Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
2.
Cell Death Differ ; 22(4): 643-53, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25301068

ABSTRACT

The death receptor Fas undergoes a variety of post-translational modifications including S-palmitoylation. This protein acylation has been reported essential for an optimal cell death signaling by allowing both a proper Fas localization in cholesterol and sphingolipid-enriched membrane nanodomains, as well as Fas high-molecular weight complexes. In human, S-palmitoylation is controlled by 23 members of the DHHC family through their palmitoyl acyltransferase activity. In order to better understand the role of this post-translational modification in the regulation of the Fas-mediated apoptosis pathway, we performed a screen that allowed the identification of DHHC7 as a Fas-palmitoylating enzyme. Indeed, modifying DHHC7 expression by specific silencing or overexpression, respectively, reduces or enhances Fas palmitoylation and DHHC7 co-immunoprecipitates with Fas. At a functional level, DHHC7-mediated palmitoylation of Fas allows a proper Fas expression level by preventing its degradation through the lysosomes. Indeed, the decrease of Fas expression obtained upon loss of Fas palmitoylation can be restored by inhibiting the lysosomal degradation pathway. We describe the modification of Fas by palmitoylation as a novel mechanism for the regulation of Fas expression through its ability to circumvent its degradation by lysosomal proteolysis.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , fas Receptor/metabolism , Acetyltransferases , Amino Acid Substitution , Apoptosis , Cell Line, Tumor , Cycloheximide/pharmacology , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Lipoylation , Lysosomes/metabolism , Protein Processing, Post-Translational , Protein Stability/drug effects , Protein Synthesis Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , fas Receptor/genetics
3.
Leuk Res ; 38(6): 662-5, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24553365

ABSTRACT

In chronic myeloid leukemia (CML), evidence is supporting the role of VEGF in growth, and survival of leukemia cells. The evaluation of plasma VEGF levels in 403 CML patients randomized within SPIRIT study to received imatinib-400mg versus imatinib+cytarabine versus imatinib+interferon (IFN) versus imatinib-600mg demonstrated that VEGF is an independent factor of BCR-ABL burden. VEGF low levels at diagnosis were associated with a progression-free survival of 100% at 48 months. Under treatment, significant lowest levels were observed in imatinib+IFN arm. These results support the use of VEGF as a parameter to predict CML evolution and let us to speculate about antiangiogenic properties of IFN.


Subject(s)
Benzamides/administration & dosage , Interferons/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Adult , Aged , Aged, 80 and over , Female , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Male , Middle Aged , Vascular Endothelial Growth Factor A/blood
4.
Cell Death Dis ; 1: e88, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-21368861

ABSTRACT

Fas ligand (FasL) is a transmembrane protein that regulates cell death in Fas-bearing cells. FasL-mediated cell death is essential for immune system homeostasis and the elimination of viral or transformed cells. Because of its potent cytotoxic activity, FasL expression at the cell surface is tightly regulated, for example, via processing by ADAM10 and SPPL2a generating soluble FasL and the intracellular fragments APL (ADAM10-processed FasL form) and SPA (SPPL2a-processed APL). In this study, we report that FasL processing by ADAM10 counteracts Fas-mediated cell death and is strictly regulated by membrane localization, interactions and modifications of FasL. According to our observations, FasL processing occurs preferentially within cholesterol and sphingolipid-rich nanodomains (rafts) where efficient Fas-FasL contact occurs, Fas receptor and FasL interaction is also required for efficient FasL processing, and FasL palmitoylation, which occurs within its transmembrane domain, is critical for efficient FasL-mediated killing and FasL processing.


Subject(s)
Apoptosis , Fas Ligand Protein/metabolism , ADAM Proteins/metabolism , ADAM10 Protein , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Cell Membrane/metabolism , Humans , Lipoylation , Membrane Proteins/metabolism , Protein Binding
5.
Cell Death Differ ; 15(12): 1824-37, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18670435

ABSTRACT

Selective compartmentalization and internalization have been shown as a means for regulating specific signals of cell surface receptors to correspond to cellular requirements and conditions. Here, we present a conserved extracellular glycosphingolipid-binding motif of Fas as one of the regulatory elements in the selection of its internalization route and consequently the signals transmitted upon ligand binding. This motif is required for clathrin-mediated internalization of Fas, which allows the transduction of its cell death signal. The loss of function of the motif drives the activated receptor to an alternative internalization route that is independent of clathrin and cholesterol-dependent rafts but dependent on ezrin, and thereby extinguishing its cell death signal while promoting its non-death functions. Through biochemical, biophysical, and genetic approaches, we present a protein/lipid-based mechanism as a key to the versatility of the signal transduction by the multifunctional Fas receptor-ligand system.


Subject(s)
Endocytosis , Extracellular Space/metabolism , Glycosphingolipids/metabolism , Signal Transduction , fas Receptor/chemistry , fas Receptor/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Animals , COS Cells , Cell Death , Cell Membrane/metabolism , Chlorocebus aethiops , Cholesterol/metabolism , Clathrin/metabolism , Cytoskeletal Proteins/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Humans , Ligands , Membrane Microdomains/metabolism , Mice , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship
6.
Cell Death Differ ; 15(3): 494-503, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18064043

ABSTRACT

The actin cytoskeleton association is required for caspase 8-independent Fas/CD95 receptor internalization, a critical step for an optimal death-inducing signaling complex formation along the endocytic pathway, leading to efficient activation of the caspase cascade and, ultimately, cell death. However, the way in which this initiation phase of Fas receptor signaling is regulated is still unknown. We report herein that, in B cells, upon Fas engagement, the tyrosine phosphatase SHP-1-regulated Vav dephosphorylation, by downmodulating the Fas-ezrin-actin linkage is a fine-tune switch-off mechanism that the cell uses as a way to terminate the receptor internalization, controlling therefore the time and extent of the DISC formation and cell death.


Subject(s)
Apoptosis , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proto-Oncogene Proteins c-vav/metabolism , fas Receptor/metabolism , Actin Cytoskeleton/metabolism , Animals , B-Lymphocytes/enzymology , B-Lymphocytes/metabolism , Cells, Cultured , Cytoskeletal Proteins/metabolism , Mice , Phosphorylation , Signal Transduction
7.
Cell Death Differ ; 14(9): 1678-87, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17557115

ABSTRACT

Fas ligand (FasL) is a type II transmembrane protein belonging to the tumor necrosis factor family. Its binding to the cognate Fas receptor triggers the apoptosis that plays a pivotal role in the maintenance of immune system homeostasis. The cell death-inducing property of FasL has been associated with its extracellular domain, which can be cleaved off by metalloprotease activity to produce soluble FasL. The fate of the remaining membrane-anchored N-terminal part of the FasL molecule has not been determined. Here we show that post-translational processing of overexpressed and endogenous FasL in T-cells by the disintegrin and metalloprotease ADAM10 generates a 17-kDa N-terminal fragment, which lacks the receptor-binding extracellular domain. This FasL remnant is membrane anchored and further processed by SPPL2a, a member of the signal peptide peptidase-like family of intramembrane-cleaving proteases. SPPL2a cleavage liberates a smaller and highly unstable fragment mainly containing the intracellular FasL domain (FasL ICD). We show that this fragment translocates to the nucleus and is capable of inhibiting gene transcription. With ADAM10 and SPPL2a we have identified two proteases implicated in FasL processing and release of the FasL ICD, which has been shown to be important for retrograde FasL signaling.


Subject(s)
ADAM Proteins/metabolism , Aspartic Acid Endopeptidases/metabolism , Fas Ligand Protein/metabolism , T-Lymphocytes/metabolism , Cell Line, Tumor , Cells, Cultured , Fas Ligand Protein/chemistry , Humans , Microscopy, Confocal , Protein Structure, Tertiary , RNA, Small Interfering
8.
Cell Death Differ ; 14(4): 795-806, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17082815

ABSTRACT

The protein Daxx promotes Fas-mediated cell death through activation of apoptosis signal-regulating kinase 1, leading to the activation of the MAPKs JNK and p38. Owing to the in utero lethality of daxx-deficient mice, the in vivo role of Daxx has been so far difficult to analyze. We have generated transgenic mice expressing a dominant-negative form of Daxx (Daxx-DN) in the T-cell lineage. We show that Daxx is recruited to the Fas receptor upon FasL engagement and that Daxx-DN expression protects activated T cells from Fas-induced cell death, by preventing the death-inducing signal complex to be properly formed. Normal lymphocyte development and homeostasis are nevertheless observed. Interestingly, we report that both in vitro and in vivo stimulation of Daxx-DN T-lymphocytes leads to increased proliferative T-cell responses. This increased proliferation is associated with a marked increase in tyrosine phosphorylation of LAT and ZAP70 as Daxx-DN favor their recruitment to the T-cell receptor (TCR) complex. These findings identify Daxx as a critical regulator of T-lymphocyte homeostasis by decreasing TCR-induced cell proliferation and by promoting Fas-mediated cell death.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Fas Ligand Protein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lymphocyte Activation , Nuclear Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , fas Receptor/physiology , Animals , Apoptosis/physiology , Caspases/metabolism , Cell Proliferation , Co-Repressor Proteins , Genes, Dominant , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Jurkat Cells , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Molecular Chaperones , T-Lymphocytes/chemistry
10.
Oncogene ; 20(10): 1164-75, 2001 Mar 08.
Article in English | MEDLINE | ID: mdl-11313860

ABSTRACT

To investigate Mad1 function in vivo, transgenic mice were generated that express a Mad1 transgene in T lineage cells under the control of the proximal lck promoter. Thymus size in lck-Mad1 transgenic mice is drastically reduced although representation of the various thymocyte sub populations appears normal. To investigate more closely any effects of Mad1 expression on thymocytes, we examined thymic selection using MHC class I-restricted H-Y-TCR transgenic mice. Mad1 expression in vivo reduces the efficiency of positive selection. Furthermore, thymocytes and splenic T cells from lck-Mad1 transgenic mice display a profound proliferative defect in response to activation with either PMA/Ionomycin or immobilized anti-CD3/CD28 antibody. This proliferative defect is not reversed by addition of exogenous IL-2 and is p53-independent. The growth inhibition caused by Mad1 is overcome by expression of active c-Myc.


Subject(s)
Lymphocyte Activation/drug effects , Mitogens/pharmacology , Phosphoproteins/biosynthesis , Repressor Proteins/biosynthesis , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , Antigens, CD/physiology , Blotting, Western , Cell Cycle Proteins , DNA Primers/chemistry , Female , Genes, myc/physiology , H-Y Antigen/immunology , Histocompatibility Antigens Class I/immunology , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Nuclear Proteins , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Resting Phase, Cell Cycle/drug effects , Reverse Transcriptase Polymerase Chain Reaction , S Phase/drug effects , Spleen/immunology , Thymus Gland/pathology , Tumor Suppressor Protein p53/metabolism
11.
Oncogene ; 19(15): 1891-900, 2000 Apr 06.
Article in English | MEDLINE | ID: mdl-10773879

ABSTRACT

In order to study the effect of c-Myc activation in T lymphocytes in vivo, we generated transgenic mice that express a 4-hydroxytamoxifen (4-OHT)-dependent switchable c-myc oncoprotein under the control of the proximal lck promoter. Activation of c-MycER causes no obvious alteration in T cell ontogeny. However, using MHC class I restricted H-Y-TCR transgenic mice, we found that c-Myc activation in vivo enhances the efficiency of positive selection. Moreover, splenic T cells derived from lck-c-mycER transgenic mice in which c-Myc had been activated exhibited increased proliferation in vitro in response to activation with anti-CD3/CD28 antibody. Activation of c-MycER also promotes apoptosis in thymocytes in vitro.


Subject(s)
Apoptosis , Gene Expression Regulation, Neoplastic , Genes, myc , T-Lymphocytes/metabolism , Animals , Cell Differentiation , Cell Division , Mice , Mice, Transgenic , T-Lymphocytes/physiology , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Thymus Gland/cytology
12.
J Biol Chem ; 275(14): 10453-62, 2000 Apr 07.
Article in English | MEDLINE | ID: mdl-10744735

ABSTRACT

Death domain-containing members of the tumor necrosis factor (TNF) receptor family ("death receptors") can induce apoptosis upon stimulation by their natural ligands or by agonistic antibodies. Activated death receptors recruit death domain adapter proteins like Fas-associated death domain protein (FADD), and this ultimately leads to proteolytic activation of the caspase cascade and cell death. Recently, FADD has also been implicated in the regulation of proliferation; functional inhibition of FADD results in p53-dependent impairment of proliferation in activated T-cells. In this study we have further analyzed T-cells derived from transgenic mice expressing a dominant negative FADD mutant (FADD DN) under control of the lck promoter in vitro so as to identify the signaling pathways that become engaged upon T-cell receptor stimulation and that are regulated by death receptors. FADD DN expression inhibits T-cell proliferation, both at the G(0) --> S transition and in the G(1) phase of continuously proliferating cells. We observe a decrease in the release of calcium from intracellular stores after T-cell receptor stimulation, whereas influx of extracellular calcium seems to be unaffected. FADD DN-expressing fibroblasts show a similarly inhibited cell growth and impaired calcium mobilization indicating that the modulation of proliferation and calcium response by death receptors is not cell type-specific.


Subject(s)
Adaptor Proteins, Signal Transducing , Calcium/metabolism , Carrier Proteins/physiology , Cell Cycle/physiology , T-Lymphocytes/physiology , 3T3 Cells , Animals , Apoptosis , Bombesin/pharmacology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Division/drug effects , Fas-Associated Death Domain Protein , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation/immunology , Interleukin-2/genetics , Interleukin-6/genetics , Ionomycin/pharmacology , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Signal Transduction , Spleen/immunology , T-Lymphocytes/cytology , Transcription, Genetic
13.
Nat Cell Biol ; 2(2): E23-5, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10655597

ABSTRACT

The CD95 protein delivers crucial signals for lymphocyte death, and may also negatively regulate T-lymphocyte activation by preventing the influx of calcium ions from the cell's exterior. The block in calcium-ion influx occurs through the activation of acidic sphingomyelinase and the release of ceramide, a metabolite that can also induce cell death.


Subject(s)
Cell Death , Lymphocyte Activation , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , fas Receptor/metabolism , Calcium Signaling , Models, Biological
14.
Genes Dev ; 13(11): 1367-81, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10364155

ABSTRACT

Expression of c-Myc sensitizes cells to a wide range of pro-apoptotic stimuli. We here show that this pro-apoptotic effect is mediated through release of mitochondrial holocytochrome c into the cytosol. First, activation of c-Myc triggers release of cytochrome c from mitochondria. This release is caspase-independent and blocked by the survival factor IGF-1. Second, c-Myc-induced apoptosis is blocked by microinjection of anticytochrome c antibody. In addition, we show that microinjection of holocytochrome c mimics the effect of c-Myc activation, sensitizing cells to DNA damage and to the CD95 pathway. Both p53 and CD95/Fas signaling have been implicated in c-Myc-induced apoptosis but neither was required for c-Myc-induced cytochrome c release. Nonetheless, inhibition of CD95 signaling in fibroblasts did prevent c-Myc-induced apoptosis, apparently by obstructing the ability of cytosolic cytochrome c to activate caspases. We conclude that c-Myc promotes apoptosis by causing the release of cytochrome c, but the ability of cytochrome c to activate apoptosis is critically dependent upon other signals.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , Cytochrome c Group/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Animals , Antibodies, Monoclonal/metabolism , Carrier Proteins/metabolism , Caspases/metabolism , Cell Line , Cytosol/metabolism , Enzyme Activation , Fas-Associated Death Domain Protein , Microinjections , Mitochondria/metabolism , Rats , Tumor Suppressor Protein p53/metabolism , X-Rays , fas Receptor/metabolism
15.
Oncogene ; 18(13): 2181-8, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10327064

ABSTRACT

The contact of natural killer (NK) cells with foreign cells and with certain virus-infected or tumor cells triggers the cytolytic machinery of NK cells. This triggering leads to exocytosis of the cytotoxic NK cell granules. The oncoproteins c-Myc and E1A render cells vulnerable to NK cell mediated cytolysis yet the mechanisms of sensitization are not well understood. In a model where foreign cells (rat fibroblasts) were cocultured with human IL-2 activated NK cells, we observed that NK cells were capable of efficiently killing their targets only if the cells overexpressed the oncogene c-Myc or E1A. Both the parental and the oncogene expressing fibroblasts similarly triggered phosphoinositide hydrolysis in the bound NK cells, demonstrating that NK cells were cytolytically activated in contact with both resistant parental and oncogene expressing sensitive target fibroblasts. The cell death was independent of wild-type p53 and was not inhibited by an anti-apoptotic protein EIB19K. These results provided evidence that c-Myc and E1A activated the NK cell induced cytolysis at a post-triggering stage of NK cell-target cell interaction. In consistence, the c-Myc and E1A overexpressing fibroblasts were more sensitive to the cytolytic effects of isolated NK cell-derived granules than parental cells. The data indicate that oncogenes activate the cytotoxicity of NK cell granules. This mechanism can have a role in directing the cytolytic action of NK cells towards the virus-infected and cancer cells.


Subject(s)
Adenovirus E1A Proteins/physiology , Cytoplasmic Granules/metabolism , Cytotoxicity, Immunologic/physiology , Killer Cells, Natural/immunology , Proto-Oncogene Proteins c-myc/physiology , Actins/chemistry , Adenovirus E1A Proteins/genetics , Animals , Apoptosis/genetics , Biopolymers , Cell Adhesion , Cell Membrane/drug effects , Exocytosis , Fas Ligand Protein , Fibroblasts/immunology , Genes, myc , Genes, p53 , Humans , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Membrane Glycoproteins/physiology , Phosphatidylinositols/physiology , Rats , Recombinant Fusion Proteins/physiology , Signal Transduction , Transfection , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology , Tumor Suppressor Protein p53/physiology , fas Receptor/physiology
16.
J Immunol ; 161(10): 5296-302, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9820502

ABSTRACT

Thy-1 is a cell surface glycoprotein expressed mainly on brain and lymphoid tissue. Although the functions of Thy-1 are incompletely understood, evidence exists that Thy-1 participates in T cell activation. To examine the functional role of Thy-1 in cutaneous immune responses in vivo, Thy-1 gene-targeted mice (Thy-1-/-) and wild-type mice (Thy-1+/+) were immunized with the hapten oxazolone. After challenge with oxazolone, contact hypersensitivity responses in Thy-1-/- mice were reduced by 25% compared with Thy-1+/+ mice. Likewise, irritant dermatitis induced by croton oil was also decreased. In addition, Thy-1-/- mice showed a significantly reduced delayed-type hypersensitivity response after injection of allogeneic spleen cells into the hind footpads of allosensitized animals when compared with Thy-1+/+ mice. Moreover, proliferative responses to immobilized anti-CD3 were decreased in peripheral Thy-1-/- lymphocytes; this decrease was associated with a significantly reduced intracellular Ca2+ influx and protein tyrosine phosphorylation, indicating impairment of early lymphocyte activation. In contrast, the T cell proliferation induced by mitogens was normal, suggesting that Thy-1 expression weakly contributes to TCR-mediated T cell activation. Epidermal Langerhans cells and bone marrow-derived dendritic cells from Thy-1-/- mice exhibited a normal expression of costimulatory surface molecules as well as an unaltered ability to stimulate allogeneic T cells. Taken together, these findings demonstrate that a lack of Thy-1 expression does not generally compromise the immune system; however, Thy-1 expression may be involved in the fine-tuning of T cell-mediated immune responses.


Subject(s)
Immunity, Cellular/genetics , Skin/immunology , Thy-1 Antigens/genetics , Animals , Antibodies, Monoclonal/pharmacology , Bone Marrow Cells/immunology , CD3 Complex/immunology , Calcium/metabolism , Calcium Signaling/immunology , Crosses, Genetic , Cytokines/metabolism , Dendritic Cells/immunology , Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , Epidermal Cells , Epidermis/immunology , Hypersensitivity, Delayed/genetics , Hypersensitivity, Delayed/immunology , Isoantigens/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Mutant Strains , Phosphorylation , Skin/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tyrosine/metabolism
17.
Trends Genet ; 14(9): 364-7, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9769732

ABSTRACT

The MYC proto-oncogene has long been implicated in the control of normal cell growth and its deregulation is associated with the development of neoplasia. The MYC protein has a well-established role as a component of signal-transduction pathways promoting both proliferation and apoptosis. Because signalling pathways that drive cell death and cell proliferation are so tightly coupled, a synergy between genetic lesions leading to suppression of cell death and those promoting cell proliferation is observed during carcinogenesis. We discuss such synergy with respect to the cooperating oncogenes MYC, RAS and BCL2.


Subject(s)
Oncogenes , Animals , Apoptosis , Cell Division , Genes, bcl-2 , Genes, myc , Genes, ras , Humans , Neoplasms/etiology , Neoplasms/genetics , Proto-Oncogene Mas , Signal Transduction
18.
Curr Biol ; 8(8): 467-70, 1998 Apr 09.
Article in English | MEDLINE | ID: mdl-9550704

ABSTRACT

Members of the tumour necrosis factor (TNF) receptor family exert pleiotropic effects and can trigger both apoptosis and proliferation [1]. In their cytoplasmic region, some of these receptors share a conserved sequence motif - the 'death domain' - which is required for transduction of the apoptotic signal by recruiting other death-domain-containing adaptor molecules like the Fas-associated protein FADD/MORT1 or the TNF receptor-associated protein TRADD [2-4]. FADD links the receptor signal to the activation of the caspase family of cysteine proteases [5,6]. Functional inactivation of individual receptor family members often fails to exhibit a distinctive phenotype, probably because of redundancy [7-9]. To circumvent this problem, we used a dominant-negative mutant of FADD (FADD-DN) which should block all TNF receptor family members that use FADD as an adaptor. We established transgenic mice expressing FADD-DN under the influence of the lck promoter and investigated the consequences of its expression in T cells. As expected, FADD-DN thymocytes were protected from death induced by CD95 (Fas/Apo1), whereas apoptosis induced by ultraviolet (UV) irradiation, anti-CD3 antibody treatment or dexamethasone was unaffected, as was spontaneous cell death. Surprisingly, however, we also observed profound inhibition of thymocyte proliferation in vivo and of activation-induced proliferation of thymocytes and mature T cells in vitro. This inhibition of proliferation was not due to increased cell death and appeared to be p53 dependent.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/immunology , T-Lymphocytes/cytology , Thymus Gland/immunology , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis , Carrier Proteins/genetics , Cell Division , Cells, Cultured , Fas-Associated Death Domain Protein , Genes, Dominant , Lymphocyte Activation , Mice , Mice, Transgenic , Mitogens/pharmacology , Mutation , Receptors, Tumor Necrosis Factor/immunology , Tumor Suppressor Protein p53/genetics , fas Receptor/pharmacology
19.
Oncogene ; 17(22): 2811-8, 1998 Dec 03.
Article in English | MEDLINE | ID: mdl-9879987

ABSTRACT

Expression of the proto-oncogene c-myc stimulates cell proliferation in the presence of the appropriate survival factors and triggers apoptosis in their absence; this dual capacity ensures that cell growth is restricted to the correct paracrine environment and is thereby strictly controlled. Recently our laboratory demonstrated that c-Myc-induced apoptosis requires the CD95 death receptor pathway and that insulin-like growth factor (IGF-1) signalling suppresses this killing. To investigate further the links between c-Myc and IGF-1 pathways in CD95-induced apoptosis, we examined the effects of c-Myc and a downstream IGF-1 survival kinase, Akt, on killing mediated by CD95 and its recruited effector proteins (FADD and caspase-8). Here, we show that c-Myc activation does not exacerbate killing induced by FADD or pro-caspase-8, which narrows the point at which c-Myc exerts its action downstream of the interaction of CD95 with its ligand and upstream of FADD. We show further that activated Akt suppresses CD95-induced apoptosis and that Akt exerts its activity at a point downstream of FADD but upstream of caspase-8. These results restrict the possible mechanisms by which CD95-induced apoptosis is modulated by death signals and survival factors.


Subject(s)
Apoptosis , Arabidopsis Proteins , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins , Signal Transduction , fas Receptor/metabolism , 3T3 Cells , Animals , Antibodies/pharmacology , Apoptosis/drug effects , Blotting, Western , Caspase 8 , Caspase 9 , Caspases/genetics , Caspases/metabolism , Cell Line , Cell Survival/drug effects , Culture Media, Serum-Free , Fatty Acid Desaturases/antagonists & inhibitors , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/metabolism , Mice , Plant Proteins/antagonists & inhibitors , Plant Proteins/genetics , Plant Proteins/metabolism , Protein Precursors/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt , Rats , Signal Transduction/drug effects , Sirolimus/pharmacology , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Transfection , fas Receptor/immunology
20.
Science ; 278(5341): 1305-9, 1997 Nov 14.
Article in English | MEDLINE | ID: mdl-9360929

ABSTRACT

Induction of apoptosis by oncogenes like c-myc may be important in restraining the emergence of neoplasia. However, the mechanism by which c-myc induces apoptosis is unknown. CD95 (also termed Fas or APO-1) is a cell surface transmembrane receptor of the tumor necrosis factor receptor family that activates an intrinsic apoptotic suicide program in cells upon binding either its ligand CD95L or antibody. c-myc-induced apoptosis was shown to require interaction on the cell surface between CD95 and its ligand. c-Myc acts downstream of the CD95 receptor by sensitizing cells to the CD95 death signal. Moreover, IGF-I signaling and Bcl-2 suppress c-myc-induced apoptosis by also acting downstream of CD95. These findings link two apoptotic pathways previously thought to be independent and establish the dependency of Myc on CD95 signaling for its killing activity.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , Membrane Glycoproteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , fas Receptor/metabolism , 3T3 Cells , Animals , Autocrine Communication , Carrier Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Fas Ligand Protein , Fas-Associated Death Domain Protein , Gene Expression Regulation , Genes, myc , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor I/physiology , Mice , Proto-Oncogene Proteins c-bcl-2/pharmacology , Proto-Oncogene Proteins c-bcl-2/physiology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...