1-RIP3 complex causes cell programmed necrosis by regulating mixed lineage kinase domain-like protein (MLKL)-mediated cell membrane rupture and dynamin-related protein 1 (Drp1)-mediated mitochondrial fission. Together, these data indicate that RIP3 plays an indispensable role in the mechanism of METH-induced neuronal programmed necrosis, which may represent a potential therapeutic target for METH-induced neurotoxicity.


Subject(s)
Gene Expression Regulation/drug effects , Methamphetamine/toxicity , Necrosis , Neurons/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Central Nervous System Stimulants/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Phosphorylation , Rats , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction
10.
Sci Signal ; 14(668)2021 02 02.
Article in English | MEDLINE | ID: mdl-33531383

ABSTRACT

Necroptosis is a lytic, proinflammatory cell death pathway, which has been implicated in host defense and, when dysregulated, the pathology of many human diseases. The central mediators of this pathway are the receptor-interacting serine/threonine protein kinases RIPK1 and RIPK3 and the terminal executioner, the pseudokinase mixed lineage kinase domain-like (MLKL). Here, we review the chronology of signaling along the RIPK1-RIPK3-MLKL axis and highlight how the subcellular compartmentalization of signaling events controls the initiation and execution of necroptosis. We propose that a network of modulators surrounds the necroptotic signaling core and that this network, rather than acting universally, tunes necroptosis in a context-, cell type-, and species-dependent manner. Such a high degree of mechanistic flexibility is likely an important property that helps necroptosis operate as a robust, emergency form of cell death.


Subject(s)
Necroptosis , Protein Kinases/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Humans , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
11.
Int J Cancer ; 148(5): 1276-1288, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33038274

ABSTRACT

The liver ischemia-reperfusion (IR) injury that occurs consequently to hepatic resection performed in patients with metastases can lead to tumor relapse for not fully understood reasons. We assessed the effects of liver IR on tumor growth and the innate immune response in a mouse model of colorectal (CR) liver metastasis. Mice subjected to liver ischemia 2 days after intrasplenic injection of CR carcinoma cells displayed a higher metastatic load in the liver, correlating with Kupffer cells (KC) death through the activation of receptor-interating protein 3 kinase (RIPK3) and caspase-1 and a recruitment of monocytes. Interestingly, the immunoregulatory mediators, tumor necrosis factor-α (TNF-α) and heme oxygenase-1 (HO-1) were strongly upregulated in recruited monocytes and were also expressed in the surviving KC following IR. Using TNFflox/flox LysMcre/wt mice, we showed that TNF deficiency in macrophages and monocytes favors tumor progression after IR. The antitumor effect of myeloid cell-derived TNF involved direct tumor cell apoptosis and a reduced expression of immunosuppressive molecules such as transforming growth factor-ß, interleukin (IL)-10, inducible nitric oxyde synthase (iNOS), IL-33 and HO-1. Conversely, a monocyte/macrophage-specific deficiency in HO-1 (HO-1flox/flox LysMcre/wt ) or the blockade of HO-1 function led to the control of tumor progression post-liver IR. Importantly, host cell RIPK3 deficiency maintains the KC number upon IR, inhibits the IR-induced innate cell recruitment, increases the TNF level, decreases the HO-1 level and suppresses the tumor outgrowth. In conclusion, tumor recurrence in host undergoing liver IR is associated with the death of antitumoral KC and the recruitment of monocytes endowed with immunosuppressive properties. In both of which HO-1 inhibition would reinforce their antitumoral activity.


Subject(s)
Colorectal Neoplasms/pathology , Heme Oxygenase-1/physiology , Liver Neoplasms/etiology , Liver Neoplasms/secondary , Liver/blood supply , Neoplasm Recurrence, Local/etiology , Reperfusion Injury/complications , Tumor Necrosis Factor-alpha/physiology , Animals , Disease Progression , Kupffer Cells/physiology , Male , Mice , Mice, Inbred C57BL , Monocytes/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology
12.
J Cereb Blood Flow Metab ; 41(7): 1623-1633, 2021 07.
Article in English | MEDLINE | ID: mdl-33210566

ABSTRACT

Receptor-interacting protein kinase-1 (RIPK1) is a master regulator of cell death and inflammation, and mediates programmed necrosis (necroptosis) via mixed-lineage kinase like (MLKL) protein. Prior studies in experimental intracerebral hemorrhage (ICH) implicated RIPK1 in the pathogenesis of neuronal death and cognitive outcome, but the relevant cell types involved and potential role of necroptosis remain unexplored. In mice subjected to autologous blood ICH, early RIPK1 activation was observed in neurons, endothelium and pericytes, but not in astrocytes. MLKL activation was detected in astrocytes and neurons but not endothelium or pericytes. Compared with WT controls, RIPK1 kinase-dead (RIPK1D138N/D138N) mice had reduced brain edema (24 h) and blood-brain barrier (BBB) permeability (24 h, 30 d), and improved postinjury rotarod performance. Mice deficient in MLKL (Mlkl-/-) had reduced neuronal death (24 h) and BBB permeability at 24 h but not 30d, and improved post-injury rotarod performance vs. WT. The data support a central role for RIPK1 in the pathogenesis of ICH, including cell death, edema, BBB permeability, and motor deficits. These effects may be mediated in part through the activation of MLKL-dependent necroptosis in neurons. The data support development of RIPK1 kinase inhibitors as therapeutic agents for human ICH.


Subject(s)
Blood-Brain Barrier/physiology , Cerebral Hemorrhage/complications , Edema/prevention & control , Inflammation/prevention & control , Necrosis , Protein Kinases/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Behavior, Animal , Cell Membrane Permeability , Edema/etiology , Edema/metabolism , Edema/pathology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , Neurons
13.
J Am Soc Nephrol ; 31(9): 2097-2115, 2020 09.
Article in English | MEDLINE | ID: mdl-32641397

ABSTRACT

BACKGROUND: Gentamicin is a potent aminoglycoside antibiotic that targets gram-negative bacteria, but nephrotoxicity limits its clinical application. The cause of gentamicin-induced AKI has been attributed mainly to apoptosis of the proximal tubule cells. However, blocking apoptosis only partially attenuates gentamicin-induced AKI in animals. METHODS: Mice treated with gentamicin for 7 days developed AKI, and programmed cell death pathways were examined using pharmacologic inhibitors and in RIPK3-deficient mice. Effects in porcine and murine kidney cell lines were also examined. RESULTS: Gentamicin caused a low level of apoptosis in the proximal tubules and significant ultrastructural alterations consistent with necroptosis, occurring predominantly in the collecting ducts (CDs), including cell and organelle swelling and rupture of the cell membrane. Upregulation of the key necroptotic signaling molecules, mixed lineage kinase domain-like pseudokinase (MLKL) and receptor-interacting serine/threonine-protein kinase 3 (RIPK3), was detected in gentamicin-treated mice and in cultured renal tubule cells. In addition, gentamicin induced apical accumulation of total and phosphorylated MLKL (pMLKL) in CDs in mouse kidney. Inhibiting a necroptotic protein, RIPK1, with necrostatin-1 (Nec-1), attenuated gentamicin-induced necrosis and upregulation of MLKL and RIPK3 in mice and cultured cells. Nec-1 also alleviated kidney inflammation and fibrosis, and significantly improved gentamicin-induced renal dysfunction in mice. Furthermore, deletion of RIPK3 in the Ripk3-/- mice significantly attenuated gentamicin-induced AKI. CONCLUSIONS: A previously unrecognized role of programmed necrosis in collecting ducts in gentamicin-induced kidney injury presents a potential new therapeutic strategy to alleviate gentamicin-induced AKI through inhibiting necroptosis.


Subject(s)
Acute Kidney Injury/chemically induced , Gentamicins/toxicity , Kidney Tubules, Collecting/drug effects , Necroptosis/drug effects , Animals , Cells, Cultured , Disease Models, Animal , Imidazoles/pharmacology , Indoles/pharmacology , Kidney Tubules, Collecting/pathology , Kidney Tubules, Collecting/ultrastructure , Mice , Mice, Inbred C57BL , Protein Kinases/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology
14.
Curr Opin Cell Biol ; 63: 186-193, 2020 04.
Article in English | MEDLINE | ID: mdl-32163825

ABSTRACT

Necroptosis and pyroptosis are inflammatory forms of regulated necrotic cell death as opposed to apoptosis that is generally considered immunologically silent. Recent studies revealed unexpected links in the pathways regulating and executing cell death in response to activation of signaling cascades inducing apoptosis, necroptosis, and pyroptosis. Emerging evidence suggests that receptor interacting protein kinase 1 and caspase-8 control the cross-talk between apoptosis, necroptosis, and pyroptosis and determine the type of cell death induced in response to activation of cell death signaling.


Subject(s)
Apoptosis/genetics , Caspase 8/physiology , Necroptosis/genetics , Pyroptosis/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Caspase 8/genetics , Caspase 8/metabolism , Humans , Necrosis/genetics , Necrosis/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics
15.
Cell ; 180(6): 1115-1129.e13, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32200799

ABSTRACT

Influenza A virus (IAV) is a lytic RNA virus that triggers receptor-interacting serine/threonine-protein kinase 3 (RIPK3)-mediated pathways of apoptosis and mixed lineage kinase domain-like pseudokinase (MLKL)-dependent necroptosis in infected cells. ZBP1 initiates RIPK3-driven cell death by sensing IAV RNA and activating RIPK3. Here, we show that replicating IAV generates Z-RNAs, which activate ZBP1 in the nucleus of infected cells. ZBP1 then initiates RIPK3-mediated MLKL activation in the nucleus, resulting in nuclear envelope disruption, leakage of DNA into the cytosol, and eventual necroptosis. Cell death induced by nuclear MLKL was a potent activator of neutrophils, a cell type known to drive inflammatory pathology in virulent IAV disease. Consequently, MLKL-deficient mice manifest reduced nuclear disruption of lung epithelia, decreased neutrophil recruitment into infected lungs, and increased survival following a lethal dose of IAV. These results implicate Z-RNA as a new pathogen-associated molecular pattern and describe a ZBP1-initiated nucleus-to-plasma membrane "inside-out" death pathway with potentially pathogenic consequences in severe cases of influenza.


Subject(s)
Influenza A virus/genetics , Necroptosis/genetics , RNA-Binding Proteins/metabolism , Animals , Apoptosis/genetics , Cell Death/genetics , Cell Line, Tumor , Female , Influenza A virus/metabolism , Male , Mice , Mice, Inbred C57BL , Necrosis/metabolism , Phosphorylation , Protein Kinases/metabolism , RNA/metabolism , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/physiology
16.
Dis Model Mech ; 13(1)2020 01 24.
Article in English | MEDLINE | ID: mdl-31953345

ABSTRACT

Receptor-interacting protein kinase 3 (RIPK3) was recently implicated in promoting atherosclerosis progression through a proposed role in macrophage necroptosis. However, RIPK3 has been connected to numerous other cellular pathways, which raises questions about its actual role in atherosclerosis. Furthermore, RIPK3 is expressed in a multitude of cell types, suggesting that it may be physiologically relevant to more than just macrophages in atherosclerosis. In this study, Ripk3 was deleted in macrophages, endothelial cells, vascular smooth muscle cells or globally on the Apoe-/- background using Cre-lox technology. To induce atherosclerosis progression, male and female mice were fed a Western diet for three months before tissue collection and analysis. Surprisingly, necroptosis markers were nearly undetectable in atherosclerotic aortas. Furthermore, en face lesion area was increased in macrophage- and endothelial-specific deletions of Ripk3 in the descending and abdominal regions of the aorta. Analysis of bone-marrow-derived macrophages and cultured endothelial cells revealed that Ripk3 deletion promotes expression of monocyte chemoattractant protein 1 (MCP-1) and E-selectin in these cell types, respectively. Western blot analysis showed upregulation of MCP-1 in aortas with Ripk3-deficient macrophages. Altogether, these data suggest that RIPK3 in macrophages and endothelial cells protects against atherosclerosis through a mechanism that likely does not involve necroptosis. This protection may be due to RIPK3-mediated suppression of pro-inflammatory MCP-1 expression in macrophages and E-selectin expression in endothelial cells. These findings suggest a novel and unexpected cell-type specific and athero-protective function for RIPK3.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Atherosclerosis/prevention & control , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Atherosclerosis/etiology , Chemokine CCL2/analysis , Chemokine CCL2/physiology , Disease Models, Animal , E-Selectin/analysis , Endothelial Cells/physiology , Interleukin-1beta/blood , Interleukin-1beta/physiology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Necroptosis
17.
Nat Commun ; 11(1): 475, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980622

ABSTRACT

We reported previously that acid-sensing ion channel 1a (ASIC1a) mediates acidic neuronal necroptosis via recruiting receptor-interacting protein kinase 1 (RIPK1) to its C terminus (CT), independent of its ion-conducting function. Here we show that the N-terminus (NT) of ASIC1a interacts with its CT to form an auto-inhibition that prevents RIPK1 recruitment/activation under resting conditions. The interaction involves glutamate residues at distal NT and is disrupted by acidosis. Expression of mutant ASIC1a bearing truncation or glutamate-to-alanine substitutions at distal NT causes constitutive cell death. The NT-CT interaction is further disrupted by N-ethylmaleimide-sensitive fusion ATPase (NSF), which associates with ASIC1a-NT under acidosis, facilitating RIPK1 interaction with ASIC1a-CT. Importantly, a membrane-penetrating synthetic peptide representing the distal 20 ASIC1a NT residues, NT1-20, reduced neuronal damage in both in vitro model of acidotoxicity and in vivo mouse model of ischemic stroke, demonstrating the therapeutic potential of targeting the auto-inhibition of ASIC1a for neuroprotection against acidotoxicity.


Subject(s)
Acid Sensing Ion Channels/chemistry , Acid Sensing Ion Channels/physiology , Necroptosis/physiology , Neurons/physiology , Acid Sensing Ion Channels/genetics , Acidosis/pathology , Acidosis/physiopathology , Amino Acid Substitution , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Models, Neurological , N-Ethylmaleimide-Sensitive Proteins/pharmacology , N-Ethylmaleimide-Sensitive Proteins/physiology , Neurons/cytology , Neuroprotection/drug effects , Neuroprotection/physiology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/physiology , Protein Conformation , Protein Interaction Domains and Motifs , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Signal Transduction , Stroke/pathology , Stroke/physiopathology
18.
Gut ; 69(1): 133-145, 2020 01.
Article in English | MEDLINE | ID: mdl-31409605

ABSTRACT

OBJECTIVE: The Fragile X mental retardation (FMR) syndrome is a frequently inherited intellectual disability caused by decreased or absent expression of the FMR protein (FMRP). Lack of FMRP is associated with neuronal degradation and cognitive dysfunction but its role outside the central nervous system is insufficiently studied. Here, we identify a role of FMRP in liver disease. DESIGN: Mice lacking Fmr1 gene expression were used to study the role of FMRP during tumour necrosis factor (TNF)-induced liver damage in disease model systems. Liver damage and mechanistic studies were performed using real-time PCR, Western Blot, staining of tissue sections and clinical chemistry. RESULTS: Fmr1null mice exhibited increased liver damage during virus-mediated hepatitis following infection with the lymphocytic choriomeningitis virus. Exposure to TNF resulted in severe liver damage due to increased hepatocyte cell death. Consistently, we found increased caspase-8 and caspase-3 activation following TNF stimulation. Furthermore, we demonstrate FMRP to be critically important for regulating key molecules in TNF receptor 1 (TNFR1)-dependent apoptosis and necroptosis including CYLD, c-FLIPS and JNK, which contribute to prolonged RIPK1 expression. Accordingly, the RIPK1 inhibitor Necrostatin-1s could reduce liver cell death and alleviate liver damage in Fmr1null mice following TNF exposure. Consistently, FMRP-deficient mice developed increased pathology during acute cholestasis following bile duct ligation, which coincided with increased hepatic expression of RIPK1, RIPK3 and phosphorylation of MLKL. CONCLUSIONS: We show that FMRP plays a central role in the inhibition of TNF-mediated cell death during infection and liver disease.


Subject(s)
Fragile X Mental Retardation Protein/physiology , Hepatitis, Viral, Animal/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/pathology , CD8-Positive T-Lymphocytes/immunology , Cell Death/drug effects , Cell Death/immunology , Cell Death/physiology , Cells, Cultured , Cholestasis/immunology , Cholestasis/metabolism , Cholestasis/pathology , Fragile X Mental Retardation Protein/metabolism , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/prevention & control , Hepatocytes/pathology , Imidazoles/pharmacology , Imidazoles/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Lymphocytic choriomeningitis virus , Male , Mice, Inbred C57BL , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/physiology
19.
Cell Death Differ ; 27(1): 297-309, 2020 01.
Article in English | MEDLINE | ID: mdl-31164717

ABSTRACT

The Siva protein, named after the Hindu God of Destruction, plays important roles in apoptosis in various contexts, including downstream of death receptor activation or p53 tumor suppressor engagement. The function of Siva in organismal development and homeostasis, however, has remained uncharacterized. Here, we generate Siva knockout mice to characterize the physiological function of Siva in vivo. Interestingly, we find that Siva deficiency causes early embryonic lethality accompanied by multiple phenotypes, including developmental delay, abnormal neural tube closure, and defective placenta and yolk sac formation. Examination of Siva expression during embryogenesis shows that Siva is expressed in both embryonic and extra-embryonic tissues, including within the mesoderm, which may explain the vascular defects observed in the placenta and yolk sac. The embryonic phenotypes caused by Siva loss are not rescued by p53 deficiency, nor do they resemble those of p53 null embryos, suggesting that the embryonic function of Siva is not related to the p53 pathway. Moreover, loss of the Ripk3 necroptosis protein does not rescue the observed lethality or developmental defects, suggesting that Siva may play a non-apoptotic role in development. Collectively, these studies reveal a key role for Siva in proper embryonic development.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Embryonic Development , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Embryo, Mammalian/blood supply , Embryo, Mammalian/metabolism , Female , Genes, Lethal , Heart/embryology , Mesoderm/metabolism , Mice , Mice, Knockout , Neural Tube/abnormalities , Phenotype , Placenta/blood supply , Pregnancy , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p53/physiology , Yolk Sac/blood supply
20.
Cell Death Differ ; 27(1): 161-175, 2020 01.
Article in English | MEDLINE | ID: mdl-31101885

ABSTRACT

The kinase RIP1 acts in multiple signaling pathways to regulate inflammatory responses and it can trigger both apoptosis and necroptosis. Its kinase activity has been implicated in a range of inflammatory, neurodegenerative, and oncogenic diseases. Here, we explore the effect of inhibiting RIP1 genetically, using knock-in mice that express catalytically inactive RIP1 D138N, or pharmacologically, using the murine-potent inhibitor GNE684. Inhibition of RIP1 reduced collagen antibody-induced arthritis, and prevented skin inflammation caused by mutation of Sharpin, or colitis caused by deletion of Nemo from intestinal epithelial cells. Conversely, inhibition of RIP1 had no effect on tumor growth or survival in pancreatic tumor models driven by mutant Kras, nor did it reduce lung metastases in a B16 melanoma model. Collectively, our data emphasize a role for the kinase activity of RIP1 in certain inflammatory disease models, but question its relevance to tumor progression and metastases.


Subject(s)
Inflammation/enzymology , Neoplasms/enzymology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Arthritis/enzymology , Cell Death , Cell Line , Cell Line, Tumor , Colitis/etiology , Colitis/prevention & control , Dermatitis/enzymology , Female , Gene Knock-In Techniques , Humans , Ileitis/etiology , Ileitis/prevention & control , Intracellular Signaling Peptides and Proteins/genetics , Male , Melanoma, Experimental/pathology , Mice , Neoplasm Metastasis , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rats , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...