Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
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
4.
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.

5.
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
6.
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
7.
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
8.
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
9.
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
10.
Nat Commun ; 6: 7515, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26104484

ABSTRACT

TLR2 promotes NLRP3 inflammasome activation via an early MyD88-IRAK1-dependent pathway that provides a priming signal (signal 1) necessary for activation of the inflammasome by a second potassium-depleting signal (signal 2). Here we show that TLR3 binding to dsRNA promotes post-translational inflammasome activation through intermediate and late TRIF/RIPK1/FADD-dependent pathways. Both pathways require the scaffolding but not the catalytic function of caspase-8 or RIPK1. Only the late pathway requires kinase competent RIPK3 and MLKL function. Mechanistically, FADD/caspase-8 scaffolding function provides a post-translational signal 1 in the intermediate pathway, whereas in the late pathway it helps the oligomerization of RIPK3, which together with MLKL provides both signal 1 and 2 for inflammasome assembly. Cytoplasmic dsRNA activates NLRP3 independent of TRIF, RIPK1, RIPK3 or mitochondrial DRP1, but requires FADD/caspase-8 in wildtype macrophages to remove RIPK3 inhibition. Our study provides a comprehensive analysis of pathways that lead to NLRP3 inflammasome activation in response to dsRNA.


Subject(s)
Carrier Proteins/metabolism , Caspase 8/metabolism , Macrophages/metabolism , Protein Kinases/metabolism , RNA, Double-Stranded/metabolism , Toll-Like Receptor 3/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Carrier Proteins/genetics , Caspase 8/genetics , Dynamins/genetics , Dynamins/metabolism , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
11.
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.

12.
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
13.
J Exp Med ; 211(6): 1093-108, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24842373

ABSTRACT

Leukemic stem cells (LSCs) isolated from acute myeloid leukemia (AML) patients are more sensitive to nuclear factor κB (NF-κB) inhibition-induced cell death when compared with hematopoietic stem and progenitor cells (HSPCs) in in vitro culture. However, inadequate anti-leukemic activity of NF-κB inhibition in vivo suggests the presence of additional survival/proliferative signals that can compensate for NF-κB inhibition. AML subtypes M3, M4, and M5 cells produce endogenous tumor necrosis factor α (TNF). Although stimulating HSPC with TNF promotes necroptosis and apoptosis, similar treatment with AML cells (leukemic cells, LCs) results in an increase in survival and proliferation. We determined that TNF stimulation drives the JNK-AP1 pathway in a manner parallel to NF-κB, leading to the up-regulation of anti-apoptotic genes in LC. We found that we can significantly sensitize LC to NF-κB inhibitor treatment by blocking the TNF-JNK-AP1 signaling pathway. Our data suggest that co-inhibition of both TNF-JNK-AP1 and NF-κB signals may provide a more comprehensive treatment paradigm for AML patients with TNF-expressing LC.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Leukemia, Myeloid, Acute/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Anthracenes/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Gene Expression Regulation, Leukemic/drug effects , HL-60 Cells , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , K562 Cells , Leukemia, Monocytic, Acute/genetics , Leukemia, Monocytic, Acute/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Myelomonocytic, Acute/genetics , Leukemia, Myelomonocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/metabolism , Mice , Mice, Knockout , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Nitriles/pharmacology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfones/pharmacology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
15.
Nat Commun ; 5: 3351, 2014.
Article in English | MEDLINE | ID: mdl-24548998

ABSTRACT

Adenylate kinase 2 (AK2), which balances adenine nucleotide pool, is a multi-functional protein. Here we show that AK2 negatively regulates tumour cell growth. AK2 forms a complex with dual-specificity phosphatase 26 (DUSP26) phosphatase and stimulates DUSP26 activity independently of its AK activity. AK2/DUSP26 phosphatase protein complex dephosphorylates fas-associated protein with death domain (FADD) and regulates cell growth. AK2 deficiency enhances cell proliferation and induces tumour formation in a xenograft assay. This anti-growth function of AK2 is associated with its DUSP26-stimulating activity. Downregulation of AK2 is frequently found in tumour cells and human cancer tissues showing high levels of phospho-FADD(Ser194). Moreover, reconstitution of AK2 in AK2-deficient tumour cells retards both cell proliferation and tumourigenesis. Consistent with this, AK2(+/-) mouse embryo fibroblasts exhibit enhanced cell proliferation with a significant alteration in phospho-FADD(Ser191). These results suggest that AK2 is an associated activator of DUSP26 and suppresses cell proliferation by FADD dephosphorylation, postulating AK2 as a negative regulator of tumour growth.


Subject(s)
Adenylate Kinase/metabolism , Dual-Specificity Phosphatases/metabolism , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Adenylate Kinase/genetics , Animals , Cell Line , Cell Proliferation/genetics , Cell Proliferation/physiology , Dual-Specificity Phosphatases/genetics , Electrophoresis, Gel, Two-Dimensional , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , HeLa Cells , Humans , In Vitro Techniques , MCF-7 Cells , Male , Mice , Mice, Nude , Mitogen-Activated Protein Kinase Phosphatases/genetics , Phosphorylation , Tandem Mass Spectrometry , Xenograft Model Antitumor Assays
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.
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
18.
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
19.
Immunol Res ; 51(2-3): 227-36, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22038529

ABSTRACT

RIP1 is an adaptor serine/threonine kinase associated with the signaling complex of death receptors (DRs) including Fas, TNFR1, and TRAIL-Rs which can initiate apoptosis. While DRs are dispensable throughout development, RIP1 deletion results in perinatal lethality. The developmental defect caused by absence of RIP1 remains unexplained. In previous studies, RIP1-deficient hematopoietic progenitors failed to reconstitute the T cell compartment and our recent data indicate a new role for RIP1 in TCR-induced activation of the pro-survival NF-κB pathway. Here, we show that RIP1 is also critical for B cell development. In addition, RIP1(-/-) B cells stimulated through LPS/TLR4 are impaired in NF-κB activation but have no major defect in the Akt pathway. Recently, RIP1 has also emerged as a critical player in necrosis-like death, necroptosis, in various cell lines. We have demonstrated that RIP1 deficiency can reverse the embryonic and T cell proliferation defects in mice lacking FADD, a caspase adaptor protein, which indicates a potential role for RIP1 in mediating in vivo necroptosis. We provide an overview and discussion of the accumulating data revealing insights into the diverse functions of RIP1 in survival and death signaling in lymphocytes.


Subject(s)
Fas-Associated Death Domain Protein/immunology , GTPase-Activating Proteins/immunology , Lymphocytes/immunology , Necrosis/immunology , Animals , Cell Survival/genetics , Fas-Associated Death Domain Protein/genetics , GTPase-Activating Proteins/genetics , Humans , Mice , Mice, Knockout , NF-kappa B/immunology , Necrosis/genetics , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction/immunology
20.
PLoS One ; 6(8): e23209, 2011.
Article in English | MEDLINE | ID: mdl-21853090

ABSTRACT

BACKGROUND: Programmed necrosis/necroptosis is an emerging form of cell death that plays important roles in mammalian development and the immune system. The pro-necrotic kinases in the receptor interacting protein (RIP) family are crucial mediators of programmed necrosis. Recent advances in necrosis research have been greatly aided by the identification of chemical inhibitors that block programmed necrosis. Necrostatin-1 (Nec-1) and its derivatives were previously shown to target the pro-necrotic kinase RIP1/RIPK1. The protective effect conferred by Nec-1 and its derivatives in many experimental model systems was often attributed to the inhibition of RIP1 function. METHODOLOGY/PRINCIPAL FINDINGS: We compared the effect of Nec-1 and siRNA-mediated silencing of RIP1 in the murine fibrosarcoma cell line L929. Treatment of L929 cells with the pan-caspase inhibitor zVAD-fmk or exogenous TNF induces necrosis. Strikingly, we found that siRNA-mediated silencing of RIP1 inhibited zVAD-fmk induced necrosis, but not TNF-induced necrosis. TNF-induced cell death in RIP1 knocked down L929 cells was inhibited by Nec-1, but not the caspase inhibitor zVAD-fmk. We found that PKA-C§ expression, but not Jnk or Erk activation, was moderately inhibited by Nec-1. Moreover, we found that Nec-1 inhibits proximal T cell receptor signaling independent of RIP1, leading to inhibition of T cell proliferation. CONCLUSIONS/SIGNIFICANCE: Our results reveal that besides RIP1, Nec-1 also targets other factors crucial for necrosis induction in L929 cells. In addition, high doses of Nec-1 inhibit other signal transduction pathways such as that for T cell receptor activation. These results highlight the importance to independently validate results obtained using Nec-1 with other approaches such as siRNA-mediated gene silencing. We propose that some of the previous published results obtained using Nec-1 should be re-evaluated in light of our findings.


Subject(s)
Imidazoles/pharmacology , Indoles/pharmacology , Lymphocyte Activation/drug effects , Necrosis/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Animals , Apoptosis/drug effects , Caspases/metabolism , Cell Proliferation/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Jurkat Cells , Lymphocyte Activation/immunology , Mice , NIH 3T3 Cells , Phosphorylation/drug effects , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...