Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Leukemia ; 23(1): 170-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18843286

ABSTRACT

This study explores whether lymphoma cell adhesion-induced B cell-activating factor (BAFF) expression in bone marrow stromal cells (BMSCs) protects B lymphoma cells from apoptosis. We first showed protection of lymphoma cells from apoptosis by conditioned medium of a stromal cell-lymphoma cell coculture, either spontaneous or induced by mitoxantrone, implying a role for soluble factor(s) in lymphoma cell survival. Addition of BAFF counteracted mitoxantrone-induced apoptosis and elicited a reduction in spontaneous apoptosis in primary lymphomas, suggesting a role of BAFF in sustaining B-cell survival. Abundant BAFF was detected in the BMSC cell line (HS-5) and primary BMSCs by flow cytometry, RT-PCR and immunoblotting. BAFF levels were 20- to 200-fold higher in BMSCs than in lymphoma cells, and lymphoma cell adhesion to BMSCs augmented BAFF secretion twofold through upregulation of BAFF gene expression. Finally, neutralization of BAFF by TACI-Ig or depletion of BAFF by small hairpin RNA (shRNA) in BMSCs significantly enhanced lymphoma cell response to chemotherapy and overcame stroma-mediated drug resistance, suggesting that lymphoma cells use BMSC-derived BAFF as a survival factor. These findings support the hypothesis that lymphoma cells interact with BMSCs, resulting in stromal niches with high BAFF concentration, and identify BMSC-derived BAFF as a functional determinant for B lymphoma cell survival in the bone marrow environment.


Subject(s)
B-Cell Activating Factor/genetics , Cell Adhesion , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/pathology , Stromal Cells/cytology , Apoptosis , Bone Marrow Cells , Cell Communication , Cell Survival , Coculture Techniques , Humans , Up-Regulation
2.
Cell Mol Life Sci ; 64(5): 632-40, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17310276

ABSTRACT

Polo-like kinase 1 (Plk1) is a highly conserved serine/threonine kinase that plays critical roles in many cell cycle events, especially in mitosis. In the present study, we identified TTDN1 as a potential interacting partner of Plk1 in yeast two-hybrid screens. Sequence analysis indicates that TTDN1 contains a consensus Plk1-binding motif at its C terminus. TTDN1 colocalizes with Plk1 at the centrosome in mitosis and the midbody during cytokinesis. TTDN1 is phosphorylated by Cdk1 in mitosis, and this is required for its interaction with Plk1. Site-directed mutagenesis indicates that TTDN1 is phosphorylated at multiple residues, including Ser93 and Ser104. Mutation of Thr120 of TTDN1 abolishes its interaction with Plk1, suggesting phosphorylation of Thr120 in the consensus Plk1-binding motif is required for its interaction with Plk1. Overexpression of TTDN1 or its knockdown by siRNA causes multi-polar spindles and multiple nuclei, suggesting that TTDN1 plays a role in regulating mitosis and cytokinesis.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Binding Sites , Cell Division , HeLa Cells , Humans , Molecular Sequence Data , Phosphorylation , Polo-Like Kinase 1
3.
Oncogene ; 25(6): 849-56, 2006 Feb 09.
Article in English | MEDLINE | ID: mdl-16186796

ABSTRACT

AMID is an apoptosis-inducing factor (AIF)-homologous and mitochondria-associated protein that has been implicated in caspase-independent apoptosis. Transcription of human AMID gene is upregulated by p53 and downregulated in tumors in comparison to their matched normal tissues, suggesting the possibility that AMID is involved in the downstream effects of p53. To investigate the physiological functions of AMID, we generated AMID-deficient mice by gene targeting. AMID-deficient mice are viable and fertile, develop normally and lack obvious phenotypic changes compared to wild-type mice up to 1 year old. AMID(-/-) mice up to 1 year old have no spontaneous tumors and show similar fibrosarcoma incidence after MCA inoculation compared to wild-type mice. AMID(-/-) embryonic fibroblasts exhibit normal proliferation but slightly increased resistance to genotoxin-induced growth arrest. These findings suggest that AMID is not required for normal development and p53-mediated tumor suppression.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Gene Expression Regulation, Developmental , Genes, Tumor Suppressor , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Animals , Apoptosis/drug effects , Apoptosis/physiology , Base Sequence , Cell Proliferation/drug effects , Cells, Cultured , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Male , Mice , Mice, Mutant Strains , Molecular Sequence Data , Mutagenicity Tests , Neoplasms/genetics , Reference Values , Sequence Homology, Amino Acid , Tumor Suppressor Protein p53/genetics
4.
Immunity ; 12(6): 633-42, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10894163

ABSTRACT

Casper (c-FLIP) associates with FADD and caspase-8 in signaling complexes downstream of death receptors like Fas. We generated Casper-deficient mice and cells and noted a duality in the physiological functions of this molecule. casper-/- embryos do not survive past day 10.5 of embryogenesis and exhibit impaired heart development. This phenotype is reminiscent of that reported for FADD-/- and caspase-8-/- embryos. However, unlike FADD-/- and caspase-8-/- cells, casper-/- embryonic fibroblasts are highly sensitive to FasL- or TNF-induced apoptosis and show rapid induction of caspase activities. NF-kappaB and JNK/SAPK activation is intact in TNF-stimulated casper-/- cells. These results suggest that Casper has two distinct roles: to cooperate with FADD and caspase-8 during embryonic development and to mediate cytoprotection against death factor-induced apoptosis.


Subject(s)
Apoptosis/immunology , Carrier Proteins/physiology , Embryonic and Fetal Development/immunology , Intracellular Signaling Peptides and Proteins , Receptors, Tumor Necrosis Factor/physiology , Animals , Apoptosis/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/genetics , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Line , Embryonic and Fetal Development/genetics , Enzyme Activation/immunology , Female , Heart/embryology , JNK Mitogen-Activated Protein Kinases , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Myocardium/enzymology , Myocardium/immunology , Myocardium/metabolism , Myocardium/pathology , NF-kappa B/metabolism , Stem Cells/enzymology , Stem Cells/immunology , Stem Cells/metabolism , Stem Cells/pathology , Tumor Necrosis Factor-alpha/physiology
5.
Proc Natl Acad Sci U S A ; 97(16): 9156-61, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10908663

ABSTRACT

TALL-1 is a recently identified member of the tumor necrosis factor (TNF) family that costimulates B lymphocyte proliferation. Here we show that B cell maturation protein (BCMA), a member of the TNF receptor family that is expressed only by B lymphocytes, specifically binds to TALL-1. A soluble receptor containing the extracellular domain of BCMA blocks the binding of TALL-1 to its receptor on the plasma membrane and inhibits TALL-1-triggered B lymphocyte costimulation. Overexpression of BCMA activates NF-kappaB, and this activation is potentiated by TALL-1. Moreover, BCMA-mediated NF-kappaB activation is inhibited by dominant negative mutants of TNF receptor-associated factor 5 (TRAF5), TRAF6, NF-kappaB-inducing kinase (NIK), and IkappaB kinase (IKK). These data indicate that BCMA is a receptor for TALL-1 and BCMA activates NF-kappaB through a TRAF5-, TRAF6-, NIK-, and IKK-dependent pathway. The identification of BCMA as a NF-kappaB-activating receptor for TALL-1 suggests molecular targets for drug development against certain immunodeficient or autoimmune diseases.


Subject(s)
B-Lymphocytes/metabolism , Membrane Proteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , B-Cell Activating Factor , B-Cell Maturation Antigen , B-Lymphocytes/cytology , Base Sequence , Cell Division/physiology , Cell Line , Cell Membrane/metabolism , DNA Primers , Humans , Ligands , Precipitin Tests , Receptors, Tumor Necrosis Factor/physiology
6.
J Biol Chem ; 275(15): 10838-44, 2000 Apr 14.
Article in English | MEDLINE | ID: mdl-10753878

ABSTRACT

Fas-associated death domain protein (FADD), caspase-8-related protein (Casper), and caspase-8 are components of the tumor necrosis factor receptor type 1 (TNF-R1) and Fas signaling complexes that are involved in TNF-R1- and Fas-induced apoptosis. Here we show that overexpression of FADD and Casper potently activates NF-kappaB. In the presence of caspase inhibitors, overexpression of caspase-8 also activates NF-kappaB. A caspase-inactive point mutant, caspase-8(C360S), activates NF-kappaB as potently as wild-type caspase-8, suggesting that caspase-8-induced apoptosis and NF-kappaB activation are uncoupled. NF-kappaB activation by FADD and Casper is inhibited by the caspase-specific inhibitors crmA and BD-fmk, suggesting that FADD- and Casper-induced NF-kappaB activation is mediated by caspase-8. FADD, Casper, and caspase-8-induced NF-kappaB activation are inhibited by dominant negative mutants of TRAF2, NIK, IkappaB kinase alpha, and IkappaB kinase beta. A dominant negative mutant of RIP inhibits FADD- and caspase-8-induced but not Casper-induced NF-kappaB activation. A mutant of Casper and the caspase-specific inhibitors crmA and BD-fmk partially inhibit TNF-R1-, TRADD, and TNF-induced NF-kappaB activation, suggesting that FADD, Casper, and caspase-8 function downstream of TRADD and contribute to TNF-R1-induced NF-kappaB activation. Moreover, activation of caspase-8 results in proteolytic processing of NIK, which is inhibited by crmA. When overexpressed, the processed fragments of NIK do not activate NF-kappaB, and the processed C-terminal fragment inhibits TNF-R1-induced NF-kappaB activation. These data indicate that FADD, Casper, and pro-caspase-8 are parts of the TNF-R1-induced NF-kappaB activation pathways, whereas activated caspase-8 can negatively regulate TNF-R1-induced NF-kappaB activation by proteolytically inactivating NIK.


Subject(s)
Arabidopsis Proteins , Carrier Proteins/physiology , Caspases/physiology , Fatty Acid Desaturases/physiology , Intracellular Signaling Peptides and Proteins , NF-kappa B/metabolism , Antigens, CD/physiology , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspase 8 , Caspase 9 , Caspase Inhibitors , Humans , Protein Serine-Threonine Kinases/physiology , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type I , NF-kappaB-Inducing Kinase
7.
J Biol Chem ; 274(43): 30603-10, 1999 Oct 22.
Article in English | MEDLINE | ID: mdl-10521444

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF family that interacts with several receptors, including TRAIL-R1, TRAIL-R2, and TRAIL-R4. TRAIL-R1 and TRAIL-R2 can induce apoptosis of cancer cells and activate the transcription factor NF-kappaB. TRAIL-R4 can activate NF-kappaB and protect cells from TRAIL-induced apoptosis. Here we show that TRAIL-R1-, TRAIL-R2-, and TRAIL-R4-induced NF-kappaB activation are mediated by a TRAF2-NIK-IkappaB kinase alpha/beta signaling cascade but is MEKK1 independent. TRAIL receptors also activate the protein kinase JNK. JNK activation by TRAIL-R1 is mediated by a TRAF2-MEKK1-MKK4 but not the TRAF2-NIK/IkappaB kinase alpha/beta signaling pathway. We also show that activation of NF-kappaB or overexpression of TRAIL-R4 does not protect TRAIL-R1-induced apoptosis. Moreover, inhibition of NF-kappaB by IkappaBalpha sensitizes cells to tumor necrosis factor- but not TRAIL-induced apoptosis. These findings suggest that TRAIL receptors induce apoptosis, NF-kappaB and JNK activation through distinct signaling pathways, and activation of NF-kappaB is not sufficient for protecting cells from TRAIL-induced apoptosis.


Subject(s)
Apoptosis/physiology , MAP Kinase Kinase 4 , MAP Kinase Kinase Kinase 1 , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/physiology , Breast Neoplasms , Cell Line , Female , Genes, Reporter , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand , Recombinant Fusion Proteins/biosynthesis , Recombinant Proteins/metabolism , Transfection , Tumor Cells, Cultured
8.
J Leukoc Biol ; 65(5): 680-3, 1999 May.
Article in English | MEDLINE | ID: mdl-10331498

ABSTRACT

Members of the tumor necrosis factor (TNF) family play important roles in modulation of immune responses. We describe the identification and cloning of a novel TNF family member that has been designated as TALL-1. TALL-1 is a 285-amino acid type II transmembrane protein. Its carboxy terminus shares approximately 35% sequence identity with the recently identified APRIL and approximately 20-25% with TNF, FasL, TRAIL, and lymphotoxin-alpha, suggesting that TALL-1 and APRIL belong to a subfamily of the TNF family of ligands. Northern blot analysis suggests that TALL-1 is expressed abundantly in peripheral blood leukocytes and weakly in spleen but is barely detectable in all other tissues examined. Reverse transcriptase-polymerase chain reaction analysis indicates that TALL-1 is specifically expressed in monocytes and macrophages but is undetectable in T and B lymphocytes. Furthermore, TALL-1 expression is dramatically down-regulated by phorbol myristate acetate/ionomycin.


Subject(s)
Down-Regulation/immunology , Membrane Proteins/biosynthesis , Mitogens/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Amino Acid Sequence , Animals , B-Cell Activating Factor , Cloning, Molecular , Down-Regulation/drug effects , Humans , Ligands , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Rabbits , Sequence Homology, Amino Acid , Tumor Necrosis Factor Ligand Superfamily Member 13 , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/genetics
9.
Science ; 279(5358): 1954-8, 1998 Mar 20.
Article in English | MEDLINE | ID: mdl-9506948

ABSTRACT

FADD (also known as Mort-1) is a signal transducer downstream of cell death receptor CD95 (also called Fas). CD95, tumor necrosis factor receptor type 1 (TNFR-1), and death receptor 3 (DR3) did not induce apoptosis in FADD-deficient embryonic fibroblasts, whereas DR4, oncogenes E1A and c-myc, and chemotherapeutic agent adriamycin did. Mice with a deletion in the FADD gene did not survive beyond day 11.5 of embryogenesis; these mice showed signs of cardiac failure and abdominal hemorrhage. Chimeric embryos showing a high contribution of FADD null mutant cells to the heart reproduce the phenotype of FADD-deficient mutants. Thus, not only death receptors, but also receptors that couple to developmental programs, may use FADD for signaling.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , Carrier Proteins/physiology , Embryonic and Fetal Development , Heart/embryology , Animals , Carrier Proteins/genetics , Cell Transformation, Neoplastic , Cells, Cultured , Doxorubicin/pharmacology , Endothelium, Vascular/embryology , Fas-Associated Death Domain Protein , Female , Gene Expression , Gene Targeting , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Oncogenes , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/genetics , fas Receptor/physiology
10.
Immunity ; 6(6): 751-63, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9208847

ABSTRACT

Caspases are cysteine proteases that play a central role in apoptosis. Caspase-8 may be the first enzyme of the proteolytic cascade activated by the Fas ligand and tumor necrosis factor (TNF). Caspase-8 is recruited to Fas and TNF receptor-1 (TNF-R1) through interaction of its prodomain with the death effector domain (DED) of the receptor-associating FADD. Here we describe a novel 55 kDa protein, Casper, that has sequence similarity to caspase-8 throughout its length. However, Casper is not a caspase since it lacks several conserved amino acids found in all caspases. Casper interacts with FADD, caspase-8, caspase-3, TRAF1, and TRAF2 through distinct domains. When overexpressed in mammalian cells, Casper potently induces apoptosis. A C-terminal deletion mutant of Casper inhibits TNF- and Fas-induced cell death, suggesting that Casper is involved in these apoptotic pathways.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , Carrier Proteins/metabolism , Carrier Proteins/physiology , Caspases , Cysteine Endopeptidases/chemistry , Intracellular Signaling Peptides and Proteins , Tumor Necrosis Factor-alpha/physiology , Viral Proteins , fas Receptor/physiology , Amino Acid Sequence , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspase 3 , Caspase 8 , Caspase 9 , Cloning, Molecular , Cysteine Endopeptidases/metabolism , Endopeptidases/metabolism , Enzyme Induction , Fas-Associated Death Domain Protein , HeLa Cells , Humans , Molecular Sequence Data , Protein Binding , Protein Processing, Post-Translational , Proteins/metabolism , Recombinant Proteins , Sequence Alignment , Sequence Deletion , Sequence Homology, Amino Acid , Serpins/metabolism , Signal Transduction , Structure-Activity Relationship , TNF Receptor-Associated Factor 1 , TNF Receptor-Associated Factor 2
11.
Proc Natl Acad Sci U S A ; 93(24): 13973-8, 1996 Nov 26.
Article in English | MEDLINE | ID: mdl-8943045

ABSTRACT

The two cell surface receptors for tumor necrosis factor (TNF) interact with a number of intracellular signal transducing proteins. The association of TRADD, a 34-kDa cytoplasmic protein containing a C-terminal death domain, with aggregated TNF receptor 1 (TNF-R1) through their respective death domains leads to NF-kappa B activation and programmed cell death. In contrast, TNF receptor 2 (TNF-R2) interacts with the TNF receptor associated factors 2/1 (TRAF2/TRAF1) heterocomplex, which mediates the recruitment of two cellular inhibitor of apoptosis proteins (c-IAP1 and c-IAP2) to TNF-R2. Here we show that the TNF-R2 signal transducers TRAF2 and c-IAP1 are a part of the TNF-R1 signaling complex. The recruitment of TRAF2 and c-IAP1 to TNF-R1 is TNF-dependent, is mediated by TRADD, and is independent of TNF-R2. These data establish the physiological involvement of TRAF2 and c-IAP1 in TNF-R1 signaling and help provide a molecular explanation for both the overlapping and distinct signals generated by the two TNF receptors.


Subject(s)
Antigens, CD/physiology , Proteins/physiology , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/physiology , Apoptosis , Cell Line , Cysteine Proteinase Inhibitors/physiology , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins , Models, Biological , NF-kappa B/metabolism , Proteins/metabolism , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , TNF Receptor-Associated Factor 1 , TNF Receptor-Associated Factor 2 , Transfection , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitin-Protein Ligases
12.
Proc Natl Acad Sci U S A ; 93(16): 8241-6, 1996 Aug 06.
Article in English | MEDLINE | ID: mdl-8710854

ABSTRACT

Tumor necrosis factor (TNF) receptor-associated factor (TRAF) proteins associate with and transduce signals from TNF receptor 2, CD40, and presumably other members of the TNF receptor superfamily. TRAF2 is required for CD40- and TNF-mediated activation of the transcription factor NF-kappa B. Here we describe the isolation and characterization of a novel TRAF-interacting protein, I-TRAF, that binds to the conserved TRAF-C domain of the three known TRAFs. Overexpression of I-TRAF inhibits TRAF2-mediated NF-kappa B activation signaled by CD40 and both TNF receptors. Thus, I-TRAF appears as a natural regulator of TRAF function that may act by maintaining TRAFs in a latent state.


Subject(s)
Adaptor Proteins, Signal Transducing , Antigens, CD/metabolism , Bacterial Proteins/physiology , Carrier Proteins/physiology , Proteins/physiology , Receptors, Tumor Necrosis Factor/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cloning, Molecular , Humans , Macromolecular Substances , Mice , Molecular Sequence Data , NF-kappa B/metabolism , Peptides/chemistry , Protein Binding , Proteins/metabolism , Receptors, Tumor Necrosis Factor, Type II , Signal Transduction , TNF Receptor-Associated Factor 2 , X-Linked Inhibitor of Apoptosis Protein
13.
Immunity ; 4(4): 387-96, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8612133

ABSTRACT

The death domain of tumor necrosis factor (TNF) receptor-1 (TNFR1) triggers distinct signaling pathways leading to apoptosis and NF-kappa B activation through its interaction with the death domain protein TRADD. Here, we show that TRADD interacts strongly with RIP, another death domain protein that was shown previously to associate with Fas antigen. We also show that RIP is a serine-threonine kinase that is recruited by TRADD to TNFR1 in a TNF-dependent process. Overexpression of the intact RIP protein induces both NF-kappa B activation and apoptosis. However, expression of the death domain of RIP Induces apoptosis, but potently inhibits NF-kappa B activation by TNF. These results suggest that distinct domains of RIP participate in the TNF signaling cascades leading to apoptosis and NF-kappa B activation.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Sequence , Animals , Apoptosis , Cell Line , Humans , Molecular Sequence Data , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , Proteins/genetics , Receptor-Interacting Protein Serine-Threonine Kinases , Recombinant Proteins/metabolism , Signal Transduction , TNF Receptor-Associated Factor 1
14.
Cell ; 84(2): 299-308, 1996 Jan 26.
Article in English | MEDLINE | ID: mdl-8565075

ABSTRACT

Tumor necrosis factor (TNF) can induce apoptosis and activate NF-kappa B through signaling cascades emanating from TNF receptor 1 (TNFR1). TRADD is a TNFR1-associated signal transducer that is involved in activating both pathways. Here we show that TRADD directly interacts with TRAF2 and FADD, signal transducers that activate NF-kappa B and induce apoptosis, respectively. A TRAF2 mutant lacking its N-terminal RING finger domain is a dominant-negative inhibitor of TNF-mediated NF-kappa B activation, but does not affect TNF-induced apoptosis. Conversely, a FADD mutant lacking its N-terminal 79 amino acids is a dominant-negative inhibitor of TNF-induced apoptosis, but does not inhibit NF-kappa B activation. Thus, these two TNFR1-TRADD signaling cascades appear to bifurcate at TRADD.


Subject(s)
Adaptor Proteins, Signal Transducing , Antigens, CD/physiology , Carrier Proteins/metabolism , Proteins , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/physiology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins , Amino Acid Sequence , Animals , Antigens, CD/metabolism , Apoptosis/physiology , Carrier Proteins/genetics , Cell Line , Fas-Associated Death Domain Protein , Humans , Interleukin-1/pharmacology , Mice , Molecular Sequence Data , NF-kappa B/physiology , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Sequence Deletion , TNF Receptor-Associated Death Domain Protein , TNF Receptor-Associated Factor 1 , TNF Receptor-Associated Factor 2 , Transcriptional Activation , Tumor Necrosis Factor-alpha/pharmacology
15.
J Cell Sci ; 108 ( Pt 9): 2955-62, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8537435

ABSTRACT

gamma-Tubulin, a relatively new member of the tubulin gene family, is localized primarily at the centrosome throughout the mammalian cell cycle and may play a key role in nucleation of cellular microtubule assembly. A transient association of gamma-tubulin at the cytoplasmic bridge of telophase mammalian cells, the midbody, is recently documented. Using immunogold electron microscopy and serial section reconstruction analysis, we show here that the transiently associated midbody gamma-tubulin is localized at the minus ends of microtubules in the midbody structure. Using antisense RNA methods we also demonstrate that a selective depletion of transiently associated midbody gamma-tubulin causes an abortive cytokinesis due to a failure in the morphogenesis of the midbody structure.


Subject(s)
Centrosome/ultrastructure , Microtubules/ultrastructure , Tubulin/genetics , Animals , Cell Division/physiology , Cell Line , HeLa Cells , Humans , Image Processing, Computer-Assisted , Morphogenesis , RNA, Antisense , Time Factors , Tubulin/biosynthesis
16.
J Cell Biol ; 130(5): 1137-47, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7657698

ABSTRACT

alpha-, beta-, and gamma-tubulins are evolutionarily highly conserved members of the tubulin gene superfamily. While the abundant members, alpha- and beta-tubulins, constitute the building blocks of cellular microtubule polymers, gamma-tubulin is a low abundance protein which localized to the pericentriolar material and may play a role in microtubule assembly. To test whether gamma-tubulin mediates the nucleation of microtubule assembly in vivo, and co-assembles with alpha- and beta-tubulins into microtubules or self-assembles into macro-molecular structures, we experimentally elevated the expression of gamma-tubulin in the cell cytoplasm. In most cells, overexpression of gamma-tubulin causes a dramatic reorganization of the cellular microtubule network. Furthermore, we show that when overexpressed, gamma-tubulin causes ectopic nucleation of microtubules which are not associated with the centrosome. In a fraction of cells, gamma-tubulin self-assembles into novel tubular structures with a diameter of approximately 50 nm (named gamma-tubules). Furthermore, unlike microtubules, gamma-tubules are resistant to cold or drug induced depolymerization. These data provide evidence that gamma-tubulin can cause nucleation of microtubule assembly and can self-assemble into novel tubular structures.


Subject(s)
Microtubules/physiology , Tubulin/physiology , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured/cytology , Centrosome/physiology , Cytoplasm/physiology , Fluorescent Antibody Technique , Gene Expression/physiology , Haplorhini , Immunohistochemistry , Microscopy, Electron , Microtubules/ultrastructure , Molecular Sequence Data , Nocodazole/pharmacology , Polymers/metabolism , Rabbits , Thermodynamics , Tubulin/genetics , Tubulin/metabolism , Tubulin/pharmacology
17.
Proc Natl Acad Sci U S A ; 90(22): 10759-63, 1993 Nov 15.
Article in English | MEDLINE | ID: mdl-8248168

ABSTRACT

The arterial wall responds to thrombosis or mechanical injury through the induction of specific gene products that increase cellular proliferation and connective tissue formation. These changes result in intimal hyperplasia that is observed in restenosis and the early phases of atherosclerosis. Transforming growth factor beta 1 (TGF-beta 1) is a secreted multi-functional protein that plays an important role in embryonal development and in repair following tissue injury. However, the function of TGF-beta 1 in vascular cell growth in vivo has not been defined. In this report, we have evaluated the role of TGF-beta 1 in the pathophysiology of intimal and medial hyperplasia by gene transfer of an expression plasmid encoding active TGF-beta 1 into porcine arteries. Expression of TGF-beta 1 in normal arteries resulted in substantial extracellular matrix production accompanied by intimal and medial hyperplasia. Increased procollagen, collagen, and proteoglycan synthesis in the neointima was demonstrated by immunohistochemistry relative to control transfected arteries. Expression of TGF-beta 1 induced a distinctly different program of gene expression and biologic response from the platelet-derived growth factor B (PDGF B) gene: procollagen synthesis induced by TGF-beta 1 was greater, and cellular proliferation was less prominent. These findings show that TGF-beta 1 differentially modulates extracellular matrix production and cellular proliferation in the arterial wall in vivo and could play a reparative role in the response to arterial injury.


Subject(s)
Arteries/pathology , Arteriosclerosis/pathology , Transforming Growth Factor beta/physiology , Animals , Arteries/metabolism , Base Sequence , Collagen/metabolism , DNA Primers/chemistry , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression , Gene Transfer Techniques , Hyperplasia , Molecular Sequence Data , Platelet-Derived Growth Factor/metabolism , Polymerase Chain Reaction , Procollagen/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-sis , RNA, Messenger/genetics , Swine , Tunica Intima/metabolism , Tunica Intima/pathology
18.
Mol Cell Biol ; 13(10): 6283-9, 1993 Oct.
Article in English | MEDLINE | ID: mdl-7692229

ABSTRACT

Vascular cell adhesion molecule 1 (VCAM-1) is expressed in both endothelial and epithelial cell types, where it contributes to lymphocyte migration to sites of inflammation. Its expression is regulated by cytokines, in part through two kappa B-like regulatory elements. Because NF-kappa B can be composed of multiple alternative subunits with differential effects on gene expression, the role of different specific NF-kappa B family members subunits in VCAM-1 regulation is unknown. In this report, we define the contribution of different NF-kappa B family members to VCAM-1 gene regulation. We show that both kappa B sites in the VCAM-1 enhancer are required to optimally stimulate gene expression, but the enhancer is differentially regulated by specific combinations of NF-kappa B subunits. At low concentrations, RelA(p65) acted in concert with the approximately 50-kDa product of p105 NF-kappa B, NF-kappa B1(p50), to stimulate transcription, and at high concentrations, RelA(p65) alone stimulated the VCAM-1 promoter. In contrast, NF-kappa B2 inhibited functional activation of the VCAM reporter by p65. Consistent with this finding, an additional binding complex was detected by using recombinant NF-kappa B2(p49)/RelA(p65) with radiolabeled VCAM kappa B site probes. Interestingly, the human immunodeficiency virus enhancer responded differently to stimulation by NF-kappa B subunits, with optimal response to p49(100)/p65. Analysis of NF-kappa B mRNA in human umbilical vein endothelial cells revealed that nfkb1, nfkb2, and relA NF-kappa B but not c-rel were induced by tumor necrosis factor alpha and lipopolysaccharide, which also induce VCAM-1. These data suggest that specific subunits of NF-kappa B regulate VCAM-1 and differentially activate other genes in these cells.


Subject(s)
Cell Adhesion Molecules/genetics , Endothelium, Vascular/metabolism , Gene Expression Regulation , I-kappa B Proteins , NF-kappa B/metabolism , Animals , B-Cell Lymphoma 3 Protein , Base Sequence , Binding Sites , Cell Line , DNA , DNA-Binding Proteins/pharmacology , Endothelium, Vascular/cytology , Enhancer Elements, Genetic , Epithelial Cells , Epithelium/metabolism , Gene Expression Regulation/drug effects , HIV/genetics , HeLa Cells , Humans , Lipopolysaccharides/pharmacology , Molecular Sequence Data , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , Proto-Oncogene Proteins/pharmacology , Swine , Transcription Factors , Transfection , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1
SELECTION OF CITATIONS
SEARCH DETAIL
...