Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Dan. med. j ; 61(3): 1-4, mar. 2014.
Article in English | BIGG - GRADE guidelines | ID: biblio-967792

ABSTRACT

Stress ulcer prophylaxis (SUP) is commonly used in the intensive care unit (ICU), and is recommended in the Surviving Sepsis Campaign guidelines 2012. The present guideline from the Danish Society of Intensive Care Medicine and the Danish Society of Anesthesiology and Intensive Care Medicine sums up current evidence and gives clinical recommendations for SUP in the ICU. The GRADE approach was used for grading the evidence (www.gradeworkinggroup.org). In conclusion, existing meta-analyses have been underpowered to reach firm conclusions. We recommend not using SUP routinely for adult critically ill patients in the ICU outside the context of randomized controlled trials (GRADE 1C). No robust evidence supports recommendations for subpopulations in the ICU such as septic, burn, trauma, cardiothoracic or enterally fed patients. However, if SUP is considered clinically indicated in individual patients, we suggest using proton pump inhibitors over histamine-2-receptor antagonists (GRADE 2C)


Subject(s)
Humans , Adult , Ulcer , Ulcer/prevention & control , Ulcer/therapy , Intensive Care Units
2.
J Biol Chem ; 276(26): 23954-61, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11287428

ABSTRACT

Recent studies suggest that trimerization of Fas is insufficient for apoptosis induction and indicate that super-aggregation of trimerized Fas might be prerequisite. For many cell surface receptors, cross-linking by multivalent ligands or antibodies induces their lateral segregation within the plasma membrane and co-localization into "caps" on one pole of the cell. In this study, we show that capping of Fas is essential for optimal function and that capping is ceramide-dependent. In Jurkat T lymphocytes and in primary cultures of hepatocytes, ceramide elevation was detected as early as 15-30 s and peaked at 1 min after CH-11 and Jo2 anti-Fas antibody treatment, respectively. Capping was detected 30 s after Fas ligation, peaked at 2 min, and was maintained at a lower level for as long as 30 min in both cell types. Ceramide generation appeared essential for capping. Acid sphingomyelinase -/- hepatocytes were defective in Jo2-induced ceramide generation, capping, and apoptosis, and nanomolar concentrations of C(16)-ceramide restored these events. To further explore the role of ceramide in capping of Fas, we employed FLAG-tagged soluble Fas ligand (sFasL), which binds trimerized Fas but is unable to induce capping or apoptosis in Jurkat cells. Cross-linking of sFasL with M2 anti-FLAG antibody induced both events. Pretreatment of cells with natural C(16)-ceramide bypassed the necessity for forced antibody cross-linking and enabled sFasL to cap and kill. The presence of intact sphingolipid-enriched membrane domains may be essential for Fas capping since their disruption with cholesterol-depleting agents abrogated capping and prevented apoptosis. These data suggest that capping is a ceramide-dependent event required for optimal Fas signaling in some cells.


Subject(s)
Apoptosis , Ceramides/physiology , fas Receptor/metabolism , fas Receptor/physiology , Animals , Antibodies/immunology , Apoptosis/drug effects , Cells, Cultured , Ceramides/biosynthesis , Ceramides/pharmacology , Fas Ligand Protein , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Jurkat Cells , Kinetics , Membrane Glycoproteins/pharmacology , Membrane Microdomains/drug effects , Mice , Mice, Knockout , Receptor Aggregation , Sphingomyelin Phosphodiesterase/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , fas Receptor/immunology
3.
Eur J Immunol ; 31(1): 243-9, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11265640

ABSTRACT

Recent studies have shown that in humans the germinal center reactions produce three types of V(D)J mutated B cells in similar proportions, i.e. Ig-switched, IgD-IgM+ (IgM-only) and IgD+IgM+ cells, and that together they form the CD27+ compartment of recirculating B cells. We investigated the Ig isotype switch capacity of these cells. Peripheral blood B subsets were sorted and IgG subclass secretion in presence or absence of IL-4 was compared in B cell assays which lead to Ig secretion in all (coculture with EL-4 thymoma cells) or only in CD27+ (CD40L stimulation) B cells. Already switched IgG+ B cells showed no significant sequential switch and IgM-only cells also had a low switch capacity, but IgD+CD27+ switched as much as IgD+CD27- B cells to all IgG subclasses. Thus, in switched B cells some alterations compromising further switch options occur frequently; IgM-only cells may result from aborted switch. However, IgD+CD27+ human B cells, extensively V(D)J mutated and "naive" regarding switch, build up a repertoire of B cells combining (1) novel cross-reactive specificities, (2) increased differentiation capacity (including after T-independent stimulation by Staphylococcus aureus Cowan I) and (3) the capacity to produce appropriate isotypes when they respond to novel pathogens.


Subject(s)
B-Lymphocyte Subsets/metabolism , Germinal Center/metabolism , Immunoglobulin Class Switching , Immunoglobulin D/biosynthesis , Immunoglobulin G/classification , Immunoglobulin M/biosynthesis , Tumor Necrosis Factor Receptor Superfamily, Member 7/analysis , Humans , Immunoglobulin G/biosynthesis , Lymphocyte Activation
4.
J Biol Chem ; 276(22): 19548-54, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11262395

ABSTRACT

The Bcl-2 family of proteins plays a central regulatory role in apoptosis. We have identified a novel, widely expressed Bcl-2 member which we have named Bcl-rambo. Bcl-rambo shows overall structural homology to the anti-apoptotic Bcl-2 members containing conserved Bcl-2 homology (BH) motifs 1, 2, 3, and 4. Unlike Bcl-2, however, the C-terminal membrane anchor region is preceded by a unique 250 amino acid insertion containing two tandem repeats. No interaction of Bcl-rambo with either anti-apoptotic (Bcl-2, Bcl-x(L), Bcl-w, A1, MCL-1, E1B-19K, and BHRF1) or pro-apoptotic (Bax, Bak, Bik, Bid, Bim, and Bad) members of the Bcl-2 family was observed. In mammalian cells, Bcl-rambo was localized to mitochondria, and its overexpression induces apoptosis that is specifically blocked by the caspase inhibitors, IAPs, whereas inhibitors controlling upstream events of either the 'death receptor' (FLIP, FADD-DN) or the 'mitochondrial' pro-apoptotic pathway (Bcl-x(L)) had no effect. Surprisingly, the Bcl-rambo cell death activity was induced by its membrane-anchored C-terminal domain and not by the Bcl-2 homology region. Thus, Bcl-rambo constitutes a novel type of pro-apoptotic Bcl-2 member that triggers cell death independently of its BH motifs.


Subject(s)
Apoptosis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/chemistry , Amino Acid Motifs , Amino Acid Sequence , Blotting, Northern , Blotting, Western , Cell Death , Cell Line , Cloning, Molecular , Cytochrome c Group/metabolism , DNA, Complementary/metabolism , Gene Library , HeLa Cells , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Models, Genetic , Molecular Sequence Data , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Tissue Distribution , Transfection
5.
J Exp Med ; 192(11): 1677-84, 2000 Dec 04.
Article in English | MEDLINE | ID: mdl-11104810

ABSTRACT

A proliferation-inducing ligand (APRIL) is a ligand of the tumor necrosis factor (TNF) family that stimulates tumor cell growth in vitro and in vivo. Expression of APRIL is highly upregulated in many tumors including colon and prostate carcinomas. Here we identify B cell maturation antigen (BCMA) and transmembrane activator and calcium modulator and cyclophilin ligand (CAML) interactor (TACI), two predicted members of the TNF receptor family, as receptors for APRIL. APRIL binds BCMA with higher affinity than TACI. A soluble form of BCMA, which inhibits the proliferative activity of APRIL in vitro, decreases tumor cell proliferation in nude mice. Growth of HT29 colon carcinoma cells is blocked when mice are treated once per week with the soluble receptor. These results suggest an important role for APRIL in tumorigenesis and point towards a novel anticancer strategy.


Subject(s)
Adaptor Proteins, Signal Transducing , B-Lymphocytes/physiology , Cell Transformation, Neoplastic , Membrane Proteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , 3T3 Cells , Animals , B-Cell Activating Factor , B-Cell Maturation Antigen , Carrier Proteins/metabolism , Cell Division , Cell Line, Transformed , HT29 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Nude , Neoplasms/therapy , Receptors, Tumor Necrosis Factor/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Solubility , Transmembrane Activator and CAML Interactor Protein , Tumor Cells, Cultured , Tumor Necrosis Factor Ligand Superfamily Member 13 , Tumor Necrosis Factor-alpha/genetics
6.
J Exp Med ; 192(5): 647-58, 2000 Sep 04.
Article in English | MEDLINE | ID: mdl-10974031

ABSTRACT

The short life span of granulocytes, which limits many inflammatory responses, is thought to be influenced by the Bcl-2 protein family, death receptors such as CD95 (Fas/APO-1), stress-activated protein kinases such as p38 mitogen-activated protein kinase (MAPK), and proinflammatory cytokines like granulocyte colony-stimulating factor (G-CSF). To clarify the roles of these various regulators in granulocyte survival, we have investigated the spontaneous apoptosis of granulocytes in culture and that induced by Fas ligand or chemotherapeutic drugs, using cells from normal, CD95-deficient lpr, or vav-bcl-2 transgenic mice. CD95-induced apoptosis, which required receptor aggregation by recombinant Fas ligand or the membrane-bound ligand, was unaffected by G-CSF treatment or Bcl-2 overexpression. Conversely, spontaneous and drug-induced apoptosis occurred normally in lpr granulocytes but were suppressed by G-CSF treatment or Bcl-2 overexpression. Although activation of p38 MAPK has been implicated in granulocyte death, their apoptosis actually was markedly accelerated by specific inhibitors of this kinase. These results suggest that G-CSF promotes granulocyte survival largely through the Bcl-2-controlled pathway, whereas CD95 regulates a distinct pathway to apoptosis that is not required for either their spontaneous or drug-induced death. Moreover, p38 MAPK signaling contributes to granulocyte survival rather than their apoptosis.


Subject(s)
Apoptosis , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/physiology , Membrane Glycoproteins/physiology , Mitogen-Activated Protein Kinases/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Cell Survival , Fas Ligand Protein , Humans , Mice , Mice, Inbred C57BL , Tumor Cells, Cultured , fas Receptor/physiology , p38 Mitogen-Activated Protein Kinases
7.
J Immunol ; 165(3): 1337-43, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10903735

ABSTRACT

Jun kinase signaling can be elicited by death receptor activation, but the mechanism and significance of this event are still unclear. It has been reported that cross-linking Abs to Fas trigger c-Jun N-terminal kinase (JNK) signaling via caspase-mediated activation of MEKK1 (JNK kinase kinase), elevation of ceramide levels or by recruitment of death domain associated protein (DAXX) to Fas. The effect of physiological ligand for Fas on JNK signaling was never investigated, although evidence is accumulating that Fas ligand is able to induce cellular responses distinct from those evoked by Ab-mediated cross-linking of Fas. Therefore, we investigated the effect of Fas ligand on JNK signaling. Like its ability to induce cell death, Fas ligand reliably activated JNK only upon extensive aggregation of the receptor. Although this was partially dependent on caspase activation, DAXX was not required. DAXX and other death receptor-associated proteins, which have been reported to bind directly or indirectly to Fas, such as receptor interacting protein (RIP) and RIP-associated ICH-1/CED-3-homologous protein with a death domain (RAIDD), were shown to be dispensable for Fas ligand-induced apoptosis.


Subject(s)
Apoptosis/immunology , Carrier Proteins/physiology , Intracellular Signaling Peptides and Proteins , Lymphocytes/enzymology , Membrane Glycoproteins/physiology , Mitogen-Activated Protein Kinases/metabolism , Nuclear Proteins , Proteins/physiology , Receptor Aggregation/immunology , fas Receptor/physiology , Adaptor Proteins, Signal Transducing , Animals , Apoptosis/genetics , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Caspases/physiology , Cell Line, Transformed , Co-Repressor Proteins , Enzyme Activation/immunology , Fas Ligand Protein , Genetic Vectors/chemical synthesis , Genetic Vectors/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases , Jurkat Cells , Ligands , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Molecular Chaperones , Protein Biosynthesis , Proteins/genetics , Receptor-Interacting Protein Serine-Threonine Kinases , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Cells, Cultured , fas Receptor/immunology , fas Receptor/metabolism
8.
Curr Biol ; 10(11): 640-8, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10837247

ABSTRACT

BACKGROUND: Activation of Fas (CD95) by its ligand (FasL) rapidly induces cell death through recruitment and activation of caspase-8 via the adaptor protein Fas-associated death domain protein (FADD). However, Fas signals do not always result in apoptosis but can also trigger a pathway that leads to proliferation. We investigated the level at which the two conflicting Fas signals diverge and the protein(s) that are implicated in switching the response. RESULTS: Under conditions in which proliferation of CD3-activated human T lymphocytes is increased by recombinant FasL, there was activation of the transcription factors NF-kappaB and AP-1 and recruitment of the caspase-8 inhibitor and FADD-interacting protein FLIP (FLICE-like inhibitory protein). Fas-recruited FLIP interacts with TNF-receptor associated factors 1 and 2, as well as with the kinases RIP and Raf-1, resulting in the activation of the NF-kappaB and extracellular signal regulated kinase (Erk) signaling pathways. In T cells these two signal pathways are critical for interleukin-2 production. Increased expression of FLIP in T cells resulted in increased production of interleukin-2. CONCLUSIONS: We provide evidence that FLIP is not simply an inhibitor of death-receptor-induced apoptosis but that it also mediates the activation of NF-kappaB and Erk by virtue of its capacity to recruit adaptor proteins involved in these signaling pathways.


Subject(s)
Carrier Proteins/metabolism , Caspase Inhibitors , Intracellular Signaling Peptides and Proteins , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Signal Transduction/physiology , T-Lymphocytes/physiology , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein , CD3 Complex/physiology , Caspase 8 , Caspase 9 , Cells, Cultured , Fas Ligand Protein , Humans , Interleukin-2/biosynthesis , Membrane Glycoproteins/pharmacology , Proteins/metabolism , Proto-Oncogene Proteins c-raf , Receptor-Interacting Protein Serine-Threonine Kinases , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , TNF Receptor-Associated Factor 1 , TNF Receptor-Associated Factor 2 , Transcription Factor AP-1/metabolism , fas Receptor/physiology
10.
J Immunol Methods ; 237(1-2): 159-73, 2000 Apr 03.
Article in English | MEDLINE | ID: mdl-10725460

ABSTRACT

TNF receptor family members fused to the constant domain of immunoglobulin G have been widely used as immunoadhesins in basic in vitro and in vivo research and in some clinical applications. In this study, we assemble soluble, high avidity chimeric receptors on a pentameric scaffold derived from the coiled-coil domain of cartilage oligomeric matrix protein (COMP). The affinity of Fas and CD40 (but not TNFR-1 and TRAIL-R2) to their ligands is increased by fusion to COMP, when compared to the respective Fc chimeras. In functional assays, Fas:COMP was at least 20-fold more active than Fas:Fc at inhibiting the action of sFasL, and CD40:COMP could block CD40L-mediated proliferation of B cells, whereas CD40:Fc could not. In conclusion, members of the TNF receptor family can display high specificity and excellent avidity for their ligands if they are adequately multimerized.


Subject(s)
Membrane Glycoproteins/antagonists & inhibitors , Animals , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , CD40 Antigens/metabolism , CD40 Ligand , Cartilage Oligomeric Matrix Protein , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/pharmacology , Fas Ligand Protein , Glycoproteins/genetics , Glycoproteins/metabolism , Glycoproteins/pharmacology , Humans , Jurkat Cells , Ligands , Lymphocyte Activation/drug effects , Matrilin Proteins , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Receptors, Fc/genetics , Receptors, Fc/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Solubility , Tumor Cells, Cultured , fas Receptor/metabolism
11.
FEBS Lett ; 468(2-3): 134-6, 2000 Feb 25.
Article in English | MEDLINE | ID: mdl-10692573

ABSTRACT

Death domain containing members of the tumor necrosis factor receptor (TNFR) superfamily can induce apoptosis or cell activation. However, the mechanisms by which these opposing programs are selected remain unclear. Frequently, NF-kappaB activation conveys protection against cell death. We show that the serine/threonine kinase RIP that is required for TNF-induced NF-kappaB activation is processed by caspase-8 into a dominant-negative (DN) fragment during death receptor-induced apoptosis, thereby leading to a blockade of NF-kappaB-mediated anti-apoptotic signals. Our results suggest that cleavage of RIP is part of an amplification loop which is triggered by Fas and most likely by other death receptors.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Cell Survival/physiology , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Binding Sites , Humans , Jurkat Cells , Models, Biological , Mutagenesis, Site-Directed , Proteins/chemistry , Proteins/genetics , Receptor-Interacting Protein Serine-Threonine Kinases , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tumor Cells, Cultured , fas Receptor/physiology
12.
Nat Immunol ; 1(6): 489-95, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11101870

ABSTRACT

Cell death is achieved by two fundamentally different mechanisms: apoptosis and necrosis. Apoptosis is dependent on caspase activation, whereas the caspase-independent necrotic signaling pathway remains largely uncharacterized. We show here that Fas kills activated primary T cells efficiently in the absence of active caspases, which results in necrotic morphological changes and late mitochondrial damage but no cytochrome c release. This Fas ligand-induced caspase-independent death is absent in T cells that are deficient in either Fas-associated death domain (FADD) or receptor-interacting protein (RIP). RIP is also required for necrotic death induced by tumor necrosis factor (TNF) and TNF-related apoptosis-inducing ligand (TRAIL). In contrast to its role in nuclear factor kappa B activation, RIP requires its own kinase activity for death signaling. Thus, Fas, TRAIL and TNF receptors can initiate cell death by two alternative pathways, one relying on caspase-8 and the other dependent on the kinase RIP.


Subject(s)
Adaptor Proteins, Signal Transducing , Caspases/metabolism , Cell Death/physiology , Proteins/metabolism , fas Receptor/metabolism , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins , Carrier Proteins/metabolism , Caspase 8 , Caspase 9 , Fas Ligand Protein , Fas-Associated Death Domain Protein , Humans , In Vitro Techniques , Jurkat Cells , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Models, Biological , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/metabolism
13.
J Exp Med ; 189(11): 1747-56, 1999 Jun 07.
Article in English | MEDLINE | ID: mdl-10359578

ABSTRACT

Members of the tumor necrosis factor (TNF) family induce pleiotropic biological responses, including cell growth, differentiation, and even death. Here we describe a novel member of the TNF family, designated BAFF (for B cell activating factor belonging to the TNF family), which is expressed by T cells and dendritic cells. Human BAFF was mapped to chromosome 13q32-34. Membrane-bound BAFF was processed and secreted through the action of a protease whose specificity matches that of the furin family of proprotein convertases. The expression of BAFF receptor appeared to be restricted to B cells. Both membrane-bound and soluble BAFF induced proliferation of anti-immunoglobulin M-stimulated peripheral blood B lymphocytes. Moreover, increased amounts of immunoglobulins were found in supernatants of germinal center-like B cells costimulated with BAFF. These results suggest that BAFF plays an important role as costimulator of B cell proliferation and function.


Subject(s)
B-Lymphocytes/immunology , Membrane Proteins/physiology , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/physiology , Amino Acid Sequence , Animals , B-Cell Activating Factor , B-Lymphocytes/cytology , Base Sequence , Cell Division , Cell Line , Chromosome Mapping , Chromosomes, Human, Pair 13/genetics , Cloning, Molecular , DNA Primers/genetics , Dendritic Cells/immunology , Humans , Ligands , Lymphocyte Activation , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Receptors, Tumor Necrosis Factor/genetics , Sequence Homology, Amino Acid , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/genetics
14.
Science ; 282(5388): 490-3, 1998 Oct 16.
Article in English | MEDLINE | ID: mdl-9774279

ABSTRACT

Toxic epidermal necrolysis (TEN, Lyell's syndrome) is a severe adverse drug reaction in which keratinocytes die and large sections of epidermis separate from the dermis. Keratinocytes normally express the death receptor Fas (CD95); those from TEN patients were found to express lytically active Fas ligand (FasL). Antibodies present in pooled human intravenous immunoglobulins (IVIG) blocked Fas-mediated keratinocyte death in vitro. In a pilot study, 10 consecutive individuals with clinically and histologically confirmed TEN were treated with IVIG; disease progression was rapidly reversed and the outcome was favorable in all cases. Thus, Fas-FasL interactions are directly involved in the epidermal necrolysis of TEN, and IVIG may be an effective treatment.


Subject(s)
Apoptosis , Immunoglobulins, Intravenous/therapeutic use , Keratinocytes/pathology , Stevens-Johnson Syndrome/therapy , fas Receptor/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Blocking/immunology , Antibodies, Blocking/therapeutic use , Child , Dermis/pathology , Disease Progression , Epidermis/pathology , Fas Ligand Protein , Female , Humans , Jurkat Cells , Keratinocytes/metabolism , Male , Membrane Glycoproteins/metabolism , Middle Aged , Pilot Projects , Stevens-Johnson Syndrome/pathology , fas Receptor/immunology
15.
J Exp Med ; 188(6): 1185-90, 1998 Sep 21.
Article in English | MEDLINE | ID: mdl-9743536

ABSTRACT

Members of the tumor necrosis factor (TNF) family induce pleiotropic biological responses, including cell growth, differentiation, and even death. Here we describe a novel member of the TNF family designated APRIL (for a proliferation-inducing ligand). Although transcripts of APRIL are of low abundance in normal tissues, high levels of mRNA are detected in transformed cell lines, and in human cancers of colon, thyroid, and lymphoid tissues in vivo. The addition of recombinant APRIL to various tumor cells stimulates their proliferation. Moreover, APRIL-transfected NIH-3T3 cells show an increased rate of tumor growth in nude mice compared with the parental cell line. These findings suggest that APRIL may be implicated in the regulation of tumor cell growth.


Subject(s)
Growth Substances/physiology , Membrane Proteins/physiology , Tumor Cells, Cultured/pathology , Tumor Necrosis Factor-alpha/physiology , 3T3 Cells , Amino Acid Sequence , Animals , Cell Division/drug effects , Humans , Ligands , Lymphoma, B-Cell , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Neoplasm Transplantation , RNA, Messenger/biosynthesis , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13
16.
J Exp Med ; 187(8): 1205-13, 1998 Apr 20.
Article in English | MEDLINE | ID: mdl-9547332

ABSTRACT

Human Fas ligand (L) (CD95L) and tumor necrosis factor (TNF)-alpha undergo metalloproteinase-mediated proteolytic processing in their extracellular domains resulting in the release of soluble trimeric ligands (soluble [s]FasL, sTNF-alpha) which, in the case of sFasL, is thought to be implicated in diseases such as hepatitis and AIDS. Here we show that the processing of sFasL occurs between Ser126 and Leu127. The apoptotic-inducing capacity of naturally processed sFasL was reduced by >1,000-fold compared with membrane-bound FasL, and injection of high doses of recombinant sFasL in mice did not induce liver failure. However, soluble FasL retained its capacity to interact with Fas, and restoration of its cytotoxic activity was achieved both in vitro and in vivo with the addition of cross-linking antibodies. Similarly, the marginal apoptotic activity of recombinant soluble TNF-related apoptosis-inducing ligand (sTRAIL), another member of the TNF ligand family, was greatly increased upon cross-linking. These results indicate that the mere trimerization of the Fas and TRAIL receptors may not be sufficient to trigger death signals. Thus, the observation that sFasL is less cytotoxic than membrane-bound FasL may explain why in certain types of cancer, systemic tissue damage is not detected, even though the levels of circulating sFasL are high.


Subject(s)
Apoptosis , Liver/pathology , Membrane Glycoproteins/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , Down-Regulation , Fas Ligand Protein , Humans , Mice , Mice, Inbred BALB C , Models, Biological , Molecular Sequence Data , Protein Binding , Receptors, Tumor Necrosis Factor/metabolism , Solubility , Tumor Necrosis Factor-alpha/metabolism
17.
FEBS Lett ; 416(3): 329-34, 1997 Oct 27.
Article in English | MEDLINE | ID: mdl-9373179

ABSTRACT

Two receptors for TRAIL, designated TRAIL-R2 and TRAIL-R3, have been identified. Both are members of the tumor necrosis factor receptor family. TRAIL-R2 is structurally similar to the death-domain-containing receptor TRAIL-R1 (DR-4), and is capable of inducing apoptosis. In contrast, TRAIL-R3 does not promote cell death. TRAIL-R3 is highly glycosylated and is membrane bound via a putative phosphatidylinositol anchor. The extended structure of TRAIL-R3 is due to the presence of multiple threonine-, alanine-, proline- and glutamine-rich repeats (TAPE repeats). TRAIL-R2 shows a broad tissue distribution, whereas the expression of TRAIL-R3 is restricted to peripheral blood lymphocytes (PBLs) and skeletal muscle. All three TRAIL receptors bind TRAIL with similar affinity, suggesting a complex regulation of TRAIL-mediated signals.


Subject(s)
Membrane Glycoproteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Sequence , Apoptosis , Apoptosis Regulatory Proteins , Cell Line , Chromosome Mapping , Cloning, Molecular , GPI-Linked Proteins , Humans , Kinetics , Lymphocytes/metabolism , Molecular Sequence Data , Muscle, Skeletal/metabolism , Polymerase Chain Reaction , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Member 10c , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Sequence Tagged Sites , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand , Transfection , Tumor Necrosis Factor Decoy Receptors
18.
J Biol Chem ; 272(30): 18827-33, 1997 Jul 25.
Article in English | MEDLINE | ID: mdl-9228058

ABSTRACT

The death-inducing receptor Fas is activated when cross-linked by the type II membrane protein Fas ligand (FasL). When human soluble FasL (sFasL, containing the extracellular portion) was expressed in human embryo kidney 293 cells, the three N-linked glycans of each FasL monomer were found to be essential for efficient secretion. Based on the structure of the closely related lymphotoxin alpha-tumor necrosis factor receptor I complex, a molecular model of the FasL homotrimer bound to three Fas molecules was generated using knowledge-based protein modeling methods. Point mutations of amino acid residues predicted to affect the receptor-ligand interaction were introduced at three sites. The F275L mutant, mimicking the loss of function murine gld mutation, exhibited a high propensity for aggregation and was unable to bind to Fas. Mutants P206R, P206D, and P206F displayed reduced cytotoxicity toward Fas-positive cells with a concomitant decrease in the binding affinity for the recombinant Fas-immunoglobulin Fc fusion proteins. Although the cytotoxic activity of mutant Y218D was unaltered, mutant Y218R was inactive, correlating with the prediction that Tyr-218 of FasL interacts with a cluster of three basic amino acid side chains of Fas. Interestingly, mutant Y218F could induce apoptosis in murine, but not human cells.


Subject(s)
Membrane Glycoproteins/metabolism , fas Receptor/metabolism , Amino Acid Sequence , Animals , Binding Sites , Chromatography, Gel , Enzyme-Linked Immunosorbent Assay , Fas Ligand Protein , Glycosylation , Humans , Jurkat Cells , Ligands , Membrane Glycoproteins/genetics , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis , Proline/metabolism , Sequence Alignment , Solubility , Species Specificity , Tyrosine/metabolism
19.
Immunity ; 7(6): 831-6, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9430228

ABSTRACT

TRAIL induces apoptosis through two closely related receptors, TRAIL-R1 (DR4) and TRAIL-R2 (DR5). Here we show that TRAIL-R1 can associate with TRAIL-R2, suggesting that TRAIL may signal through heteroreceptor signaling complexes. Both TRAIL receptors bind the adaptor molecules FADD and TRADD, and both death signals are interrupted by a dominant negative form of FADD and by the FLICE-inhibitory protein FLIP. The recruitment of TRADD may explain the potent activation of NF-kappaB observed by TRAIL receptors. Thus, TRAIL receptors can signal both death and gene transcription, functions reminiscent of those of TNFR1 and TRAMP, two other members of the death receptor family.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , Carrier Proteins/metabolism , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Cell Line , Cytoplasm/metabolism , Fas-Associated Death Domain Protein , Humans , Jurkat Cells , Receptors, TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...