Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 192
Filter
2.
Cell Death Differ ; 22(1): 58-73, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25236395

ABSTRACT

Cells exposed to extreme physicochemical or mechanical stimuli die in an uncontrollable manner, as a result of their immediate structural breakdown. Such an unavoidable variant of cellular demise is generally referred to as 'accidental cell death' (ACD). In most settings, however, cell death is initiated by a genetically encoded apparatus, correlating with the fact that its course can be altered by pharmacologic or genetic interventions. 'Regulated cell death' (RCD) can occur as part of physiologic programs or can be activated once adaptive responses to perturbations of the extracellular or intracellular microenvironment fail. The biochemical phenomena that accompany RCD may be harnessed to classify it into a few subtypes, which often (but not always) exhibit stereotyped morphologic features. Nonetheless, efficiently inhibiting the processes that are commonly thought to cause RCD, such as the activation of executioner caspases in the course of apoptosis, does not exert true cytoprotective effects in the mammalian system, but simply alters the kinetics of cellular demise as it shifts its morphologic and biochemical correlates. Conversely, bona fide cytoprotection can be achieved by inhibiting the transduction of lethal signals in the early phases of the process, when adaptive responses are still operational. Thus, the mechanisms that truly execute RCD may be less understood, less inhibitable and perhaps more homogeneous than previously thought. Here, the Nomenclature Committee on Cell Death formulates a set of recommendations to help scientists and researchers to discriminate between essential and accessory aspects of cell death.


Subject(s)
Apoptosis , Signal Transduction , Animals , Humans , Terminology as Topic
3.
Cell Death Differ ; 17(1): 14-24, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19893571

ABSTRACT

The study of death receptor (DR) signaling has led to the discovery of new signaling paradigms, including the first example of direct receptor-mediated activation of a protease (caspase-8) that functions as a second messenger to initiate a 'death cascade' of downstream protease activation. More recently, this receptor system has underscored the importance of ubiquitin modification in NF-kappaB activation. Both degradative lysine 48-linked polyubiquitin and scaffolding lysine 63-linked polyubiquitin have an essential role in signal propagation. Remarkably, a negative feedback process, termed ubiquitin editing, regulates signaling that emanates from certain DRs. Ubiquitin editing is mediated by a complex interplay between the ubiquitination and deubiquitination machinery, resulting in the replacement of signal enhancing lysine 63-linked polyubiquitin with signal extinguishing lysine 48-linked polyubiquitin. The ubiquitination machinery and its regulation in the context of DR signaling are discussed herein.


Subject(s)
Receptors, Death Domain/metabolism , Ubiquitin/metabolism , CARD Signaling Adaptor Proteins/metabolism , Caspase 8/metabolism , Fas-Associated Death Domain Protein/metabolism , Signal Transduction , TNF Receptor-Associated Death Domain Protein/metabolism , Ubiquitination
5.
J Cell Sci ; 114(Pt 20): 3771-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11707529

ABSTRACT

Poly(ADP-ribosyl)ation is an important mechanism for the maintenance of genomic integrity in response to DNA damage. The enzyme responsible for poly(ADP-ribose) synthesis, poly(ADP-ribose) polymerase 1 (PARP-1), has been implicated in two distinct modes of cell death induced by DNA damage, namely apoptosis and necrosis. During the execution phase of apoptosis, PARP-1 is specifically proteolyzed by caspases to produce an N-terminal DNA-binding domain (DBD) and a C-terminal catalytic fragment. The functional consequence of this proteolytic event is not known. However, it has recently been shown that overactivation of full-length PARP-1 can result in energy depletion and necrosis in dying cells. Here, we investigate the molecular basis for the differential involvement of PARP-1 in these two types of cellular demise. We show that the C-terminal apoptotic fragment of PARP-1 loses its DNA-dependent catalytic activity upon cleavage with caspase 3. However, the N-terminal apoptotic fragment, retains a strong DNA-binding activity and totally inhibits the catalytic activity of uncleaved PARP-1. This dominant-negative behavior was confirmed and extended in cellular extracts where DNA repair was completely inhibited by nanomolar concentrations of the N-terminal fragment. Furthermore, overexpression of the apoptotic DBD in mouse fibroblast inhibits endogenous PARP-1 activity very efficiently in vivo, thereby confirming our biochemical observations. Taken together, these experiments indicate that the apoptotic DBD of PARP-1 acts cooperatively with the proteolytic inactivation of the enzyme to trans-inhibit NAD hydrolysis and to maintain the energy levels of the cell. These results are consistent with a model in which cleavage of PARP-1 promotes apoptosis by preventing DNA repair-induced survival and by blocking energy depletion-induced necrosis.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Peptide Fragments/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Caspase 3 , Cell Line , DNA/metabolism , DNA Damage , DNA Repair/physiology , Dexamethasone/pharmacology , Humans , Mice
6.
J Exp Med ; 194(10): 1441-8, 2001 Nov 19.
Article in English | MEDLINE | ID: mdl-11714751

ABSTRACT

During an immune response naive T helper (Th) cells differentiate into two functionally distinct subsets, Th1 and Th2, based on their cytokine secretion profile and immunomodulatory function. c-Jun amino terminal kinase (JNK) regulates Th cell differentiation by activating a transcriptional program required for cytokine production. We have recently identified a TNFR superfamily death domain-containing molecule, death receptor (DR)6, which potently activates JNK. T cells from DR6-deficient mice are substantially impaired in JNK activation. When DR6(-/-) mice were challenged with protein antigen, their T cells hyperproliferate and display a profound polarization toward a Th2 response whereas Th1 differentiation is not equivalently affected. In addition, DR6(-/)- T cells showed preference toward Th2 differentiation in vitro. The phenotype seen in the DR6(-/)- mice is not due to the apoptotic pathway. Therefore, DR6, working through JNK, rather than apoptosis, functions to attenuate the Th2 response. This is the first demonstration of a role in the activation and differentiation of Th cells by DR6 in particular and DRs in general.


Subject(s)
Mitogen-Activated Protein Kinases/physiology , Receptors, Tumor Necrosis Factor/physiology , T-Lymphocytes/physiology , Amino Acid Sequence , Animals , Apoptosis , Cell Differentiation , Cloning, Molecular , Female , Immunoglobulin E/biosynthesis , Immunoglobulin G/biosynthesis , JNK Mitogen-Activated Protein Kinases , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Receptors, Tumor Necrosis Factor/deficiency
7.
Curr Biol ; 11(19): 1547-52, 2001 Oct 02.
Article in English | MEDLINE | ID: mdl-11591325

ABSTRACT

BLyS (also called BAFF, TALL-1, THANK, and zTNF4), a TNF superfamily member, binds two receptors, TACI and BCMA, and regulates humoral immune responses [1-7]. These two receptors also bind APRIL [7-10], another TNF superfamily member. The results from TACI(-/-) and BCMA(-/-) mice suggest the existence of additional receptor(s) for BLyS. The TACI knockout gives the paradoxical result of B cells being hyperresponsive, suggesting an inhibitory role for this receptor [11, 12], while BCMA null mice have no discernable phenotype [13]. Here we report the identification of a third BLyS receptor (BR3; BLyS receptor 3). This receptor is unique in that, in contrast to TACI and BCMA, BR3 only binds BLyS. Treatment of antigen-challenged mice with BR3-Fc inhibited antibody production, indicating an essential role for BLyS, but not APRIL, in this response. A critical role for BR3 in B cell ontogeny is underscored by our data showing that the BR3 gene had been inactivated by a discrete, approximately 4.7 kb gene insertion event that disrupted the 3' end of the BR3 gene in A/WySnJ mice, which lack peripheral B cells.


Subject(s)
B-Lymphocytes/physiology , Membrane Proteins/metabolism , Mutagenesis , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Sequence , Animals , B-Cell Activating Factor , B-Cell Activation Factor Receptor , B-Cell Maturation Antigen , B-Lymphocytes/metabolism , Base Sequence , COS Cells , Chlorocebus aethiops , DNA, Complementary , Humans , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Receptors, Tumor Necrosis Factor/classification , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spleen/metabolism , Transmembrane Activator and CAML Interactor Protein , Tumor Necrosis Factor-alpha/genetics
8.
Protein Sci ; 10(9): 1911-8, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11514682

ABSTRACT

PYRIN domains were identified recently as putative protein-protein interaction domains at the N-termini of several proteins thought to function in apoptotic and inflammatory signaling pathways. The approximately 95 residue PYRIN domains have no statistically significant sequence homology to proteins with known three-dimensional structure. Using secondary structure prediction and potential-based fold recognition methods, however, the PYRIN domain is predicted to be a member of the six-helix bundle death domain-fold superfamily that includes death domains (DDs), death effector domains (DEDs), and caspase recruitment domains (CARDs). Members of the death domain-fold superfamily are well established mediators of protein-protein interactions found in many proteins involved in apoptosis and inflammation, indicating further that the PYRIN domains serve a similar function. An homology model of the PYRIN domain of CARD7/DEFCAP/NAC/NALP1, a member of the Apaf-1/Ced-4 family of proteins, was constructed using the three-dimensional structures of the FADD and p75 neurotrophin receptor DDs, and of the Apaf-1 and caspase-9 CARDs, as templates. Validation of the model using a variety of computational techniques indicates that the fold prediction is consistent with the sequence. Comparison of a circular dichroism spectrum of the PYRIN domain of CARD7/DEFCAP/NAC/NALP1 with spectra of several proteins known to adopt the death domain-fold provides experimental support for the structure prediction.


Subject(s)
Apoptosis , Proteins/chemistry , Amino Acid Sequence , Circular Dichroism , Cytoskeletal Proteins , Models, Molecular , Molecular Sequence Data , Protein Folding , Protein Structure, Tertiary , Proteins/metabolism , Pyrin , Sequence Alignment , Sequence Homology, Amino Acid
9.
Nat Immunol ; 2(7): 632-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11429548

ABSTRACT

Interactions of the tumor necrosis factor superfamily members B lymphocyte stimulator (BLyS) and a proliferation-inducing ligand (APRIL) with their receptors-transmembrane activator and CAML interactor (TACI) and B cell maturation molecule (BCMA)-on B cells play an important role in the humoral immune response. Whereas BCMA is restricted to B cells, TACI is also expressed on activated T cells; we show here that TACI-Fc blocks the activation of T cells in vitro and inhibits antigen-specific T cell activation and priming in vivo. In a mouse model for rheumatoid arthritis (RA), an autoimmune disease that involves both B and T cell components, TACI-Fc treatment substantially inhibited inflammation, bone and cartilage destruction and disease development. Thus, BLyS and/or APRIL are important not only for B cell function but for T cell-mediated immune responses. Inhibition of these ligands might have therapeutic benefits for autoimmune diseases, such as RA, that involve both B and T cells.


Subject(s)
Arthritis, Rheumatoid/immunology , Lymphocyte Activation/immunology , Membrane Proteins , Receptors, Tumor Necrosis Factor/immunology , T-Lymphocytes/immunology , Animals , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/pathology , B-Lymphocytes/immunology , Collagen/adverse effects , Disease Models, Animal , Joints/pathology , Ligands , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/immunology , Transmembrane Activator and CAML Interactor Protein
10.
Nat Immunol ; 2(7): 638-43, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11429549

ABSTRACT

The tumor necrosis factor (TNF)-related ligand B lymphocyte stimulator (BLyS) binds two TNF receptor family members, transmembrane activator and calcium-modulating and cyclophilin ligand interactor (TACI) and B cell maturation molecule (BCMA). Mice that are transgenic for BLyS show B cell accumulation, activation and autoimmune lupus-like nephritis. The existence of at least two distinct BLyS receptors raises the question of the relative contribution of each to B cell functions. We therefore generated mice that were deficient in TACI. TACI-/- mice showed increased B cell accumulation and marked splenomegaly. Isolated TACI-/- B cells hyperproliferated and produced increased amounts of immunoglobulins in vitro. In vivo antigen challenge resulted in enhanced antigen-specific antibody production. Thus, TACI may play an unexpected inhibitory role in B cell activation that helps maintain immunological homeostasis.


Subject(s)
B-Lymphocytes/immunology , Lymphocyte Activation/immunology , Receptors, Tumor Necrosis Factor/immunology , Animals , Antigens/immunology , B-Cell Activating Factor , B-Lymphocytes/cytology , Cell Division , Cells, Cultured , Immunoglobulin M/biosynthesis , Membrane Proteins/immunology , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Tumor Necrosis Factor/genetics , Spleen/pathology , Transmembrane Activator and CAML Interactor Protein , Tumor Necrosis Factor-alpha/immunology , gamma-Globulins/immunology
11.
Science ; 291(5507): 1279-84, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11181990

ABSTRACT

A comparison of the proteins encoded in the recently (nearly) completed human genome to those from the fly and nematode genomes reveals a major increase in the complexity of the apoptotic molecular machinery in vertebrates, in terms of both the number of proteins involved and their domain architecture. Several components of the apoptotic system are shared by humans and flies, to the exclusion of nematodes, which seems to support the existence of a coelomate clade in animal evolution. A considerable repertoire of apoptotic protein domains was detected in Actinomycetes and Cyanobacteria, which suggests a major contribution of horizontal gene transfer to the early evolution of apoptosis.


Subject(s)
Apoptosis , Evolution, Molecular , Genome, Human , Genome , Proteins/chemistry , Proteins/genetics , Amino Acid Motifs , Animals , Apoptosis/genetics , Caenorhabditis elegans/genetics , Conserved Sequence , Drosophila melanogaster/genetics , Helminth Proteins/chemistry , Helminth Proteins/genetics , Helminth Proteins/physiology , Humans , Insect Proteins/chemistry , Insect Proteins/genetics , Insect Proteins/physiology , Protein Structure, Tertiary , Proteins/physiology , Sequence Alignment , Sequence Analysis, DNA , Species Specificity
12.
Mol Cell ; 6(4): 961-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11090634

ABSTRACT

Caspases are cysteine proteases essential to apoptosis. We have identified two families of caspase-like proteins, Paracaspases (found in metazoans and Dictyostelium) and metacaspases (found in plants, fungi, and protozoa). Metazoan paracaspase prodomains contain a death domain and immunoglobulin domains. Several plant metacaspase prodomains contain zinc finger motifs resembling those in the plant hypersensitive response/cell death protein Isd-1. The human paracaspase prodomain binds Bcl10, a protein involved in the t(1;14)(p22;q32) translocation of mucosa-associated lymphoid tissue (MALT) lymphoma. Another MALT lymphoma translocation, t(11;18)(q21;q21), fuses the IAP-2 gene to the MLT1/MALT1 locus, which encodes the human paracaspase. We find that this fusion activates NF-kappaB and that the caspase domain is required for this function, since mutation of the conserved catalytic cysteine attenuates NF-kappaB activation.


Subject(s)
Caspases/genetics , Lymphoma, B-Cell, Marginal Zone/enzymology , Lymphoma, B-Cell, Marginal Zone/genetics , Amino Acid Sequence , Animals , Caspases/chemistry , Chromosome Mapping , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 14 , Cloning, Molecular , Dictyostelium/enzymology , Dictyostelium/genetics , Humans , Molecular Sequence Data , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Transfection , Translocation, Genetic , Zinc Fingers
13.
Curr Biol ; 10(21): 1359-66, 2000 Nov 02.
Article in English | MEDLINE | ID: mdl-11084335

ABSTRACT

BACKGROUND: Inhibitors of apoptosis (IAPs) are a family of cell death inhibitors found in viruses and metazoans. All IAPs have at least one baculovirus IAP repeat (BIR) motif that is essential for their anti-apoptotic activity. IAPs physically interact with a variety of pro-apoptotic proteins and inhibit apoptosis induced by diverse stimuli. This allows them to function as sensors and inhibitors of death signals that emanate from a variety of pathways. RESULTS: Here we report the characterization of ML-IAP, a novel human IAP that contains a single BIR and RING finger motif. ML-IAP is a powerful inhibitor of apoptosis induced by death receptors and chemotherapeutic agents, probably functioning as a direct inhibitor of downstream effector caspases. Modeling studies of the structure of the BIR domain revealed it to closely resemble the fold determined for the BIR2 domain of X-IAP. Deletion and mutational analysis demonstrated that integrity of the BIR domain was required for anti-apoptotic function. Tissue survey analysis showed expression in a number of embryonic tissues and tumor cell lines. In particular, the majority of melanoma cell lines expressed high levels of ML-IAP in contrast to primary melanocytes, which expressed undetectable levels. These melanoma cells were significantly more resistant to drug-induced apoptosis. CONCLUSIONS: ML-IAP, a novel human IAP, inhibits apoptosis induced by death receptors and chemotherapeutic agents. The BIR of ML-IAP possesses an evolutionarily conserved fold that is necessary for anti-apoptotic activity. Elevated expression of ML-IAP renders melanoma cells resistant to apoptotic stimuli and thereby potentially contributes to the pathogenesis of this malignancy.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , Carrier Proteins/metabolism , Melanoma/metabolism , Neoplasm Proteins/metabolism , Adult , Amino Acid Motifs , Amino Acid Sequence , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carrier Proteins/chemistry , Carrier Proteins/genetics , Caspase Inhibitors , Caspases/genetics , Caspases/metabolism , Cell Line , Doxorubicin/pharmacology , Genes, Reporter , Humans , Inhibitor of Apoptosis Proteins , Melanocytes/metabolism , Melanoma/genetics , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Protein Conformation , Protein Structure, Tertiary , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Tumor Cells, Cultured , fas Receptor/metabolism
14.
Cell ; 103(1): 99-111, 2000 Sep 29.
Article in English | MEDLINE | ID: mdl-11051551

ABSTRACT

ProIL-1beta is a proinflammatory cytokine that is proteolytically processed to its active form by caspase-1. Upon receipt of a proinflammatory stimulus, an upstream adaptor, RIP2, binds and oligomerizes caspase-1 zymogen, promoting its autoactivation. ICEBERG is a novel protein that inhibits generation of IL-1beta by interacting with caspase-1 and preventing its association with RIP2. ICEBERG is induced by proinflammatory stimuli, suggesting that it may be part of a negative feedback loop. Consistent with this, enforced retroviral expression of ICEBERG inhibits lipopolysaccharide-induced IL-1beta generation. The structure of ICEBERG reveals it to be a member of the death-domain-fold superfamily. The distribution of surface charge is complementary to the homologous prodomain of caspase-1, suggesting that charge-charge interactions mediate binding of ICEBERG to the prodomain of caspase-1.


Subject(s)
Amino Acid Sequence/physiology , Carrier Proteins/genetics , Caspase 1/metabolism , Inflammation Mediators/metabolism , Interleukin-1/antagonists & inhibitors , Interleukin-1/biosynthesis , Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases/metabolism , Caspase 1/chemistry , Cells, Cultured , Molecular Sequence Data , Monomeric GTP-Binding Proteins/metabolism , Monomeric GTP-Binding Proteins/pharmacology , Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases , Sequence Analysis, Protein , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , ran GTP-Binding Protein
15.
Science ; 290(5491): 523-7, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-11039935

ABSTRACT

Ectodysplasin, a member of the tumor necrosis factor family, is encoded by the anhidrotic ectodermal dysplasia (EDA) gene. Mutations in EDA give rise to a clinical syndrome characterized by loss of hair, sweat glands, and teeth. EDA-A1 and EDA-A2 are two isoforms of ectodysplasin that differ only by an insertion of two amino acids. This insertion functions to determine receptor binding specificity, such that EDA-A1 binds only the receptor EDAR, whereas EDA-A2 binds only the related, but distinct, X-linked ectodysplasin-A2 receptor (XEDAR). In situ binding and organ culture studies indicate that EDA-A1 and EDA-A2 are differentially expressed and play a role in epidermal morphogenesis.


Subject(s)
Epidermis/metabolism , I-kappa B Proteins , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Cell Line , DNA-Binding Proteins/metabolism , Ectodermal Dysplasia/genetics , Ectodysplasins , Epidermis/embryology , Humans , In Situ Hybridization , Ligands , Mice , Models, Molecular , Molecular Sequence Data , Morphogenesis , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Phosphorylation , Point Mutation , Protein Conformation , Proteins/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6 , Transfection
16.
Annu Rev Biochem ; 69: 217-45, 2000.
Article in English | MEDLINE | ID: mdl-10966458

ABSTRACT

Apoptosis, a physiological process for killing cells, is critical for the normal development and function of multicellular organisms. Abnormalities in cell death control can contribute to a variety of diseases, including cancer, autoimmunity, and degenerative disorders. Signaling for apoptosis occurs through multiple independent pathways that are initiated either from triggering events within the cell or from outside the cell, for instance, by ligation of death receptors. All apoptosis signaling pathways converge on a common machinery of cell destruction that is activated by a family of cysteine proteases (caspases) that cleave proteins at aspartate residues. Dismantling and removal of doomed cells is accomplished by proteolysis of vital cellular constituents, DNA degradation, and phagocytosis by neighboring cells. This article reviews current knowledge of apoptosis signaling, lists several pressing questions, and presents a novel model to explain the biochemical and functional interactions between components of the cell death regulatory machinery.


Subject(s)
Apoptosis/physiology , Signal Transduction/physiology , Animals , Caspases/physiology , Humans , Models, Biological , Neoplasms/pathology , Neoplasms/physiopathology , Proto-Oncogene Proteins c-bcl-2/physiology , Receptors, Tumor Necrosis Factor/physiology
17.
Curr Biol ; 10(13): 785-8, 2000 Jun 29.
Article in English | MEDLINE | ID: mdl-10898980

ABSTRACT

BLyS (also called TALL-1, THANK, or BAFF) [1] [2] [3] [4] is a member of the tumor necrosis factor (TNF) gene family that stimulates proliferation and immunoglobulin production by B cells. BLyS interacts with the TNF receptor (TNFR) homologue TACI (transmembrane activator and CAML-interactor) [5], and treatment of mice with a TACI-Fc fusion protein abolishes germinal center formation after antigenic challenge [6]. Here we report a novel interaction between BLyS and another TNFR homologue, BCMA (B cell maturation antigen) [7] [8]. Further, the TNF homologue APRIL [9], a close relative of BLyS, also bound to BCMA and TACI. BLyS or APRIL activated nuclear factor-kappaB (NF-kappaB) through TACI and BCMA, and each ligand stimulated immunoglobulin M (IgM) production by peripheral blood B cells. These results define a dual ligand-receptor system that may play an important role in humoral immunity.


Subject(s)
Membrane Proteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Sequence , Animals , B-Cell Activating Factor , B-Cell Maturation Antigen , CHO Cells , Cell Line , Cricetinae , Humans , Immunoglobulin M/metabolism , Leukocytes/cytology , Leukocytes/drug effects , Leukocytes/immunology , Membrane Proteins/genetics , Molecular Sequence Data , NF-kappa B/metabolism , Protein Binding , Receptors, Tumor Necrosis Factor/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Sequence Homology, Amino Acid , Transmembrane Activator and CAML Interactor Protein , Tumor Necrosis Factor Ligand Superfamily Member 13 , Tumor Necrosis Factor-alpha/genetics
18.
Cancer Res ; 60(4): 847-53, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10706092

ABSTRACT

The intractability of malignant gliomas to multimodality treatments plays a large part in their extremely poor prognosis. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a novel member of the tumor necrosis factor (TNF) family that induces apoptosis preferentially in tumor cells through binding to its cognate death receptors, DR4 and DR5. Here we show that the DNA-damaging chemotherapeutic drugs, cis-diamminedichloroplatinum(II) (CDDP) and etoposide, elicited increased expression of DR5 in human glioma cells. Exposure of such cells in vitro to soluble human TRAIL in combination with CDDP or etoposide resulted in synergistic cell death that could be blocked by soluble TRAIL-neutralizing DR5-Fc or the caspase inhibitors, Z-Asp-CH2-DCB and CrmA. Moreover, systemic in vivo administration of TRAIL with CDDP synergistically suppressed both tumor formation and growth of established s.c. human glioblastoma xenografts in nude mice by inducing apoptosis without causing significant general toxicity. The combination treatment resulted in complete and durable remission in 29% of mice with the established s.c. xenografts and also significantly extended the survival of mice bearing intracerebral xenografts. These results provide preclinical proof-of-principle for a novel therapeutic strategy in which the death ligand, TRAIL, is safely combined with conventional DNA-damaging chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Glioma/drug therapy , Membrane Glycoproteins/pharmacology , Receptors, Tumor Necrosis Factor/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis Regulatory Proteins , Cisplatin/pharmacology , DNA/drug effects , DNA Damage , Drug Synergism , Female , Glioma/metabolism , Glioma/pathology , Humans , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Receptors, TNF-Related Apoptosis-Inducing Ligand , TNF-Related Apoptosis-Inducing Ligand , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Suppressor Protein p53/physiology
19.
J Exp Med ; 191(3): 463-74, 2000 Feb 07.
Article in English | MEDLINE | ID: mdl-10662792

ABSTRACT

Initiation of T cell antigen receptor (TCR) signaling is dependent on Lck, a Src family kinase. The Src-like adaptor protein (SLAP) contains Src homology (SH)3 and SH2 domains, which are highly homologous to those of Lck and other Src family members. Because of the structural similarity between Lck and SLAP, we studied its potential role in TCR signaling. Here, we show that SLAP is expressed in T cells, and that when expressed in Jurkat T cells it can specifically inhibit TCR signaling leading to nuclear factor of activated T cells (NFAT)-, activator protein 1 (AP-1)-, and interleukin 2-dependent transcription. The SH3 and SH2 domains of SLAP are required for maximal attenuation of TCR signaling. This inhibitory activity can be bypassed by the combination of phorbol myristate acetate (PMA) and ionomycin, suggesting that SLAP acts proximally in the TCR signaling pathway. SLAP colocalizes with endosomes in Jurkat and in HeLa cells, and is insoluble in mild detergents. In stimulated Jurkat cells, SLAP associates with a molecular signaling complex containing CD3zeta, ZAP-70, SH2 domain-containing leukocyte protein of 76 kD (SLP-76), Vav, and possibly linker for activation of T cells (LAT). These results suggest that SLAP is a negative regulator of TCR signaling.


Subject(s)
Adaptor Proteins, Signal Transducing , Proto-Oncogene Proteins pp60(c-src)/pharmacology , Receptors, Antigen, T-Cell/drug effects , 3T3 Cells , Animals , Calcium/metabolism , Cell Line , Detergents , Down-Regulation , HeLa Cells , Humans , Jurkat Cells , Mice , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , RNA, Messenger/metabolism , Signal Transduction/drug effects , Subcellular Fractions/metabolism , Transcription, Genetic/drug effects , Transfection , src Homology Domains
20.
Nat Immunol ; 1(1): 37-41, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10881172

ABSTRACT

B lymphocyte stimulator (BLyS) is a member of the tumor necrosis factor (TNF) superfamily. BLyS stimulates proliferation of, and immunoglobulin production by, B cells. However, the relative importance of BLyS in physiological B cell activation is unclear. We identified a B cell receptor for BLyS through expression cloning as TACI, an orphan TNF receptor homologue of unknown function. Binding of BLyS to TACI activated signaling by nuclear factor-kappa B (NF-kappa B). In vitro soluble TACI-Fc fusion protein blocked BLyS-induced NF-kappa B activation in B lymphoma cells and IgM production in peripheral blood B cells. In vivo treatment of immunized mice with TACI-Fc inhibited production of antigen-specific IgM and IgGI antibodies and abolished splenic germinal center (GC) formation. Thus, BLyS activity must play a critical role in the humoral immune response.


Subject(s)
Antibody Formation/immunology , Membrane Proteins/immunology , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , B-Cell Activating Factor , COS Cells , Cloning, Molecular , Humans , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Tumor Necrosis Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...