Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Mucosal Immunol ; 14(5): 1160-1171, 2021 09.
Article in English | MEDLINE | ID: mdl-34045680

ABSTRACT

Regulation of epithelial cell death has emerged as a key mechanism controlling immune homeostasis in barrier surfaces. Necroptosis is a type of regulated necrotic cell death induced by receptor interacting protein kinase 3 (RIPK3) that has been shown to cause inflammatory pathologies in different tissues. The role of regulated cell death and particularly necroptosis in lung homeostasis and disease remains poorly understood. Here we show that mice with Airway Epithelial Cell (AEC)-specific deficiency of Fas-associated with death domain (FADD), an adapter essential for caspase-8 activation, developed exacerbated allergic airway inflammation in a mouse model of asthma induced by sensitization and challenge with house dust mite (HDM) extracts. Genetic inhibition of RIPK1 kinase activity by crossing to mice expressing kinase inactive RIPK1 as well as RIPK3 or MLKL deficiency prevented the development of exaggerated HDM-induced asthma pathology in FADDAEC-KO mice, suggesting that necroptosis of FADD-deficient AECs augmented the allergic immune response. These results reveal a role of AEC necroptosis in amplifying airway allergic inflammation and suggest that necroptosis could contribute to asthma exacerbations caused by respiratory virus infections inducing AEC death.


Subject(s)
Allergens/immunology , Asthma/etiology , Asthma/metabolism , Necroptosis/immunology , Pyroglyphidae/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Airway Remodeling , Animals , Asthma/pathology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers , Disease Models, Animal , Disease Progression , Disease Susceptibility , Enzyme Activation , Fas-Associated Death Domain Protein/deficiency , Immunoglobulin E/immunology , Immunohistochemistry , Mice , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Respiratory Mucosa/pathology
2.
Mol Cell ; 81(2): 370-385.e7, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33271062

ABSTRACT

The mechanisms of cellular energy sensing and AMPK-mediated mTORC1 inhibition are not fully delineated. Here, we discover that RIPK1 promotes mTORC1 inhibition during energetic stress. RIPK1 is involved in mediating the interaction between AMPK and TSC2 and facilitate TSC2 phosphorylation at Ser1387. RIPK1 loss results in a high basal mTORC1 activity that drives defective lysosomes in cells and mice, leading to accumulation of RIPK3 and CASP8 and sensitization to cell death. RIPK1-deficient cells are unable to cope with energetic stress and are vulnerable to low glucose levels and metformin. Inhibition of mTORC1 rescues the lysosomal defects and vulnerability to energetic stress and prolongs the survival of RIPK1-deficient neonatal mice. Thus, RIPK1 plays an important role in the cellular response to low energy levels and mediates AMPK-mTORC1 signaling. These findings shed light on the regulation of mTORC1 during energetic stress and unveil a point of crosstalk between pro-survival and pro-death pathways.


Subject(s)
Autophagy-Related Protein 5/genetics , Fas-Associated Death Domain Protein/genetics , Intestine, Large/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Animals, Newborn , Autophagy-Related Protein 5/deficiency , Caspase 8/genetics , Caspase 8/metabolism , Cell Death/genetics , Fas-Associated Death Domain Protein/deficiency , Gene Expression Regulation , Glucose/antagonists & inhibitors , Glucose/pharmacology , HEK293 Cells , HT29 Cells , Humans , Intestine, Large/drug effects , Intestine, Large/pathology , Jurkat Cells , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Metformin/antagonists & inhibitors , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Signal Transduction , Sirolimus/pharmacology , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism
3.
Eur J Med Chem ; 201: 112337, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32659605

ABSTRACT

With the aim to develop new chemical tools based on simplified natural metabolites to help deciphering the molecular mechanism of necroptosis, simplified benzazole fragments including 2-aminobenzimidazole and the 2-aminobenzothiazole analogs were prepared during the synthesis of the marine benzosceptrin B. Conpounds inhibiting the RIPK1 protein kinase were discovered. A library of 54 synthetic analogs were prepared and evaluated through a phenotypic screen using the inhibition of the necrotic cell death induced by TNF-α in human Jurkat T cells deficient for the FADD protein. This article reports the design, synthesis and biological evaluation of a series of 2-aminobenzazoles on the necroptotic cell death through the inhibition of RIPK1 protein kinase. The 2-aminobenzimidazole and 2-aminobenzothiazole platforms presented herein can serve as novel chemical tools to study the molecular regulation of necroptosis and further develop lead drug candidates for chronic pathologies involving necroptosis.


Subject(s)
Imidazoles/pharmacology , Necroptosis/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Binding Sites , Drug Design , Fas-Associated Death Domain Protein/deficiency , Humans , Imidazoles/chemical synthesis , Imidazoles/metabolism , Jurkat Cells , Molecular Docking Simulation , Molecular Structure , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Pyrroles/chemical synthesis , Pyrroles/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Structure-Activity Relationship
4.
J Mol Med (Berl) ; 98(4): 569-583, 2020 04.
Article in English | MEDLINE | ID: mdl-32072232

ABSTRACT

Necrosis with inflammation plays a crucial role in acute respiratory distress syndrome (ARDS). Receptor-interacting protein 3 (RIPK3) regulates a newly discovered programmed form of necrosis called necroptosis. However, the underlying mechanism of necroptosis in ARDS remains unknown. Thus, the purpose of this study was to examine the possible involvement of RIPK3 in ARDS-associated necroptosis. RIPK3 protein levels were found to be significantly elevated in the plasma and bronchoalveolar lavage fluid of ARDS patients. Next, we utilised a mouse model of severe ARDS induced with high-dose lipopolysaccharide and found that lung injury was mainly due to RIPK3-mixed lineage kinase domain-like pseudokinase (MLKL)-mediated necroptosis and endothelial dysfunction. The activation of RIPK3-MLKL by tumour necrosis factor receptor 1 (TNFR1) and TNFR1-associated death domain protein (TRADD) required catalytically active RIPK1 and the inhibition of Fas-associated protein with death domain (FADD)/caspase-8 catalytic activity. We further showed that the molecular chaperone heat shock protein 90 (Hsp90)/p23, as a novel RIPK3- and MLKL-interacting complex, played an important role in RIP-MLKL-mediated necroptosis, inflammation and endothelial dysfunction in the pulmonary vasculature, which resulted in ARDS. Collectively, the results of our study indicate that necroptosis is an important mechanism of cell death in ARDS and the inhibition of necroptosis may be a therapeutic intervention for ARDS. KEY MESSAGES: Lung injury in high-dose LPS-induced severe ARDS is mainly due to RIP3-MLKL-mediated necroptosis and endothelial dysfunction. Chaperone HSP90/p23 is a novel RIP3- and MLKL-interacting complex in HPAECs. HSP90/p23 is a novel RIP3- and MLKL-interacting complex in RIP-MLKL-mediated necroptosis, inflammation and endothelial dysfunction.


Subject(s)
Endothelial Cells/metabolism , HSP90 Heat-Shock Proteins/metabolism , Multiprotein Complexes/metabolism , Prostaglandin-E Synthases/metabolism , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Fas-Associated Death Domain Protein/deficiency , Gene Expression , Humans , Mice , Necroptosis/drug effects , Necroptosis/genetics , Prostaglandin-E Synthases/blood , Protein Binding , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Respiratory Distress Syndrome/pathology , Respiratory Mucosa
5.
J Photochem Photobiol B ; 194: 32-45, 2019 May.
Article in English | MEDLINE | ID: mdl-30904584

ABSTRACT

Sun or therapy-related ultraviolet B (UVB) irradiation induces different cell death modalities such as apoptosis, necrosis/necroptosis and autophagy. Understanding of mechanisms implicated in regulation and execution of cell death program is imperative for prevention and treatment of skin diseases. An essential component of death-inducing complex is Fas-associated protein with death domain (FADD), involved in conduction of death signals of different death modalities. The purpose of this study was to enlighten the role of FADD in the selection of cell death mode after narrow-band UVB (NB-UVB) irradiation using specific cell death inhibitors (carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]- fluoromethylketone (zVAD-fmk), Necrostatin-1 and 3-Methyladenine) and FADD-deficient (FADD-/-) mouse embryonic fibroblasts (MEFs) and their wild type (wt) counterparts. The results imply that lack of FADD sensitized MEFs to induction of receptor-interacting protein 1 (RIPK1)-dependent apoptosis by the generation of reactive oxygen species (ROS), but without activation of the proteins p53, Bax and Bcl-2 as well as without the enrolment of calpain-2. Autophagy was established as a contributing factor to NB-UVB-induced death execution. By contrast, wt cells triggered intrinsic apoptotic pathway that was resistant to the inhibition by zVAD-fmk and Necrostatin-1 pointing to the mechanism overcoming the cell survival. These findings support the role of FADD in prevention of autophagy-dependent apoptosis.


Subject(s)
Apoptosis/radiation effects , Autophagy/radiation effects , Fas-Associated Death Domain Protein/deficiency , Fibroblasts/cytology , Fibroblasts/radiation effects , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Ultraviolet Rays , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Caspases/metabolism , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Damage , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Imidazoles/pharmacology , Indoles/pharmacology , Mice , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
6.
Cell Death Differ ; 24(8): 1459-1469, 2017 08.
Article in English | MEDLINE | ID: mdl-28574501

ABSTRACT

RIP1 is an essential regulator of TNF-induced signaling complexes mediating NF-κB activation, apoptosis and necroptosis. Loss of Rip1 rescues the embryonic lethality of Fadd or Caspase-8-deficient mice, even though the double knockout mice die shortly after birth like Rip1-deficient mice. Recent studies demonstrated that mice expressing RIP1 kinase-dead mutants developed normally and resisted necroptotic stimuli in vitro and in vivo. However, the impact of RIP1 kinase activity on Fadd-/- embryonic development remains unknown. Here, we engineered two RIP1 kinase inactive mutant mouse lines, a Rip1K45A/K45A mouse line as previously reported and a novel Rip1Δ/Δ mouse line with an altered P-loop in the kinase domain. While RIP1K45A could not rescue the embryonic lethality of Fadd-deficient mice at E11.5, RIP1Δ rescued lethality of Fadd-/- mice at E11.5 and Fadd-/-Rip1Δ/Δ mice eventually died at E16.5 due to excessive death of fetal liver cells and unregulated inflammation. Under necropotosis-inducing conditions, comparing to Rip1K45A/K45A cells, Rip1Δ/Δcells displayed reduced phosphorylation and oligomerization of RIP3 and MLKL, which lead to increased cell viability. Thus, our study provides genetic evidence that different kinase inactive mutations have distinct impacts on the embryogenesis of Fadd-deficient mice, which might attribute to their extents of protection on necroptosis signaling.


Subject(s)
Embryonic Development/genetics , Fas-Associated Death Domain Protein/genetics , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Developmental , Animals , Embryo, Mammalian , Fas-Associated Death Domain Protein/deficiency , Female , GTPase-Activating Proteins/deficiency , Genes, Lethal , Mice , Mice, Knockout , Mutation , Phosphorylation , Pregnancy , Protein Multimerization , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction
7.
Mol Pharmacol ; 92(1): 30-47, 2017 07.
Article in English | MEDLINE | ID: mdl-28461585

ABSTRACT

Pannexin-1 (Panx1) channels mediate the efflux of ATP and AMP from cancer cells in response to induction of extrinsic apoptosis by death receptors or intrinsic apoptosis by chemotherapeutic agents. We previously described the accumulation of extracellular ATP /AMP during chemotherapy-induced apoptosis in Jurkat human leukemia cells. In this study, we compared how different signaling pathways determine extracellular nucleotide pools in control Jurkat cells versus Jurkat lines that lack the Fas-associated death domain (FADD) or receptor-interacting protein kinase 1 (RIP1) cell death regulatory proteins. Tumor necrosis factor-α induced extrinsic apoptosis in control Jurkat cells and necroptosis in FADD-deficient cells; treatment of both lines with chemotherapeutic drugs elicited similar intrinsic apoptosis. Robust extracellular ATP/AMP accumulation was observed in the FADD-deficient cells during necroptosis, but not during apoptotic activation of Panx1 channels. Accumulation of extracellular ATP/AMP was similarly absent in RIP1-deficient Jurkat cells during apoptotic responses to chemotherapeutic agents. Apoptotic activation triggered equivalent proteolytic gating of Panx1 channels in all three Jurkat cell lines. The differences in extracellular ATP/AMP accumulation correlated with cell-line-specific expression of ectonucleotidases that metabolized the released ATP/AMP. CD73 mRNA, and αß-methylene-ADP-inhibitable ecto-AMPase activity were elevated in the FADD-deficient cells. In contrast, the RIP1-deficient cells were defined by increased expression of tartrate-sensitive prostatic acid phosphatase as a broadly acting ectonucleotidase. Thus, extracellular nucleotide accumulation during regulated tumor cell death involves interplay between ATP/AMP efflux pathways and different cell-autonomous ectonucleotidases. Differential expression of particular ectonucleotidases in tumor cell variants will determine whether chemotherapy-induced activation of Panx1 channels drives accumulation of immunostimulatory ATP versus immunosuppressive adenosine within the tumor microenvironment.


Subject(s)
Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Antineoplastic Agents/toxicity , Connexins/metabolism , Fas-Associated Death Domain Protein/deficiency , Nerve Tissue Proteins/metabolism , Nuclear Pore Complex Proteins/deficiency , Apoptosis/drug effects , Apoptosis/physiology , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Humans , Jurkat Cells , RNA-Binding Proteins , Up-Regulation/drug effects , Up-Regulation/physiology
8.
Cell Rep ; 19(4): 798-808, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28445730

ABSTRACT

RIPK3 mediates cell death and regulates inflammatory responses. Although genetic studies have suggested that RIPK3-MLKL-mediated necroptosis leads to embryonic lethality in Fadd or Caspase-8-deficient mice, the exact mechanisms are not fully understood. Here, we generated Ripk3 mutant mice by altering the RIPK3 kinase domain (Ripk3Δ/Δ mice), thus abolishing its kinase activity. Ripk3Δ/Δ cells were resistant to necroptosis stimulation in vitro, and Ripk3Δ/Δ mice were protected from necroptotic diseases. Although the Ripk3Δ/Δ mutation rescued embryonic lethality in Fadd-/- embryos, Fadd-/-Ripk3Δ/Δ mice died within 1 day after birth due to massive inflammation. These results indicate that Ripk3 ablation rescues embryonic lethality in Fadd-deficient mice by suppressing two RIPK3-mediating processes: necroptosis during embryogenesis and inflammation during postnatal development in Fadd-/- mice.


Subject(s)
Apoptosis , Fas-Associated Death Domain Protein/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis/drug effects , Caspase 8/genetics , Caspase 8/metabolism , Ceruletide/toxicity , Chemokines/metabolism , Cytokines/analysis , Cytokines/metabolism , Embryo, Mammalian/metabolism , Embryonic Development/drug effects , Fas-Associated Death Domain Protein/deficiency , HEK293 Cells , Humans , Inflammation , Lipopolysaccharides/toxicity , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Knockout , Mutagenesis , Necrosis , Oligopeptides/pharmacology , Pancreatitis/chemically induced , Pancreatitis/pathology , Phosphorylation , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
10.
Oncotarget ; 7(17): 24572-84, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27013580

ABSTRACT

FADD (Fas-associated protein with death domain) is a classical adaptor protein in apoptosis. Increasing evidences have shown that FADD is also implicated in cell cycle progression, proliferation and tumorigenesis. The role of FADD in cancer remains largely unexplored. In this study, In Silico Analysis using Oncomine and Kaplan Meier plotter revealed that FADD is significantly up-regulated in breast cancer tissues and closely associated with a poor prognosis in patients with breast cancer. To better understanding the FADD functions in breast cancer, we performed proteomics analysis by LC-MS/MS detection and found that Rheb-mTORC1 pathway was dysregulated in MCF-7 cells when FADD knockdown. The mTORC1 pathway is a key regulator in many processes, including cell growth, metabolism and autophagy. Here, FADD interference down-regulated Rheb expression and repressed mTORC1 activity in breast cancer cell lines. The autophagy was induced by FADD deficiency in MCF7 or MDA-231 cells but rescued by recovering Rheb expression. Similarly, growth defect in FADD-knockdown cells was also restored by Rheb overexpression. These findings implied a novel role of FADD in tumor progression via Rheb-mTORC1 pathway in breast cancer.


Subject(s)
Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Fas-Associated Death Domain Protein/metabolism , Ras Homolog Enriched in Brain Protein/biosynthesis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Autophagy/physiology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Fas-Associated Death Domain Protein/deficiency , Fas-Associated Death Domain Protein/genetics , Female , Humans , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 1/metabolism , Ras Homolog Enriched in Brain Protein/genetics , Signal Transduction
11.
Cell Rep ; 13(9): 2027-36, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26655912

ABSTRACT

Apoptosis is generally believed to be a process that requires several hours, in contrast to non-programmed forms of cell death that can occur in minutes. Our findings challenge the time-consuming nature of apoptosis as we describe the discovery and characterization of a small molecule, named Raptinal, which initiates intrinsic pathway caspase-dependent apoptosis within minutes in multiple cell lines. Comparison to a mechanistically diverse panel of apoptotic stimuli reveals that Raptinal-induced apoptosis proceeds with unparalleled speed. The rapid phenotype enabled identification of the critical roles of mitochondrial voltage-dependent anion channel function, mitochondrial membrane potential/coupled respiration, and mitochondrial complex I, III, and IV function for apoptosis induction. Use of Raptinal in whole organisms demonstrates its utility for studying apoptosis in vivo for a variety of applications. Overall, rapid inducers of apoptosis are powerful tools that will be used in a variety of settings to generate further insight into the apoptotic machinery.


Subject(s)
Apoptosis , Cyclopentanes/chemistry , Fluorenes/chemistry , Animals , Apoptosis/drug effects , Caspase 3/chemistry , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Complement C8/deficiency , Complement C8/genetics , Cyclopentanes/pharmacokinetics , Cyclopentanes/toxicity , Embryo, Nonmammalian/metabolism , Fas-Associated Death Domain Protein/deficiency , Fas-Associated Death Domain Protein/genetics , Fluorenes/pharmacokinetics , Fluorenes/toxicity , Half-Life , Humans , Jurkat Cells , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Zebrafish
12.
Oncogene ; 34(47): 5796-806, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-25867066

ABSTRACT

Necroptosis represents a key programmed cell death pathway involved in various physiological and pathophysiological conditions. However, the role of reactive oxygen species (ROS) in necroptotic signaling has remained unclear. In the present study, we identify ROS as critical regulators of BV6/tumor necrosis factor-α (TNFα)-induced necroptotic signaling and cell death. We show that BV6/TNFα-induced cell death depends on ROS production, as several ROS scavengers such as butylated hydroxyanisole, N-acetylcysteine, α-tocopherol and ethyl pyruvate significantly rescue cell death. Before cell death, BV6/TNFα-stimulated ROS generation promotes stabilization of the receptor-interacting protein kinase 1 (RIP1)/RIP3 necrosome complex via a potential positive feedback loop, as on the one hand radical scavengers attenuate RIP1/RIP3 necrosome assembly and phosphorylation of mixed lineage kinase domain like (MLKL), but on the other hand silencing of RIP1 or RIP3 reduces ROS production. Although MLKL knockdown effectively decreases BV6/TNFα-induced cell death, it does not affect RIP1/RIP3 interaction and only partly reduces ROS generation. Moreover, the deubiquitinase cylindromatosis (CYLD) promotes BV6/TNFα-induced ROS generation and necrosome assembly even in the presence of BV6, as CYLD silencing attenuates these events. Genetic silencing of phosphoglycerate mutase 5 or dynamin-related protein 1 (Drp1) fails to protect against BV6/TNFα-induced cell death. By demonstrating that ROS are involved in regulating BV6/TNFα-induced necroptotic signaling, our study provides new insights into redox regulation of necroptosis.


Subject(s)
Oligopeptides/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Cell Death/drug effects , Fas-Associated Death Domain Protein/deficiency , Humans , Jurkat Cells , Nuclear Pore Complex Proteins/metabolism , Phosphorylation , Protein Kinases/metabolism , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
14.
Nature ; 513(7516): 90-4, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25132550

ABSTRACT

Necroptosis has emerged as an important pathway of programmed cell death in embryonic development, tissue homeostasis, immunity and inflammation. RIPK1 is implicated in inflammatory and cell death signalling and its kinase activity is believed to drive RIPK3-mediated necroptosis. Here we show that kinase-independent scaffolding RIPK1 functions regulate homeostasis and prevent inflammation in barrier tissues by inhibiting epithelial cell apoptosis and necroptosis. Intestinal epithelial cell (IEC)-specific RIPK1 knockout caused IEC apoptosis, villus atrophy, loss of goblet and Paneth cells and premature death in mice. This pathology developed independently of the microbiota and of MyD88 signalling but was partly rescued by TNFR1 (also known as TNFRSF1A) deficiency. Epithelial FADD ablation inhibited IEC apoptosis and prevented the premature death of mice with IEC-specific RIPK1 knockout. However, mice lacking both RIPK1 and FADD in IECs displayed RIPK3-dependent IEC necroptosis, Paneth cell loss and focal erosive inflammatory lesions in the colon. Moreover, a RIPK1 kinase inactive knock-in delayed but did not prevent inflammation caused by FADD deficiency in IECs or keratinocytes, showing that RIPK3-dependent necroptosis of FADD-deficient epithelial cells only partly requires RIPK1 kinase activity. Epidermis-specific RIPK1 knockout triggered keratinocyte apoptosis and necroptosis and caused severe skin inflammation that was prevented by RIPK3 but not FADD deficiency. These findings revealed that RIPK1 inhibits RIPK3-mediated necroptosis in keratinocytes in vivo and identified necroptosis as a more potent trigger of inflammation compared with apoptosis. Therefore, RIPK1 is a master regulator of epithelial cell survival, homeostasis and inflammation in the intestine and the skin.


Subject(s)
Apoptosis , Epithelial Cells/cytology , Epithelial Cells/pathology , Homeostasis , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Caspase 8/metabolism , Cell Survival , Epithelial Cells/metabolism , Fas-Associated Death Domain Protein/deficiency , Fas-Associated Death Domain Protein/metabolism , Female , Inflammation/metabolism , Inflammation/pathology , Intestinal Mucosa/metabolism , Intestines/cytology , Intestines/pathology , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Paneth Cells/metabolism , Paneth Cells/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/metabolism , Skin/cytology , Skin/metabolism , Skin/pathology
15.
Biochem Biophys Res Commun ; 451(2): 202-7, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-25078620

ABSTRACT

Fas-associated protein with death domain (FADD) has been implicated in T lymphocytes, but the nature of FADD-dependent mechanism in early T cell development has not been completely elucidated. In this study, using T-cell specific deletion mice, we observed that FADD deficiency in thymocytes led to increased apoptosis and reduced cell numbers, which may be attributed to the reduction of Glut1 expression and correspondingly decreased glucose uptake. Furthermore, an abnormal transduction of Akt signaling was discovered in FADD(-/-) thymocytes, which may be responsible for the declined Glut1 expression. Collectively, our results demonstrate the new function of FADD in glucose metabolism and survival of early T cells.


Subject(s)
Fas-Associated Death Domain Protein/metabolism , Glucose/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , Apoptosis , Biological Transport, Active , Cell Differentiation , Cell Survival , Cells, Cultured , Fas-Associated Death Domain Protein/deficiency , Fas-Associated Death Domain Protein/genetics , Glucose Transporter Type 1/metabolism , Mice , Mice, Knockout , Models, Biological , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
16.
PLoS One ; 8(9): e73537, 2013.
Article in English | MEDLINE | ID: mdl-24058479

ABSTRACT

AIM: As technological interventions treating acute myocardial infarction (MI) improve, post-ischemic heart failure increasingly threatens patient health. The aim of the current study was to test whether FADD could be a potential target of gene therapy in the treatment of heart failure. METHODS: Cardiomyocyte-specific FADD knockout mice along with non-transgenic littermates (NLC) were subjected to 30 minutes myocardial ischemia followed by 7 days of reperfusion or 6 weeks of permanent myocardial ischemia via the ligation of left main descending coronary artery. Cardiac function were evaluated by echocardiography and left ventricular (LV) catheterization and cardiomyocyte death was measured by Evans blue-TTC staining, TUNEL staining, and caspase-3, -8, and -9 activities. In vitro, H9C2 cells transfected with ether scramble siRNA or FADD siRNA were stressed with chelerythrin for 30 min and cleaved caspase-3 was assessed. RESULTS: FADD expression was significantly decreased in FADD knockout mice compared to NLC. Ischemia/reperfusion (I/R) upregulated FADD expression in NLC mice, but not in FADD knockout mice at the early time. FADD deletion significantly attenuated I/R-induced cardiac dysfunction, decreased myocardial necrosis, and inhibited cardiomyocyte apoptosis. Furthermore, in 6 weeks long term permanent ischemia model, FADD deletion significantly reduced the infarct size (from 41.20 ± 3.90% in NLC to 26.83 ± 4.17% in FADD deletion), attenuated myocardial remodeling, improved cardiac function and improved survival. In vitro, FADD knockdown significantly reduced chelerythrin-induced the level of cleaved caspase-3. CONCLUSION: Taken together, our results suggest FADD plays a critical role in post-ischemic heart failure. Inhibition of FADD retards heart failure progression. Our data supports the further investigation of FADD as a potential target for genetic manipulation in the treatment of heart failure.


Subject(s)
Fas-Associated Death Domain Protein/genetics , Heart Failure/genetics , Myocardial Reperfusion Injury/genetics , Myocytes, Cardiac/metabolism , Ventricular Dysfunction, Left/genetics , Animals , Apoptosis/drug effects , Benzophenanthridines/pharmacology , Caspase 3/genetics , Caspase 3/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cells, Cultured , Coronary Vessels/surgery , Disease Models, Animal , Fas-Associated Death Domain Protein/antagonists & inhibitors , Fas-Associated Death Domain Protein/deficiency , Gene Expression Regulation , Heart Failure/metabolism , Heart Failure/pathology , Male , Mice , Mice, Knockout , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Severity of Illness Index , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology
17.
Mol Biosyst ; 9(8): 2063-78, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23689606

ABSTRACT

Fas-associated death domain (FADD) is a classical adaptor protein involved in tumor necrosis factor receptor family-mediated apoptosis. Besides being an essential instrument in cell death, it also plays key roles in cell proliferation and survival. The current study shows for the first time that FADD is probably associated with energy metabolism and proteolysis. It has been reported that embryonic death caused by FADD deficiency in mice was not attributable to impaired apoptosis. Furthermore, mice bearing the substitution in FADD of serine 191 to aspartic acid exhibited leaner body size than both wild-type control and serine 191 to alanine mutant mice, indicating metabolic disorders. To study these non-apoptotic effects of FADD, a comprehensive strategy of proteomics identification combined with bioinformatic analyses and further cell biology validation was utilized to identify differentially-expressed proteins in FADD-deficient mouse embryonic fibroblasts (MEFs). A total of 45 unique proteins were determined to be significantly changing due to FADD deficiency. Network analysis of these proteins using MetaCore™ suggested induction of transcriptional factors that are too low to be detected by two-dimensional gels and identified an enriched cluster of changed proteins that are involved in cellular metabolic processes, including lipid metabolism, fatty acids metabolism, glycolysis, tricarboxylic acid (TCA) cycle, and oxidative phosphorylation. Fatty acids ß-oxidation was found to be enhanced in FADD-deficient cells. In addition, five proteins related to the ubiquitin-proteasome (UP) pathway were also specifically up-regulated in the FADD(-/-) MEFs. Finally, the c-Myc gene represents a convergent hub lying at the center of dysregulated pathways, and was up-regulated in FADD knockout cells. Taken together, these studies show that impaired mitochondrial function and proteolysis may play pivotal roles in the dysfunction associated with FADD deficiency-induced disorders, probably including embryonic lethality. The link between FADD and cell metabolism may provide us new insight for understanding the crosstalk of independent cell signaling pathways.


Subject(s)
Fas-Associated Death Domain Protein/genetics , Fibroblasts/metabolism , Mitochondria/genetics , Proteasome Endopeptidase Complex/genetics , Proteome , Animals , Cell Line , Electrophoresis, Gel, Two-Dimensional , Embryo, Mammalian , Fas-Associated Death Domain Protein/deficiency , Fibroblasts/pathology , Gene Expression Regulation , Gene Knockout Techniques , Mice , Mitochondria/pathology , Molecular Sequence Annotation , Proteasome Endopeptidase Complex/metabolism , Protein Interaction Mapping , Proteolysis , Signal Transduction
18.
J Immunol ; 189(12): 5508-12, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23144495

ABSTRACT

Fas, a TNF family receptor, is activated by the membrane protein Fas ligand expressed on various immune cells. Fas signaling triggers apoptosis and induces inflammatory cytokine production. Among the Fas-induced cytokines, the IL-1ß family cytokines require proteolysis to gain biological activity. Inflammasomes, which respond to pathogens and danger signals, cleave IL-1ß cytokines via caspase-1. However, the mechanisms by which Fas regulates IL-1ß activation remain unresolved. In this article, we demonstrate that macrophages exposed to TLR ligands upregulate Fas, which renders them responsive to receptor engagement by Fas ligand. Fas signaling activates caspase-8 in macrophages and dendritic cells, leading to the maturation of IL-1ß and IL-18 independently of inflammasomes or RIP3. Hence, Fas controls a novel noncanonical IL-1ß activation pathway in myeloid cells, which could play an essential role in inflammatory processes, tumor surveillance, and control of infectious diseases.


Subject(s)
Caspase 8/physiology , Interleukin-18/biosynthesis , Interleukin-1beta/biosynthesis , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , fas Receptor/physiology , Animals , Caspase 8/genetics , Caspase 8/metabolism , Dendritic Cells/enzymology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Enzyme Activation/immunology , Fas-Associated Death Domain Protein/deficiency , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/physiology , Inflammasomes/metabolism , Inflammasomes/physiology , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Signal Transduction/immunology
19.
Cell Rep ; 1(5): 401-7, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22675671

ABSTRACT

Caspase-8, the initiator caspase of the death receptor pathway of apoptosis, its adapter molecule, FADD, required for caspase-8 activation, and cFLIPL, a caspase-8-like protein that lacks a catalytic site and blocks caspase-8-mediated apoptosis, are each essential for embryonic development. Animals deficient in any of these genes present with E10.5 embryonic lethality. Recent studies have shown that development in caspase-8-deficient mice is rescued by ablation of RIPK3, a kinase that promotes a form of programmed, necrotic cell death. Here, we show that FADD, RIPK3 double-knockout mice develop normally but that the lethal effects of cFLIP deletion are not rescued by RIPK3 deficiency. Remarkably, in mice lacking FADD, cFLIP, and RIPK3, embryonic development is normal. This can be explained by the convergence of two cell processes: the enzymatic activity of the FADD-caspase-8-cFLIPL complex blocks RIPK3-dependent signaling (including necrosis), whereas cFLIPL blocks RIPK3-independent apoptosis promoted by the FADD-caspase-8 complex.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Caspase 8/physiology , Embryonic Development/physiology , Fas-Associated Death Domain Protein/physiology , Signal Transduction/physiology , Animals , Apoptosis/genetics , Apoptosis/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspase 8/genetics , Embryonic Development/genetics , Fas-Associated Death Domain Protein/deficiency , Fas-Associated Death Domain Protein/genetics , Gene Deletion , Mice , Mice, Knockout , Necrosis/genetics , Necrosis/physiopathology , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Signal Transduction/genetics
20.
Neoplasia ; 13(10): 971-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22028622

ABSTRACT

Searching for new strategies to bypass apoptosis resistance, we investigated the potential of the Smac mimetic BV6 in Jurkat leukemia cells deficient in key molecules of the death receptor pathway. Here, we demonstrate for the first time that Smac mimetic primes apoptosis-resistant, FADD- or caspase-8-deficient leukemia cells for TNFα-induced necroptosis in a synergistic manner. In contrast to TNFα, Smac mimetic significantly enhances CD95-induced apoptosis in wild-type but not in FADD-deficient cells. Interestingly, Smac mimetic- and TNFα-mediated cell death occurs without characteristic features of apoptosis (i.e., caspase activation, DNA fragmentation) in FADD-deficient cells. By comparison, Smac mimetic and TNFα trigger activation of caspase-8, -9, and -3 and DNA fragmentation in wild-type cells. Consistently, the caspase inhibitor zVAD.fmk fails to block Smac mimetic- and TNFα-triggered cell death in FADD- or caspase-8-deficient cells, while it confers protection in wild-type cells. By comparison, necrostatin-1, an RIP1 kinase inhibitor, abolishes Smac mimetic- and TNFα-induced cell death in FADD- or caspase-8-deficient. Thus, Smac mimetic enhances TNFα-induced cell death in leukemia cells via two distinct pathways in a context-dependent manner: it primes apoptosis-resistant cells lacking FADD or caspase-8 to TNFα-induced, RIP1-dependent and caspase-independent necroptosis, whereas it sensitizes apoptosis-proficient cells to TNFα-mediated, caspase-dependent apoptosis. These findings have important implications for the therapeutic exploitation of necroptosis as an alternative cell death program to overcome apoptosis resistance.


Subject(s)
Apoptosis/drug effects , Biomimetic Materials/pharmacology , Caspase 8/metabolism , Fas-Associated Death Domain Protein/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis Regulatory Proteins , Blotting, Western , Caspase 3/metabolism , Caspase 9/metabolism , DNA Fragmentation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Enzyme Activation/drug effects , Fas-Associated Death Domain Protein/deficiency , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Microscopy, Fluorescence , Mitochondrial Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...