Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Proc Natl Acad Sci U S A ; 115(15): 3930-3935, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29581256

ABSTRACT

Necroptosis, a form of regulated necrotic cell death, is governed by RIP1/RIP3-mediated activation of MLKL. However, the signaling process leading to necroptotic death remains to be elucidated. In this study, we found that PUMA, a proapoptotic BH3-only Bcl-2 family member, is transcriptionally activated in an RIP3/MLKL-dependent manner following induction of necroptosis. The induction of PUMA, which is mediated by autocrine TNF-α and enhanced NF-κB activity, contributes to necroptotic death in RIP3-expressing cells with caspases inhibited. On induction, PUMA promotes the cytosolic release of mitochondrial DNA and activation of the DNA sensors DAI/Zbp1 and STING, leading to enhanced RIP3 and MLKL phosphorylation in a positive feedback loop. Furthermore, deletion of PUMA partially rescues necroptosis-mediated developmental defects in FADD-deficient embryos. Collectively, our results reveal a signal amplification mechanism mediated by PUMA and cytosolic DNA sensors that is involved in TNF-driven necroptotic death in vitro and in vivo.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , DNA-Binding Proteins/metabolism , Glycoproteins/metabolism , Membrane Proteins/metabolism , Necrosis/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Cytosol/metabolism , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , Glycoproteins/genetics , Humans , Membrane Proteins/genetics , Mice , Mitochondria/genetics , Mitochondria/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Necrosis/genetics , Necrosis/physiopathology , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Proto-Oncogene Proteins/genetics , RNA-Binding Proteins , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Proteins/genetics
2.
Nature ; 471(7338): 373-6, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21368761

ABSTRACT

FADD is a common adaptor shared by several death receptors for signalling apoptosis through recruitment and activation of caspase 8 (refs 1-3). Death receptors are essential for immune homeostasis, but dispensable during embryogenesis. Surprisingly, Fadd(-/-) mice die in utero and conditional deletion of FADD leads to impaired lymphocyte proliferation. How FADD regulates embryogenesis and lymphocyte responses has been a long-standing enigma. FADD could directly bind to RIP1 (also known as RIPK1), a serine/threonine kinase that mediates both necrosis and NF-κB activation. Here we show that Fadd(-/-) embryos contain raised levels of RIP1 and exhibit massive necrosis. To investigate a potential in vivo functional interaction between RIP1 and FADD, null alleles of RIP1 were crossed into Fadd(-/-) mice. Notably, RIP1 deficiency allowed normal embryogenesis of Fadd(-/-) mice. Conversely, the developmental defect of Rip1(-/-) lymphocytes was partially corrected by FADD deletion. Furthermore, RIP1 deficiency fully restored normal proliferation in Fadd(-/-) T cells but not in Fadd(-/-) B cells. Fadd(-/-)Rip1(-/-) double-knockout T cells are resistant to death induced by Fas or TNF-α and show reduced NF-κB activity. Therefore, our data demonstrate an unexpected cell-type-specific interplay between FADD and RIP1, which is critical for the regulation of apoptosis and necrosis during embryogenesis and lymphocyte function.


Subject(s)
Embryo, Mammalian/metabolism , Fas-Associated Death Domain Protein/metabolism , GTPase-Activating Proteins/metabolism , Genetic Complementation Test , Lymphocytes/cytology , Animals , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Proliferation , Embryo, Mammalian/enzymology , Embryo, Mammalian/pathology , Embryonic Development/genetics , Fas-Associated Death Domain Protein/deficiency , Fas-Associated Death Domain Protein/genetics , Female , GTPase-Activating Proteins/deficiency , GTPase-Activating Proteins/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Male , Mice , Mice, Knockout , Necrosis/genetics
3.
J Biol Chem ; 287(15): 12455-68, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22362782

ABSTRACT

Autophagy and apoptosis are two evolutionarily conserved processes that regulate cell fate in response to cytotoxic stress. However, the functional relationship between these two processes remains far from clear. Here, we demonstrate an autophagy-dependent mechanism of caspase-8 activation and initiation of the apoptotic cascade in response to SKI-I, a pan-sphingosine kinase inhibitor, and bortezomib, a proteasome inhibitor. Autophagy is induced concomitantly with caspase-8 activation, which is responsible for initiation of the caspase cascade and the mitochondrial amplification loop that is required for full execution of apoptosis. Inhibition of autophagosome formation by depletion of Atg5 or Atg3 results in a marked suppression of caspase-8 activation and apoptosis. Although caspase-8 self-association depends on p62/SQSTM1, its self-processing requires the autophagosomal membrane. Caspase-8 forms a complex with Atg5 and colocalizes with LC3 and p62. Moreover, FADD, an adaptor protein for caspase-8 activation, associates with Atg5 on Atg16L- and LC3-positive autophagosomal membranes and loss of FADD suppresses cell death. Taken together, these results indicate that the autophagosomal membrane serves as a platform for an intracellular death-inducing signaling complex (iDISC) that recruits self-associated caspase-8 to initiate the caspase-8/-3 cascade.


Subject(s)
Apoptosis , Autophagy , Caspase 8/metabolism , Cell Membrane/metabolism , Death Domain Receptor Signaling Adaptor Proteins/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy-Related Protein 5 , Autophagy-Related Proteins , Caspase 3/metabolism , Cell Membrane/enzymology , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation , Fas-Associated Death Domain Protein/metabolism , Gene Knockout Techniques , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Hydrazines/pharmacology , Leukemia, Myeloid, Acute , Lysosomal Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Binding , Protein Multimerization , Protein Transport , Pyrazoles/pharmacology , Sequestosome-1 Protein , Tumor Cells, Cultured , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
4.
Apoptosis ; 18(9): 1106-19, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23801080

ABSTRACT

Recent data show that anti-CD20 therapy is effective for some autoimmune diseases, including multiple sclerosis (MS). However, the efficacy of anti-CD20 therapy for MS is largely limited because anti-CD20 antibodies target only B cells. In previous studies, we have investigated the function of MS4a4B, a novel CD20 homologue, in T cell proliferation. Here, we found that MS4a4B regulates not only T cell proliferation but also T cell apoptosis. Knockdown of MS4a4B by MS4a4B-siRNA or MS4a4B-shRNA-expressing vector promoted apoptosis in primary T cells and T32 cell line. In contrast, vector-driven over-expression of MS4a4B reduced apoptosis in EL-4 cells. Machinery analysis showed that MS4a4B-mediated T cell survival was associated with decreased activity of caspases 3, 8 and 9. Interestingly, binding of anti-MS4a4B antibodies to T cells induced activated T cells to undergo apoptosis. To test whether anti-MS4a4B antibody interferes with MS4a4B-mediated protection of T cells, we injected anti-MS4a4B antibodies into mice with experimental autoimmune encephalomyelitis (EAE). The results show that anti-MS4a4B treatment ameliorated the severity of EAE, accompanied by decreased Th1 and Th17 cell responses and reduced levels of pro-inflammatory cytokines in the central nervous system, suggesting that MS4a4B may serve as a target of antibody-based therapy for T cell-mediated diseases.


Subject(s)
Antibodies/therapeutic use , Apoptosis , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Membrane Proteins/genetics , Membrane Proteins/immunology , T-Lymphocytes/cytology , Animals , Cell Proliferation , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Gene Knockout Techniques , Humans , Mice , Mice, Inbred C57BL , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , T-Lymphocytes/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology
5.
Nat Cell Biol ; 8(8): 855-62, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16845383

ABSTRACT

The tumour suppressor p53 induces apoptosis or cell-cycle arrest in response to genotoxic and other stresses. In unstressed cells, the anti-proliferative effects of p53 are restrained by mouse double minute 2 (Mdm2), a ubiquitin ligase (E3) that promotes p53 ubiquitination and degradation. Mdm2 also mediates its own degradation through auto-ubiquitination. It is unclear how the cis- and trans-E3 activities of Mdm2, which have opposing effects on cell fate, are differentially regulated. Here, we show that death domain-associated protein (Daxx) is required for Mdm2 stability. Downregulation of Daxx decreases Mdm2 levels, whereas overexpression of Daxx strongly stabilizes Mdm2. Daxx simultaneously binds to Mdm2 and the deubiquitinase Hausp, and it mediates the stabilizing effect of Hausp on Mdm2. In addition, Daxx enhances the intrinsic E3 activity of Mdm2 towards p53. On DNA damage, Daxx dissociates from Mdm2, which correlates with Mdm2 self-degradation. These findings reveal that Daxx modulates the function of Mdm2 at multiple levels and suggest that the disruption of the Mdm2-Daxx interaction may be important for p53 activation in response to DNA damage.


Subject(s)
Carrier Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-mdm2/metabolism , Animals , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Co-Repressor Proteins , DNA Damage , Endopeptidases/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HCT116 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Microscopy, Fluorescence , Models, Biological , Molecular Chaperones , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Proto-Oncogene Proteins c-mdm2/genetics , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin/metabolism , Ubiquitin Thiolesterase , Ubiquitin-Specific Peptidase 7
6.
J Immunol ; 186(1): 203-13, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21115735

ABSTRACT

Signal transduction mediated by Fas-associated death domain protein (FADD) represents a paradigm of coregulation of apoptosis and cellular proliferation. During apoptotic signaling induced by death receptors including Fas, FADD is required for the recruitment and activation of caspase 8. In addition, a death receptor-independent function of FADD is essential for embryogenesis. In previous studies, FADD deficiency in embryonic stem cells resulted in a complete lack of B cells and dramatically reduced T cell numbers, as shown by Rag1(-/-) blastocyst complementation assays. However, T-specific FADD-deficient mice contained normal numbers of thymocytes and slightly reduced peripheral T cell numbers, whereas B cell-specific deletion of FADD led to increased peripheral B cell numbers. It remains undetermined what impact an FADD deficiency has on hematopoietic stem cells and progenitors. The current study analyzed the effect of simultaneous deletion of FADD in multiple cell types, including bone marrow cells, by using the IFN-inducible Mx1-cre transgene. The resulting FADD mutant mice did not develop lymphoproliferation diseases, unlike Fas-deficient mice. Instead, a time-dependent depletion of peripheral FADD-deficient lymphocytes was observed. In the bone marrow, a lack of FADD led to a dramatic decrease in the hematopoietic stem cells and progenitor-enriched population. Furthermore, FADD-deficient bone marrow cells were defective in their ability to generate lymphoid, myeloid, and erythroid cells. Thus, the results revealed a temporal requirement for FADD. Although dispensable during lymphopoiesis post lineage commitment, FADD plays a critical role in early hematopoietic stages in the bone marrow.


Subject(s)
Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Fas-Associated Death Domain Protein/deficiency , Fas-Associated Death Domain Protein/genetics , Hematopoiesis/genetics , Hematopoiesis/immunology , Lymphocyte Depletion/methods , Animals , Bone Marrow Cells/metabolism , Cell Lineage/genetics , Cell Lineage/immunology , Cells, Cultured , Erythroid Precursor Cells/immunology , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/pathology , GTP-Binding Proteins/genetics , Gene Deletion , Green Fluorescent Proteins/genetics , Integrases/genetics , Lymphoid Progenitor Cells/immunology , Lymphoid Progenitor Cells/metabolism , Lymphoid Progenitor Cells/pathology , Mice , Mice, Knockout , Mice, Transgenic , Myeloid Progenitor Cells/immunology , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Myxovirus Resistance Proteins , Time Factors , fas Receptor/deficiency , fas Receptor/genetics
7.
Am J Respir Crit Care Med ; 183(8): 1043-54, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-20959557

ABSTRACT

RATIONALE: Acute lung injury and the acute respiratory distress syndrome are characterized by increased lung oxidant stress and apoptotic cell death. The contribution of epithelial cell apoptosis to the development of lung injury is unknown. OBJECTIVES: To determine whether oxidant-mediated activation of the intrinsic or extrinsic apoptotic pathway contributes to the development of acute lung injury. METHODS: Exposure of tissue-specific or global knockout mice or cells lacking critical components of the apoptotic pathway to hyperoxia, a well-established mouse model of oxidant-induced lung injury, for measurement of cell death, lung injury, and survival. MEASUREMENTS AND MAIN RESULTS: We found that the overexpression of SOD2 prevents hyperoxia-induced BAX activation and cell death in primary alveolar epithelial cells and prolongs the survival of mice exposed to hyperoxia. The conditional loss of BAX and BAK in the lung epithelium prevented hyperoxia-induced cell death in alveolar epithelial cells, ameliorated hyperoxia-induced lung injury, and prolonged survival in mice. By contrast, Cyclophilin D-deficient mice were not protected from hyperoxia, indicating that opening of the mitochondrial permeability transition pore is dispensable for hyperoxia-induced lung injury. Mice globally deficient in the BH3-only proteins BIM, BID, PUMA, or NOXA, which are proximal upstream regulators of BAX and BAK, were not protected against hyperoxia-induced lung injury suggesting redundancy of these proteins in the activation of BAX or BAK. CONCLUSIONS: Mitochondrial oxidant generation initiates BAX- or BAK-dependent alveolar epithelial cell death, which contributes to hyperoxia-induced lung injury.


Subject(s)
Acute Lung Injury/etiology , Respiratory Mucosa/physiopathology , Animals , Apoptosis/physiology , Peptidyl-Prolyl Isomerase F , Cyclophilins/physiology , Disease Models, Animal , Hyperoxia/complications , Hyperoxia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Oxidative Stress/physiology , Pulmonary Alveoli/chemistry , Pulmonary Alveoli/cytology , Pulmonary Alveoli/physiopathology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/analysis , Respiratory Mucosa/metabolism , Superoxide Dismutase/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
8.
J Immunol ; 182(1): 207-15, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19109151

ABSTRACT

Fas/Apo-1 signals through the FADD (Fas-associated death domain) adaptor protein, which recruits and activates the apical caspase 8 and leads to apoptosis. Cellular FLIP (cFLIP) is a homolog of caspase 8 and is also capable of binding to FADD. Previous studies suggest that cFLIP could either enhance or inhibit apoptosis and lead to NF-kappaB and Erk1/2 activation. Like FADD or caspase 8 deficiency, a lack of cFLIP disrupts embryogenesis and T cell proliferation. It has been demonstrated that B cells lacking either FADD or caspase 8 were defective in both Fas-induced apoptosis and TLR-induced proliferation, which indicates that these death-inducing proteins have an additional role in regulating innate immunity. To analyze the function of cFLIP in B cells, conditional deletion of cFLIP was induced by using CD19(Cre). The resulting B cell-specific cFLIP-deficient mice were found to have reduced numbers of peripheral B cells that were hypersensitive to Fas-induced apoptosis and impaired in proliferation induced by TLRs and the BCR. Furthermore, there was aberrant expression of costimulatory proteins and activation markers in cFLIP-deficient B cells. Whereas LPS-induced activation of NF-kappaB and Erk1/2 appears to be unaffected, p38 and Jnk were spontaneously activated and hyperinduced in cFLIP-deficient B cells. Therefore, these data revealed novel functions of cFLIP in B cells.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/enzymology , CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Cell Proliferation , Mitogen-Activated Protein Kinase 9/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis/immunology , B-Lymphocytes/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Bone Marrow Cells/immunology , CASP8 and FADD-Like Apoptosis Regulating Protein/deficiency , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/immunology , Mice , Mice, Knockout , Mice, Transgenic
9.
Int Immunol ; 21(6): 691-703, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19395373

ABSTRACT

The mammalian Nit1 protein is homologous to plant and bacterial nitrilases. In flies and worms, Nit1 is fused to the 5' end of Fhit, suggesting that Nit1 may functionally interact with the Fhit pathway. Fhit has been shown to play a role of a tumor suppressor. Somatic loss of Fhit in human tissues is associated with a wide variety of cancers. Deletion of Fhit results in a predisposition to induced and spontaneous tumors in mice. It has been suggested that Nit1 collaborates with Fhit in tumor suppression. Similar to mice lacking Fhit, Nit1-deficient mice are more sensitive to carcinogen-induced tumors. It was previously shown that ectopic expression of Nit1 or Fhit led to caspase activation and apoptosis, and that both proteins may play a role in DNA damage-induced apoptosis. In this study, we analyzed the physiological function of Nit1 in T cells using Nit1-knockout mice. Nit1-deficient T cells can undergo apoptosis induced by DNA damage due to irradiation and chemical treatment. However, apoptosis induced by Fas or Ca(++) signals appeared to be compromised. Additionally, Nit1 deficiency resulted in T cell hyperproliferative responses induced by TCR stimulation. The expressions of T cell activation markers were elevated in Nit1(-/-) T cells. There was a spontaneous cell cycle entry and enhanced cell cycle progression in Nit1(-/-) T cells. These data indicate that Nit1 is a novel negative regulator in primary T cells.


Subject(s)
Aminohydrolases/immunology , Cell Proliferation , Feedback, Physiological , T-Lymphocytes/metabolism , Aminohydrolases/genetics , Aminohydrolases/metabolism , Animals , Apoptosis/genetics , Apoptosis/immunology , Calcium/immunology , Calcium/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Cell Cycle Proteins/metabolism , DNA Damage/immunology , Lymphocyte Activation , Mice , Mice, Knockout , Sequence Homology, Amino Acid , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Thymus Gland/pathology , fas Receptor/immunology , fas Receptor/metabolism
10.
Cell Death Dis ; 10(3): 245, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30867408

ABSTRACT

RIPK1 has emerged as a key effector in programmed necrosis or necroptosis. This function of RIPK1 is mediated by its protein serine/threonine kinase activity and through the downstream kinase RIPK3. Deletion of RIPK1 prevents embryonic lethality in mice lacking FADD, a signaling adaptor protein required for activation of Caspase 8 in extrinsic apoptotic pathways. This indicates that FADD-mediated apoptosis inhibits RIPK1-dependent necroptosis to ensure successful embryogenesis. However, the molecular mechanism for this critical regulation remains unclear. In the current study, a novel mouse model has been generated, by disrupting a potential caspase cleavage site at aspartic residue (D)324 in RIPK1. Interestingly, replacing D324 with alanine (A) in RIPK1 results in midgestation lethality, similar to the embryonic defect in FADD-/- mice but in stark contrast to the normal embryogenesis of RIPK1-/- null mutant mice. Surprisingly, disrupting the downstream RIPK3 alone is insufficient to rescue RIPK1D324A/D324A mice from embryonic lethality, unless FADD is deleted simultaneously. Further analyses reveal a paradoxical role for RIPK1 in promoting caspase activation and apoptosis in embryos, a novel mechanism previously unappreciated.


Subject(s)
Apoptosis/genetics , Embryonic Development/genetics , Necroptosis/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis/drug effects , Caspase 8/metabolism , Fas-Associated Death Domain Protein/genetics , Fibroblasts , Genes, Lethal , Lymphadenopathy/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Necroptosis/drug effects , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Splenomegaly/genetics , T-Lymphocytes , Tumor Necrosis Factor-alpha/pharmacology
11.
Nat Commun ; 10(1): 705, 2019 02 11.
Article in English | MEDLINE | ID: mdl-30741936

ABSTRACT

TRADD is an adaptor for TNFR1-induced apoptosis and NFκB activation. However, TRADD-deficient mice undergo normal development and contain normal lymphoid populations, which contrasts with an embryonic defect in mice lacking FADD, the shared adaptor mediating apoptosis. Recent studies indicate FADD suppresses embryonic necroptosis mediated by RIPK1. TRADD was suggested to also mediate necroptosis. Here we report that targeting TRADD fails to rescue Fadd-/- embryos from necroptosis, and ablation of TRADD rescues Ripk1-/- mice from perinatal lethality when RIPK3-mediated necroptosis is disabled. The resulting Ripk1-/-Ripk3-/-Tradd-/- mice survive until early adulthood, but die thereafter. A single allele of Tradd is optimal for survival of Ripk1-/-Ripk3-/-Tradd+/- mice. We show that TRADD plays a more dominating role in NFκB-signaling than RIPK1. While RIPK1 protects thymocytes from TNFα-induced apoptosis, TRADD promotes this process. The data demonstrate that TRADD is critical in perinatal and adult mice lacking RIPK1 and RIPK3, which has not been appreciated in prior studies.


Subject(s)
Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Death Domain Protein/metabolism , Animals , Apoptosis/drug effects , Caspase 8/genetics , Caspase 8/metabolism , Cell Death , Cell Proliferation/drug effects , Fas-Associated Death Domain Protein/metabolism , Fibroblasts , Gene Deletion , Gene Expression Regulation , Intestines/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/pharmacology , Signal Transduction , Survival Analysis , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , TNF Receptor-Associated Death Domain Protein/genetics , TNF Receptor-Associated Death Domain Protein/pharmacology , Thymocytes/drug effects , Transcriptome , Tumor Necrosis Factor-alpha
12.
Nat Commun ; 10(1): 3759, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31434890

ABSTRACT

Autophagy cargo recognition and clearance are essential for intracellular protein quality control. SQSTM1/p62 sequesters intracellular aberrant proteins and mediates cargo delivery for their selective autophagic degradation. The formation of p62 non-membrane-bound liquid compartments is critical for its function as a cargo receptor. The regulation of p62 phase separation/condensation has yet been poorly characterised. Using an unbiased yeast two-hybrid screening and complementary approaches, we found that DAXX physically interacts with p62. Cytoplasmic DAXX promotes p62 puncta formation. We further elucidate that DAXX drives p62 liquid phase condensation by inducing p62 oligomerisation. This effect promotes p62 recruitment of Keap1 and subsequent Nrf2-mediated stress response. The present study suggests a mechanism of p62 phase condensation by a protein interaction, and indicates that DAXX regulates redox homoeostasis, providing a mechanistic insight into the prosurvival function of DAXX.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoplasm/metabolism , NF-E2-Related Factor 2/metabolism , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Sequestosome-1 Protein/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Autophagy/physiology , Cell Line , Co-Repressor Proteins , Drosophila , Female , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Male , Mice , Molecular Chaperones , Nuclear Proteins/genetics , Protein Binding , Protein Folding , Protein Interaction Domains and Motifs
13.
J Inequal Appl ; 2018(1): 44, 2018.
Article in English | MEDLINE | ID: mdl-29497264

ABSTRACT

The purpose of this paper is to solve fractional calculus of variational Herglotz problem depending on an Atangana-Baleanu fractional derivative. Since the new Atangana-Baleanu fractional derivative is non-singular and non-local, the Euler-Lagrange equations are proposed for the problems of Herglotz. Fractional variational Herglotz problems of variable order are considered and two cases are shown. The Noether-type theorem with this new fractional derivative is proved. Several typical examples of the results of this paper are expressed in this paper.

14.
Mol Cell Biol ; 24(13): 5914-22, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15199146

ABSTRACT

Oxidative stress and reactive oxygen species (ROS) can elicit and modulate various physiological and pathological processes, including cell death. However, the mechanisms controlling ROS-induced cell death are largely unknown. Data from this study suggest that receptor-interacting protein (RIP) and tumor necrosis factor receptor (TNFR)-associated factor 2 (TRAF2), two key effector molecules of TNF signaling, are essential for ROS-induced cell death. We found that RIP(-/-) or TRAF2(-/-) mouse embryonic fibroblasts (MEF) are resistant to ROS-induced cell death when compared to wild-type cells, and reconstitution of RIP and TRAF2 gene expression in their respective deficient MEF cells restored their sensitivity to H(2)O(2)-induced cell death. We also found that RIP and TRAF2 form a complex upon H(2)O(2) exposure, but without the participation of TNFR1. The colocalization of RIP with a membrane lipid raft marker revealed a possible role of lipid rafts in the transduction of cell death signal initiated by H(2)O(2). Finally, our results demonstrate that activation of c-Jun NH(2)-terminal kinase 1 is a critical event downstream of RIP and TRAF2 in mediating ROS-induced cell death. Therefore, our study uncovers a novel signaling pathway regulating oxidative stress-induced cell death.


Subject(s)
Oxidative Stress , Proteins/physiology , Animals , Cell Death , Cell Line , Humans , Hydrogen Peroxide/pharmacology , Jurkat Cells , Mice , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinases/metabolism , Protein Binding/drug effects , Proteins/genetics , Proteins/metabolism , Reactive Oxygen Species/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases , Signal Transduction , TNF Receptor-Associated Factor 2 , Transfection
15.
PLoS One ; 12(3): e0174011, 2017.
Article in English | MEDLINE | ID: mdl-28301594

ABSTRACT

Daxx was originally isolated as a Fas-binding protein. However, the in vivo function of Daxx in Fas-induced apoptosis has remained enigmatic. Fas plays an important role in homeostasis in the immune system. Fas gene mutations lead to autoimmune-lymphoproliferation (lpr) diseases characterized by hyperplasia of secondary lymphoid organs. It is well established that the FADD adaptor binds to Fas, and recruits/activates caspase 8. However, additional proteins including Daxx have also been indicated to associate with Fas. It was proposed that Daxx mediates a parallel apoptotic pathway that is independent of FADD and caspase 8, but signals through ASK1-mediated apoptotic pathway. However, because the deletion of Daxx leads to embryonic lethality, the in vivo function of Daxx has not been properly analyzed. In the current study, analysis was performed using a conditional mutant mouse in which Daxx was deleted specifically in T cells. The data show that Daxx-/- T cells were able to undergo normal Fas-induced apoptosis. While containing normal thymocyte populations, the T cell-specific Daxx-/- mice have a reduced peripheral T cell pool. Importantly, Daxx-deficient T cells displayed increased death responses upon activation through TCR stimulation. These results unequivocally demonstrated that Daxx does not mediate Fas-induced apoptosis, but rather that it plays a critical role in survival responses in primary mature T cells.


Subject(s)
Apoptosis/physiology , Carrier Proteins/physiology , Cell Survival/physiology , Intracellular Signaling Peptides and Proteins/physiology , Nuclear Proteins/physiology , T-Lymphocytes/cytology , fas Receptor/metabolism , Animals , Carrier Proteins/metabolism , Cell Proliferation , Co-Repressor Proteins , Flow Cytometry , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Molecular Chaperones , Nuclear Proteins/metabolism
16.
Cell Death Dis ; 7(9): e2379, 2016 09 29.
Article in English | MEDLINE | ID: mdl-27685623

ABSTRACT

The death receptor, Fas, triggers apoptotic death and is essential for maintaining homeostasis in the peripheral lymphoid organs. RIP1 was originally cloned when searching for Fas-binding proteins and was later shown to associate also with the signaling complex of TNFR1. Although Fas exclusively induces apoptosis, TNFR1 primarily activates the pro-survival/pro-inflammatory NF-κB pathway. Mutations in Fas lead to lymphoproliferative (lpr) diseases, and deletion of TNFR1 results in defective innate immune responses. However, the function of RIP1 in the adult lymphoid system has not been well understood, primarily owing to perinatal lethality in mice lacking the entire RIP1 protein in germ cells. This current study investigated the requirement for RIP1 in the T lineage using viable RIP1 mutant mice containing a conditional and kinase-dead RIP1 allele. Disabling the kinase activity of RIP1 had no obvious impact on the T-cell compartment. However, T-cell-specific deletion of RIP1 led to a severe T-lymphopenic condition, owing to a dramatically reduced mature T-cell pool in the periphery. Interestingly, the immature T-cell compartment in the thymus appeared intact. Further analysis showed that mature RIP1-/- T cells were severely defective in antigen receptor-induced proliferative responses. Moreover, the RIP1-/- T cells displayed greatly increased death and contained elevated caspase activities, an indication of apoptosis. In total, these results revealed a novel, kinase-independent function of RIP1, which is essential for not only promoting TCR-induced proliferative responses but also in blocking apoptosis in mature T cells.


Subject(s)
GTPase-Activating Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Deletion , Lymphocyte Activation/immunology , Mice , Receptors, Antigen, T-Cell/metabolism , Receptors, Death Domain/metabolism , T-Lymphocytes/drug effects , Thymus Gland/cytology , Tumor Necrosis Factor-alpha/pharmacology
17.
Cell Rep ; 16(12): 3247-3259, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27498868

ABSTRACT

MLKL, a key component downstream of RIPK3, is suggested to be a terminal executor of necroptosis. Genetic studies have revealed that Ripk3 ablation rescues embryonic lethality in Fadd- or Caspase-8-deficient mice. Given that RIPK3 has also been implicated in non-necroptotic pathways including apoptosis and inflammatory signaling, it remains unclear whether the lethality in Fadd(-/-) mice is indeed caused by necropotosis. Here, we show that genetic deletion of Mlkl rescues the developmental defect in Fadd-deficient mice and that Fadd(-/-)Mlkl(-/-) mice are viable and fertile. Mlkl(-/-)Fadd(-/-) mice display significantly accelerated lymphoproliferative disease characterized by lymphadenopathy and splenomegaly when compared to Ripk3(-/-)Fadd(-/-) mice. Mlkl(-/-)Fadd(-/-) bone-marrow-derived macrophages and dendritic cells have impaired NLRP3 inflammasome activation associated with defects in ASC speck formation and NF-κB-dependent NLRP3 transcription. Our findings reveal that MLKL and FADD play critical roles in preventing lymphoproliferative disease and activating the NLRP3 inflammasome.


Subject(s)
Fas-Associated Death Domain Protein/immunology , Inflammasomes/immunology , Lymphoproliferative Disorders/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Protein Kinases/immunology , Animals , Fas-Associated Death Domain Protein/genetics , Lymphoproliferative Disorders/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Kinases/genetics
18.
Cell Rep ; 15(11): 2449-61, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27264187

ABSTRACT

Tumor necrosis factor (TNF) induces necroptosis, a RIPK3/MLKL-dependent form of inflammatory cell death. In response to infection by Gram-negative bacteria, multiple receptors on macrophages, including TLR4, TNF, and type I IFN receptors, are concurrently activated, but it is unclear how they crosstalk to regulate necroptosis. We report that TLR4 activates CASPASE-8 to cleave and remove the deubiquitinase cylindromatosis (CYLD) in a TRIF- and RIPK1-dependent manner to disable necroptosis in macrophages. Inhibiting CASPASE-8 leads to CYLD-dependent necroptosis caused by the TNF produced in response to TLR4 ligation. While lipopolysaccharides (LPS)-induced necroptosis was abrogated in Tnf(-/-) macrophages, a soluble TNF antagonist was not able to do so in Tnf(+/+) macrophages, indicating that necroptosis occurs in a cell-autonomous manner. Surprisingly, TNF-mediated auto-necroptosis of macrophages requires type I IFN, which primes the expression of key necroptosis-signaling molecules, including TNFR2 and MLKL. Thus, the TNF necroptosis pathway is regulated by both negative and positive crosstalk.


Subject(s)
Apoptosis , Cysteine Endopeptidases/metabolism , Cytoprotection , Interferon Type I/metabolism , Macrophages/metabolism , Proteolysis , Tumor Necrosis Factor-alpha/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Caspase 8/metabolism , Deubiquitinating Enzyme CYLD , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Necrosis , Protein Kinases/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism
19.
Front Cell Dev Biol ; 3: 12, 2015.
Article in English | MEDLINE | ID: mdl-25767797

ABSTRACT

RIP1 is an adaptor kinase originally identified as being able to associate with TNFR1 and Fas, and is later shown to be involved in signaling induced by TLRs. Major signaling pathways regulated by RIP1 include necroptosis, apoptosis, and pro-survival/inflammation NF-κB activation. Previous studies show that RIP1 deficiency has no effect on mouse embryogenesis, but blocks postnatal development. This phenotype could not readily be explained, since mice lacking TNFR1, Fas, or TLRs show no apparent developmental defect. Certain types of RIP1-deficient cells are hypersensitive to TNF-induced apoptosis. However, in our previous study, deletion of the apoptotic adaptor protein, FADD, provides marginal improvement of postnatal development of rip1 (-/-) mice. Remarkably, the current data shows that haploid insufficiency of RIP3, a known mediator of necroptosis, allowed survival of rip1 (-/-) fadd (-/-) mice beyond weaning age, although the resulting rip1(-/-)fadd(-/-) rip3(+/-) mice were significant smaller in size and weight. Moreover, complete absence of RIP3 further improved postnatal development of the resulting rip1 (-/-) fadd (-/-) rip3 (-/-) mice, which display normal size and weight. In such triple knockout (TKO) mice, lymphocytes underwent normal development, but progressively accumulated as mice age. This lymphoproliferative (lpr) disease in TKO mice is, however, less severe than that of fadd(-/-)rip3 (-/-) double knockout mice. In total, the data show that the postnatal developmental defect in rip1 (-/-) mice is due in part to FADD-mediated apoptosis as well as RIP3-dependent necroptosis. Moreover, the function of RIP1 contributes to development of lpr diseases.

20.
Sci Signal ; 8(361): ra9, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25628462

ABSTRACT

Genomic amplification of the gene encoding and phosphorylation of the protein FADD (Fas-associated death domain) is associated with poor clinical outcome in lung cancer and in head and neck cancer. Activating mutations in the guanosine triphosphatase RAS promotes cell proliferation in various cancers. Increased abundance of phosphorylated FADD in patient-derived tumor samples predicts poor clinical outcome. Using immunohistochemistry analysis and in vivo imaging of conditional mouse models of KRAS(G12D)-driven lung cancer, we found that the deletion of the gene encoding FADD suppressed tumor growth, reduced the proliferative index of cells, and decreased the activation of downstream effectors of the RAS-MAPK (mitogen-activated protein kinase) pathway that promote the cell cycle, including retinoblastoma (RB) and cyclin D1. In mouse embryonic fibroblasts, the induction of mitosis upon activation of KRAS required FADD and the phosphorylation of FADD by CK1α (casein kinase 1α). Deleting the gene encoding CK1α in KRAS mutant mice abrogated the phosphorylation of FADD and suppressed lung cancer development. Phosphorylated FADD was most abundant during the G2/M phase of the cell cycle, and mass spectrometry revealed that phosphorylated FADD interacted with kinases that mediate the G2/M transition, including PLK1 (Polo-like kinase 1), AURKA (Aurora kinase A), and BUB1 (budding uninhibited by benzimidazoles 1). This interaction was decreased in cells treated with a CKI-7, a CK1α inhibitor. Therefore, as the kinase that phosphorylates FADD downstream of RAS, CK1α may be a therapeutic target for KRAS-driven lung cancer.


Subject(s)
Casein Kinase Ialpha/metabolism , Fas-Associated Death Domain Protein/metabolism , Lung Neoplasms/genetics , Mutation, Missense/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Aurora Kinase A/metabolism , Blotting, Western , Cell Cycle Proteins/metabolism , DNA Primers/genetics , Genotype , Histological Techniques , Immunoprecipitation , Luminescent Measurements , Mass Spectrometry , Mice , Phosphorylation , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , X-Ray Microtomography , Polo-Like Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL