201
|
Kishimoto H, Sprent J. Strong TCR Ligation Without Costimulation Causes Rapid Onset of Fas-Dependent Apoptosis of Naive Murine CD4+ T Cells. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.4.1817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Abstract
Activation-induced cell death of T cells typically occurs late in the primary response after a prior proliferative response. Here, we describe a novel form of cell death in which purified naive murine CD4+ cells undergo apoptosis within 18 h in vitro after strong TCR ligation. Such rapid-onset TCR-mediated death of T cells does not involve cell division and is Fas-dependent, inhibited by CD28 (and IL-6) costimulation and enhanced by IL-4 and IL-7; by contrast, spontaneous death of CD4+ cells cultured alone is Fas-independent and inhibited by IL-4 and IL-7. TCR-mediated Fas-dependent death of CD4+ cells is prevented by combined TCR/Fas ligation and by drugs that inhibit calcineurin-dependent signaling and mitogen-activated protein kinase MEK1 activation.
Collapse
Affiliation(s)
- Hidehiro Kishimoto
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037
| | - Jonathan Sprent
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
202
|
Löffler M, Ausserlechner MJ, Tonko M, Hartmann BL, Bernhard D, Geley S, Helmberg A, Kofler R. c-Myc does not prevent glucocorticoid-induced apoptosis of human leukemic lymphoblasts. Oncogene 1999; 18:4626-31. [PMID: 10467407 DOI: 10.1038/sj.onc.1202820] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Due to their growth arrest- and apoptosis-inducing ability, glucocorticoids (GC) are widely used in the therapy of various lymphoid malignancies. The signal transduction pathways leading to this clinically-relevant form of apoptosis have, however, not been sufficiently elucidated. GC bind to their specific receptor, a ligand-activated transcription factor of the Zn-finger type, that activates or represses transcription of GC-responsive genes. Previous studies in leukemia cells suggested that transcriptional repression of c-myc expression might be the crucial event in GC-induced apoptosis, although in other systems, c-Myc apparently increased the sensitivity to cell-death inducers. To address this controversy, we stably transfected the GC-sensitive human T-ALL cell line CEM-C7H2 with constructs allowing tetracycline-regulated expression of c-Myc. Subsequent analyses of these cell lines showed that overexpression of c-Myc per se had little, if any, effect on cell viability, although it rendered the cells more sensitive to apoptosis induced by low serum, confirming the functionality of the expressed transgene. More importantly, however, when the cells were treated with GC in the presence of exogenous c-Myc, they underwent apoptosis exceeding that in cells treated in the absence of transgenic c-Myc. The data indicate that c-myc downregulation is not critical for induction of cell-death by GC in this system, and support the notion that c-Myc sensitizes cells to apoptosis-inducing agents.
Collapse
Affiliation(s)
- M Löffler
- Institute for General and Experimental Pathology, Division of Molecular Pathophysiology, University of Innsbruck, Innsbruck, Austria
| | | | | | | | | | | | | | | |
Collapse
|
203
|
Abstract
In the pathogenesis of cervical carcinoma there are three major components, two of them related to the role of human papillomaviruses (HPV). First, the effect of viral E6 and E7 proteins. Second, the integration of viral DNA in chromosomal regions associated with well known tumour phenotypes. Some of these viral integrations occur recurrently at specific chromosomal locations, such as 8q24 and 12q15, both harbouring HPV18 and HPV16. And third, there are other recurrent genetic alterations not linked to HPV. Recurrent losses of heterozygosity (LOH) have been detected in chromosome regions 3p14-22, 4p16, 5p15, 6p21-22, 11q23, 17p13.3 without effect on p53, 18q12-22 and 19q13, all of them suggesting the alteration of putative tumour suppressor genes not yet identified. Recurrent amplification has been mapped to 3q+ arm, with the common region in 3q24-28 in 90% of invasive carcinomas. The mutator phenotype, microsatellite instability, plays a minor role and is detected in only 7% of cervical carcinomas. The development of cervical carcinoma requires the sequential occurrence and selection of several genetic alterations. The identification of the specific genes involved, and their correlation with specific tumour properties and stages could improve the understanding and perhaps the management of cervical carcinoma.
Collapse
Affiliation(s)
- P A Lazo
- Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Universidad de Salamanca, Spain
| |
Collapse
|
204
|
Affiliation(s)
- M J Arends
- University of Cambridge, Department of Histopathology, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
205
|
Abstract
Neutrophils undergo constitutive apoptosis when aged ex vivo. Recent studies have indicated roles for Fas/CD95 and the nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase system in this process. We have investigated the role of protein kinase C (PKC) in neutrophil death. We show that there is proteolysis and activation of the novel isoform PKCδ in aged neutrophils and that this process is accelerated by the addition of an agonistic Fas antibody. PKCδ proteolysis occurs before the onset of any detectable features of apoptosis and pharmacologic inhibition of this enzyme inhibits neutrophil apoptosis. PKCδ cleavage and activation is dependent on caspase-8/FADD-like interleukin-1β converting enzyme (FLICE)–mediated processing of caspase-3/CPP32. Neutrophil survival is prolonged by the addition of broad spectrum (BD.fmk) or caspase-8 targeted (zIETD.fmk) peptide caspase inhibitors. Inhibition of PKCδ does not prevent apoptosis triggered by factor withdrawal in immature hematopoietic cells, including normal human CD34+ progenitors indicating that within a given lineage, the mechanisms of apoptosis may be differentiation-stage–specific. Ex vivo aging of neutrophils leads to the increasing production of reactive oxygen species and this is attenuated in cells treated with either caspase or PKCδ inhibitors. Proteolytically activated PKCδ acts as a molecular link between the Fas/CD95 receptor and the NADPH-oxidase system and plays a central role in regulating the process of neutrophil apoptosis.
Collapse
|
206
|
Abstract
Fas ligation in the presence of cycloheximide induced Jun N-terminal kinase 1 (JNK1) and JNK2 phosphorylation, caspase activation and cell death in the IL-3-dependent cell line BAF3. Fas-mediated apoptosis was prevented by expression of dominant negative FADD but not inhibited by IL-3. To investigate the role of JNK activation in this process, we examined cells over-expressing a JNK-specific phosphatase M3/6. M3/6 prevented Fas stimulation of JNK, but did not affect Fas-mediated caspase activation or cell death, demonstrating that JNK activation is not required for these processes.
Collapse
Affiliation(s)
- W Low
- Department of Immunology, University College London, UK
| | | | | | | |
Collapse
|
207
|
Yeh WC, Hakem R, Woo M, Mak TW. Gene targeting in the analysis of mammalian apoptosis and TNF receptor superfamily signaling. Immunol Rev 1999; 169:283-302. [PMID: 10450525 DOI: 10.1111/j.1600-065x.1999.tb01323.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Apoptosis, or programmed cell death (PCD), is the subject of much current investigative interest. Developing embryos and many adult organ systems require the tight coupling of cellular proliferation and PCD to ensure proper organogenesis and optimal tissue function. Over the past decade, our knowledge of the genetic basis underlying the execution of apoptosis in mammals has progressed enormously, thanks largely to groundbreaking studies performed in the nematode Caenorhabditis elegans. In contrast, the components of the signaling apparatus that links the various death stimuli and the receptors they stimulate to the execution mechanism remain relatively unknown. It is only in the past 4 years that studies of signal transduction via members of the tumor necrosis factor (TNF) receptor superfamily have identified a plethora of novel signaling proteins, including molecules that are directly involved in apoptosis signaling, and others that regulate the induction of cell death. This two-part review focuses on the biology of apoptosis and signaling through members of the TNF receptor superfamily as revealed by the study of gene-targeted "knockout" mice. These genetic mutant animals are invaluable tools not only for confirming or refuting a proposed function of a particular gene in an in vivo setting, but also for uncovering novel functions for a gene that were not anticipated from conventional in vitro experiments. In the field of apoptosis, as for many other areas of biomedical research, knockout mice and cell lines can be used as models for studying human disease, with the ultimate goal of developing therapeutic strategies.
Collapse
Affiliation(s)
- W C Yeh
- Amgen Institute, Department of Medical Biophysics, University of Toronto, Canada
| | | | | | | |
Collapse
|
208
|
Matsuo Y, Sugimoto A, Harashima A, Nishizaki C, Ishimaru F, Kondo E, Katayama Y, Harada M, Orita K. Establishment and characterization of a novel ALL-L3 cell line (BALM-18): induction of apoptosis by anti-IgM and inhibition of apoptosis by bone marrow stroma cells. Leuk Res 1999; 23:559-68. [PMID: 10374849 DOI: 10.1016/s0145-2126(99)00046-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
A human acute lymphoblastic leukemia (ALL) cell line, BALM-18, was established from the peripheral blood specimen of a patient with B cell ALL L3 type (ALL-L3) at diagnosis using bone marrow stroma cells (BST) as feeder cells. The primary leukemia cells did not grow without feeder cells. As with the primary leukemia cells, BALM-18 showed an immunophenotype of Burkitt's lymphoma group I [CD10+, CD20+, CD23-, CD39-, CD77+] and carried the t(8;14)(q24;q32) chromosomal abnormality which is highly associated with ALL-L3 and Burkitt's lymphoma. It also revealed a significantly low level of bcl-2 protein. Strikingly, anti-human IgM antibody did induce apoptosis in induction experiments. However, it was reversed by the addition of anti-CD40 antibody or BST cells, whereas the culture supernatant of the stroma cells did not show any effect on the inhibition of apoptosis. BALM-18 may be useful for analyzing both the mechanisms of anti-IgM induced apoptosis and signaling during the inhibition of apoptosis by CD40 or BST cells.
Collapse
Affiliation(s)
- Y Matsuo
- Fujisaki Cell Center, Hayashibara Biochemical Labs. Inc., Okayama, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
209
|
Juin P, Hueber AO, Littlewood T, Evan G. c-Myc-induced sensitization to apoptosis is mediated through cytochrome c release. Genes Dev 1999; 13:1367-81. [PMID: 10364155 PMCID: PMC316765 DOI: 10.1101/gad.13.11.1367] [Citation(s) in RCA: 263] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Expression of c-Myc sensitizes cells to a wide range of pro-apoptotic stimuli. We here show that this pro-apoptotic effect is mediated through release of mitochondrial holocytochrome c into the cytosol. First, activation of c-Myc triggers release of cytochrome c from mitochondria. This release is caspase-independent and blocked by the survival factor IGF-1. Second, c-Myc-induced apoptosis is blocked by microinjection of anticytochrome c antibody. In addition, we show that microinjection of holocytochrome c mimics the effect of c-Myc activation, sensitizing cells to DNA damage and to the CD95 pathway. Both p53 and CD95/Fas signaling have been implicated in c-Myc-induced apoptosis but neither was required for c-Myc-induced cytochrome c release. Nonetheless, inhibition of CD95 signaling in fibroblasts did prevent c-Myc-induced apoptosis, apparently by obstructing the ability of cytosolic cytochrome c to activate caspases. We conclude that c-Myc promotes apoptosis by causing the release of cytochrome c, but the ability of cytochrome c to activate apoptosis is critically dependent upon other signals.
Collapse
Affiliation(s)
- P Juin
- Imperial Cancer Research Fund, London WC2A 3PX, UK
| | | | | | | |
Collapse
|
210
|
Abstract
Much recent research on c-Myc has focused on how it drives apoptosis. c-Myc is widely known as a crucial regulator of cell proliferation in normal and neoplastic cells, but until relatively recently its apoptotic properties, which appear to be intrinsic, were not fully appreciated. Its death-dealing aspects have gained wide attention in part because of their potential therapeutic utility in advanced malignancy, where c-Myc is frequently deregulated and where novel modalities are badly needed. Although its exact function remains obscure, c-Myc is a transcription factor and advances have been made in characterizing target genes which may mediate its apoptotic properties. Candidate regulators and effectors are also emerging. Among recent findings are connections to the CD95/Fas and TNF pathways and roles for the tumor suppressor p19ARF and the c-Myc-interacting adaptor protein Binl in mediating cell death. In this review I summarize the data establishing a role for c-Myc in apoptosis in diverse settings and present a modified dual signal model for c-Myc function. It is proposed that c-Myc induces apoptosis through separate 'death priming' and 'death triggering' mechanisms in which 'death priming' and mitogenic signals are coordinated. Investigation of the mechanisms that underlie the triggering steps may offer new therapeutic opportunities.
Collapse
|
211
|
Hashiramoto A, Sano H, Maekawa T, Kawahito Y, Kimura S, Kusaka Y, Wilder RL, Kato H, Kondo M, Nakajima H. C-myc antisense oligodeoxynucleotides can induce apoptosis and down-regulate Fas expression in rheumatoid synoviocytes. ARTHRITIS AND RHEUMATISM 1999; 42:954-62. [PMID: 10323451 DOI: 10.1002/1529-0131(199905)42:5<954::aid-anr14>3.0.co;2-j] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE To investigate the role of c-myc in the pathogenesis of rheumatoid arthritis (RA) and the mechanism of synovial apoptosis. METHODS Using cultured human synoviocytes from patients with RA and c-myc antisense oligodeoxynucleotides (AS ODN), we examined the inhibition of cell proliferation by the MTT assay and the induction of apoptosis with TUNEL staining and fluorescence microscopy. In addition, the effect of c-myc on down-regulation of Fas expression was analyzed by flow cytometry, cytotoxicity assay, and reverse transcriptase-polymerase chain reaction. RESULTS Treatment with c-myc AS ODN induced inhibition of cell proliferation, along with down-regulation of c-Myc protein and c-myc messenger RNA (mRNA) expression. The morphologic changes of synovial cell death were typical of apoptosis. In addition, c-myc AS ODN treatment down-regulated expression of Fas mRNA but not Fas antigen. Analysis of the involvement of the caspase cascade revealed that the cytotoxic activity of c-myc AS ODN was completely blocked by inhibitors of both caspase 1 (YVAD-FMK) and caspase 3 (DEVD-FMK). CONCLUSION Our results strongly suggest that c-myc AS ODN might be a useful therapeutic tool in RA and clarify that cell death by c-myc AS ODN is induced through the caspase cascade, similar to Fas-induced apoptosis. In addition, combination therapy with anti-Fas antibody and c-myc AS ODN reduced Fas-dependent cytotoxicity.
Collapse
|
212
|
Peli J, Schröter M, Rudaz C, Hahne M, Meyer C, Reichmann E, Tschopp J. Oncogenic Ras inhibits Fas ligand-mediated apoptosis by downregulating the expression of Fas. EMBO J 1999; 18:1824-31. [PMID: 10202146 PMCID: PMC1171268 DOI: 10.1093/emboj/18.7.1824] [Citation(s) in RCA: 127] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Tumor growth is the result of deregulated tissue homeostasis which is maintained through the delicate balance of cell growth and apoptosis. One of the most efficient inducers of apoptosis is the death receptor Fas. We report here that oncogenic Ras (H-Ras) downregulates Fas expression and renders cells of fibroblastic and epitheloid origin resistant to Fas ligand-induced apoptosis. In Ras-transformed cells, Fas mRNA is absent. Inhibition of DNA methylation restores Fas expression. H-Ras signals via the PI 3-kinase pathway to downregulate Fas, suggesting that the known anti-apoptotic effect of the downstream PKB/Akt kinase may be mediated, at least in part, by the repression of Fas expression. Thus, the oncogenic potential of H-ras may reside on its capacity not only to promote cellular proliferation, but also to simultaneously inhibit Fas-triggered apoptosis.
Collapse
Affiliation(s)
- J Peli
- Swiss Cancer Research Institute (ISREC), BIL Biomedical Research Center, Chemin des Boveresses 155, Switzerland
| | | | | | | | | | | | | |
Collapse
|
213
|
Abstract
Chemotherapy of malignant tumors, including acute leukemias, is largely based on empirical data and clinical experience. In recent years, it has become clear that anticancer drugs induce apoptosis in target cells. Activation of the CD95 system initiated by induction of CD95 ligand/receptor interaction has been shown to contribute to drug-induced apoptosis in certain systems. However, some groups also have shown that drug-induced apoptosis occurs independently of CD95 triggering. Drug-induced activation of apoptotic pathways appears to include parallel or sequential activation of death receptor systems and, most importantly, changes in mitochondrial function with concomitant release of apoptogenic factors. Activation of downstream caspases, such as caspase 3 or caspase 9, seems to be the most critical mediator of a drug-induced cell death. The identification of the crucial elements of drug-induced apoptosis and activation of effector caspases will certainly provide new insights into the molecular determinants of chemosensitivity or chemoresistance of malignant tumors and leukemias. Copyright 1999 Harcourt Publishers Ltd.
Collapse
|
214
|
Klefstrom J, Kovanen PE, Somersalo K, Hueber AO, Littlewood T, Evan GI, Greenberg AH, Saksela E, Timonen T, Alitalo K. c-Myc and E1A induced cellular sensitivity to activated NK cells involves cytotoxic granules as death effectors. Oncogene 1999; 18:2181-8. [PMID: 10327064 DOI: 10.1038/sj.onc.1202546] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The contact of natural killer (NK) cells with foreign cells and with certain virus-infected or tumor cells triggers the cytolytic machinery of NK cells. This triggering leads to exocytosis of the cytotoxic NK cell granules. The oncoproteins c-Myc and E1A render cells vulnerable to NK cell mediated cytolysis yet the mechanisms of sensitization are not well understood. In a model where foreign cells (rat fibroblasts) were cocultured with human IL-2 activated NK cells, we observed that NK cells were capable of efficiently killing their targets only if the cells overexpressed the oncogene c-Myc or E1A. Both the parental and the oncogene expressing fibroblasts similarly triggered phosphoinositide hydrolysis in the bound NK cells, demonstrating that NK cells were cytolytically activated in contact with both resistant parental and oncogene expressing sensitive target fibroblasts. The cell death was independent of wild-type p53 and was not inhibited by an anti-apoptotic protein EIB19K. These results provided evidence that c-Myc and E1A activated the NK cell induced cytolysis at a post-triggering stage of NK cell-target cell interaction. In consistence, the c-Myc and E1A overexpressing fibroblasts were more sensitive to the cytolytic effects of isolated NK cell-derived granules than parental cells. The data indicate that oncogenes activate the cytotoxicity of NK cell granules. This mechanism can have a role in directing the cytolytic action of NK cells towards the virus-infected and cancer cells.
Collapse
Affiliation(s)
- J Klefstrom
- Biochemistry of the Cell Nucleus Laboratory, Imperial Cancer Research Fund, London, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
215
|
Papas KK, Sun L, Roos ES, Gounarides JS, Shapiro M, Nalin CM. Change in lactate production in Myc-transformed cells precedes apoptosis and can be inhibited by Bcl-2 overexpression. FEBS Lett 1999; 446:338-42. [PMID: 10100870 DOI: 10.1016/s0014-5793(99)00240-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
As a result of Myc-dependent transcription of the LDH-A gene, Myc-transformed cells (Rat1-Myc) exhibit increased lactate production rates (LPR) even under aerobic conditions (the Warburg effect). Recently, the increased susceptibility to stress-induced apoptosis associated with Myc transfection has been linked to the overexpression of the LDH-A gene. In this report we demonstrate that the overexpression of the anti-apoptotic protein Bcl-2 in Rat1-Myc cells (Rat1-Myc-Bcl-2) reduces the molar ratio of lactate production to glucose consumption (Y(L/G)). The Bcl-2 induced reduction in Y(L/G) may be associated with reduced expression of the LDH-A gene, or a decrease in LDH-A activity. Stimulation of apoptosis by staurosporine, a protein kinase C inhibitor, reduces the LPR in Rat1-Myc cells in a dose-dependent manner. The staurosporine effect on the LPR is rapid and precedes the execution phase of apoptosis as defined by caspase activation and PARP cleavage. This effect on LPR is completely blocked by Bcl-2 overexpression. Serum starvation alone does not affect the LPR of Rat1-Myc or Rat1-Myc-Bcl-2 cells; however, the effect of staurosporine on the LPR of Rat1-Myc cells is potentiated by serum starvation. These data demonstrate that Bcl-2 overexpression reduces the Y(L/G) in Rat1-Myc cells, perhaps via a reduction in the activity or expression of the LDH-A gene, and this reduction may desensitize cells to some pro-apoptotic stimuli. The reduction in LPR in response to staurosporine may be an early step in the induction of apoptosis in Rat1-Myc cells. By abolishing the reduction in LPR, Bcl-2 may protect Rat1-Myc cells from staurosporine-induced apoptosis. Moreover, the lack of effect by serum starvation on the LPR supports a model in which serum starvation induces apoptosis through a pathway distinct from that of the staurosporine and glucose-dependent apoptotic pathway(s) in Myc-transformed cells.
Collapse
Affiliation(s)
- K K Papas
- Core Technologies/Analytics and Bio-NMR US, Novartis Institute for Biomedical Research, Novartis Pharmaceuticals Corporation, Summit, NJ 07901, USA
| | | | | | | | | | | |
Collapse
|
216
|
Tang D, Lahti JM, Grenet J, Kidd VJ. Cycloheximide-induced T-cell death is mediated by a Fas-associated death domain-dependent mechanism. J Biol Chem 1999; 274:7245-52. [PMID: 10066786 DOI: 10.1074/jbc.274.11.7245] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cycloheximide (CHX) can contribute to apoptotic processes, either in conjunction with another agent (e.g. tumor necrosis factor-alpha) or on its own. However, the basis of this CHX-induced apoptosis has not been clearly established. In this study, the molecular mechanisms of CHX-induced cell death were examined in two different human T-cell lines. In T-cells undergoing CHX-induced apoptosis (Jurkat), but not in T-cells resistant to the effects of CHX (CEM C7), caspase-8 and caspase-3 were activated. However, the Fas ligand was not expressed in Jurkat cells either before or after treatment with CHX, suggesting that the activation of these caspases does not involve the Fas receptor. To determine whether CHX-induced apoptosis was mediated by a Fas-associated death domain (FADD)-dependent mechanism, a FADD-DN protein was expressed in cells prior to CHX treatment. Its expression effectively inhibited CHX-induced cell death, suggesting that CHX-mediated apoptosis primarily involves a FADD-dependent mechanism. Since CHX treatment did not result in the induction of Fas or FasL, and neutralizing anti-Fas and anti-tumor necrosis factor receptor-1 antibodies did not block CHX-mediated apoptosis, these results may also indicate that FADD functions in a receptor-independent manner. Surprisingly, death effector filaments containing FADD and caspase-8 were observed during CHX treatment of Jurkat, Jurkat-FADD-DN, and CEM C7 cells, suggesting that their formation may be necessary, but not sufficient, for cell death.
Collapse
Affiliation(s)
- D Tang
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38101, USA
| | | | | | | |
Collapse
|
217
|
Fulda S, Lutz W, Schwab M, Debatin KM. MycN sensitizes neuroblastoma cells for drug-induced apoptosis. Oncogene 1999; 18:1479-86. [PMID: 10050884 DOI: 10.1038/sj.onc.1202435] [Citation(s) in RCA: 101] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Amplification of the MYCN gene is found in a large proportion of neuroblastoma and considered as an adverse prognostic factor. To investigate the effect of ectopic MycN expression on the susceptibility of neuroblastoma cells to cytotoxic drugs we used a human neuroblastoma cell line harboring tetracycline-controlled expression of MycN. Neither conditional expression of MycN alone nor low drug concentrations triggered apoptosis. However, when acting in concert, MycN and cytotoxic drugs efficiently induced cell death. Apoptosis depended on mitochondrial permeability transition and activation of caspases, since the mitochondrion-specific inhibitor bongkrekic acid and the caspase inhibitor zVAD-fmk almost completely abrogated apoptosis. Loss of mitochondrial transmembrane potential and release of cytochrome c from mitochondria preceded activation of caspase-8 and caspase-3 and cleavage of PARP. CD95 expression was upregulated by treatment with cytotoxic drugs, while MycN cooperated with cytotoxic drugs to increase sensitivity to CD95-induced apoptosis and enhancing CD95-L expression. MycN overexpression and cytotoxic drugs also synergized to induce p53 and Bax protein expression, while Bcl-2 and Bcl-X(L) protein levels remained unchanged. Since amplification of MYCN is usually associated with a poor prognosis, these findings suggest that dysfunctions in apoptosis pathways may be a mechanism by which MycN-induced apoptosis of neuroblastoma cells is inhibited.
Collapse
Affiliation(s)
- S Fulda
- University Children's Hospital, Ulm, Germany
| | | | | | | |
Collapse
|
218
|
Choi C, Park JY, Lee J, Lim JH, Shin EC, Ahn YS, Kim CH, Kim SJ, Kim JD, Choi IS, Choi IH. Fas Ligand and Fas Are Expressed Constitutively in Human Astrocytes and the Expression Increases with IL-1, IL-6, TNF-α, or IFN-γ. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.4.1889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Fas ligand (FasL) and Fas are mediators of apoptosis, which are implicated in the peripheral deletion of autoimmune cells, activation-induced T cell death, and cytotoxicity mediated by CD8+ T cells. Fas is also believed to be involved in several central nervous system diseases, but until now, the effector cells expressing FasL in the brain have not been identified. We investigated the expression levels of Fas and FasL with the stimulation of cytokines and the possible effector cells targeting Fas-bearing cells. Our data demonstrated that: 1) FasL is expressed constitutively on astrocytes taken from a fetus or an adult and that its expression increases when these cells are treated with IL-1, IL-6, or TNF-α in which the pretreatment of IFN-γ triggers astrocytes to express more FasL; 2) astrocytes induce apoptosis in MOLT-4 cells through FasL; 3) Fas is also expressed constitutively and is up-regulated by IL-1, IL-6, or TNF-α in which the pretreatment of IFN-γ triggers astrocytes to express more Fas; 4) apoptosis occurs when fetal astrocytes are treated with agonistic anti-Fas IgM Ab after culture with IFN-γ and TNF-α; and 5) TNF-related apoptosis inducing ligand is up-regulated in fetal astrocytes with stimuli of IL-1 or TNF-α. These findings suggest a possible role of astrocytes in the induction of apoptosis in central nervous system diseases.
Collapse
Affiliation(s)
- Chulhee Choi
- *Microbiology and Institute for Immunology and Immunological Diseases,
- †Neurology, and
| | - Joo Young Park
- ‡Pharmacology, Yonsei University College of Medicine, Seoul, Korea; and
| | - Jeonggi Lee
- *Microbiology and Institute for Immunology and Immunological Diseases,
| | - Jung-Hee Lim
- *Microbiology and Institute for Immunology and Immunological Diseases,
| | - Eui-Cheol Shin
- *Microbiology and Institute for Immunology and Immunological Diseases,
| | - Young Soo Ahn
- §Wonju College of Medicine, Yonsei University, Wonju, Korea
| | - Chul-Hoon Kim
- §Wonju College of Medicine, Yonsei University, Wonju, Korea
| | - Se-Jong Kim
- *Microbiology and Institute for Immunology and Immunological Diseases,
| | - Joo-Deuk Kim
- *Microbiology and Institute for Immunology and Immunological Diseases,
| | | | - In-Hong Choi
- *Microbiology and Institute for Immunology and Immunological Diseases,
| |
Collapse
|
219
|
Abstract
Though p53-induced apoptosis plays an important role in tumor suppression, the mechanism(s) by which p53 induces apoptosis is still unclear. To elucidate the p53-induced apoptotic pathway, we examined the role of p53 transactivation activity and caspase in J138V5C cells carrying a human temperature-sensitive (ts) p53 mutant (138Ala-->Val). The results showed that p53-induced apoptosis was not blocked by cycloheximide, which effectively prevented the expression of p53 target genes, indicating that transactivation was not essential for p53-induced apoptosis in this system. Western blot analysis showed that PARP, CPP32 and ICH-1 precursors were cleaved during apoptosis. The CPP32-preferential tetrapeptide inhibitor Ac-DEVD-CHO blocked the cleavage of ICH-1 and PARP precursors, suggesting that CPP32 or some other DEVD-sensitive caspase(s) is the upstream activator of ICH-1. We also examined the role of the Fas pathway by using Fas and Fas ligand-neutralizing antibodies. Both antibodies failed to block p53-induced apoptosis, suggesting that the Fas pathway was not essential for p53-induced apoptosis in this system. Taken together, our results indicate that p53-induced, transactivation-independent apoptosis in Jurkat cells involves sequential activation of CPP32 or some other DEVD-sensitive caspase(s) and ICH-1, via a Fas-independent pathway.
Collapse
Affiliation(s)
- C Gao
- Department of Molecular Cellular Oncology, Tokyo Medical and Dental University
| | | |
Collapse
|
220
|
Burtscher I, Christofori G. The IGF/IGF-1 receptor signaling pathway as a potential target for cancer therapy. Drug Resist Updat 1999; 2:3-8. [PMID: 11504463 DOI: 10.1054/drup.1998.0061] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Expression of insulin-like growth factors (IGFs) and their cognate receptor, the IGF-1 receptor, is frequently upregulated during the development of many types of cancer. Besides stimulating cell cycle progression and the transformation status of tumor cells, a wealth of recent experimental data suggests that IGF-mediated signaling exerts a central tumor-promoting function through the repression of tumor cell apoptosis. These functions are all conveyed by the IGF-1 receptor, thus making it an attractive target for therapeutic intervention. Notably, inhibition of IGF-mediated survival function appears to synergize with conventional chemotherapeutic ablation of tumor cells, raising the possibility of combinatorial cancer therapies with significantly reduced side-effects. Copyright 1999 Harcourt Publishers Ltd.
Collapse
Affiliation(s)
- Ingo Burtscher
- Research Institute of Molecular Pathology, Vienna, Austria
| | | |
Collapse
|
221
|
Abstract
The human CASP8 gene, whose product is also known as caspase 8 and FLICE, encodes an interleukin-1beta converting enzyme (ICE)-related cysteine protease that is activated by the engagement of several different death receptors. Caspase 8 is immediately recruited to the Fas receptor once it oligomerizes, and its protease activity is crucial for the apoptotic response generated by the resulting death-inducing signaling complex (DISC). We report here that the CASP8 gene contains at least 11 exons spanning approximately 30kb on human chromosome band 2q33-34. This region of human chromosome 2 was previously reported as the location of the CASP10 gene, whose product is closely related to caspase 8. Chromosome 2 band q33-34 is also involved in tumorigenesis, with loss of heterogeneity (LOH) being reported in a number of tumors. We also report EcoRI and HindIII polymorphisms that may prove to be useful in disease analysis. Both caspases 8 and 10 contain long pro-domains with duplicated death effector domains (DEDs), as well as their corresponding cysteine protease catalytic domains. Thus, it appears that CASP8 and CASP10 have evolved by tandem gene duplication, much like the CASP1, CASP4 and CASP5 gene cluster on human chromosome 11q22.2-22.3.
Collapse
Affiliation(s)
- J Grenet
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis TN 38101, USA
| | | | | | | | | |
Collapse
|
222
|
Hengstschläger M, Hölzl G, Hengstschläger-Ottnad E. Different regulation of c-Myc- and E2F-1-induced apoptosis during the ongoing cell cycle. Oncogene 1999; 18:843-8. [PMID: 9989838 DOI: 10.1038/sj.onc.1202342] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The transcription factors c-Myc and E2F-1 have been shown to harbour both mitogenic and apoptotic properties. Both factors have been implicated in the regulation of the transition from the G1 phase to the S phase in the mammalian cell cycle. However, whether cell death triggered by these molecules is dependent on the cell's position in the ongoing cell cycle remained elusive. Using centrifugal elutriation we here show for the first time that c-Myc induces apoptosis in G1 and in G2 phase, whereas E2F-1-induced apoptosis specifically occurs in G1. S phase cells are resistant to cell death triggered by these factors. We demonstrate that this is not a general phenomenon, since S phase cells are susceptible to apoptosis induced by treatment with actinomycin D and to the anti-apoptotic activity of Bcl-2. Our data indicate that S phase cells harbour specific protective activities against c-Myc- and E2F-1-induced apoptosis. Our results demonstrate that these transcription factors, although probably sharing specific apoptotic pathways, also take distinct routes to induce cell death and that apoptosis can occur at different phases of the cell cycle depending on the apoptotic stimulus. In this report we present the usefulness of a new approach to determine the regulation of apoptosis in the ongoing unperturbated cell cycle. This approach has clear implications for the identification of target genes involved in the regulation of cell death.
Collapse
Affiliation(s)
- M Hengstschläger
- Obstetrics and Gynecology, University of Vienna, Department of Prenatal Diagnosis and Therapy, Austria
| | | | | |
Collapse
|
223
|
Genestier L, Kasibhatla S, Brunner T, Green DR. Transforming growth factor beta1 inhibits Fas ligand expression and subsequent activation-induced cell death in T cells via downregulation of c-Myc. J Exp Med 1999; 189:231-9. [PMID: 9892606 PMCID: PMC2192981 DOI: 10.1084/jem.189.2.231] [Citation(s) in RCA: 145] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/1998] [Revised: 08/13/1998] [Indexed: 12/13/2022] Open
Abstract
Activation-induced cell death (AICD) is a process that regulates the size and the duration of the primary immune T cell response. In this report, we investigated the mechanisms involved in the regulation of AICD by transforming growth factor beta1 (TGF-beta1). We found that TGF-beta1 decreased apoptosis of human T cells or T cell hybridomas after activation by anti-CD3. This decrease was associated with inhibition of Fas (Apo-1/CD95) ligand (FasL) expression, whereas Fas signaling was not affected by TGF-beta1. In parallel, TGF-beta1 inhibited c-Myc expression in T cell hybridomas, and ectopic expression of a chimeric molecule composed of c-Myc and the steroid binding domain of the estrogen receptor (Myc-ER) blocked both the inhibition of FasL and the decrease of AICD induced by TGF-beta1, providing that 4-hydroxytamoxifen was present. These results identify one mechanism by which TGF-beta1 blocks AICD to allow the clonal expansion of effector T cells and the generation of memory T cells during immune responses.
Collapse
Affiliation(s)
- L Genestier
- Division of Cellular Immunology, La Jolla Institute for Allergy and Immunology, San Diego, California 92121, USA
| | | | | | | |
Collapse
|
224
|
Donzelli M, Bernardi R, Negri C, Prosperi E, Padovan L, Lavialle C, Brison O, Scovassi AI. Apoptosis-prone phenotype of human colon carcinoma cells with a high level amplification of the c-myc gene. Oncogene 1999; 18:439-48. [PMID: 9927200 DOI: 10.1038/sj.onc.1202309] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Although apoptosis can be induced by the enforced expression of exogenously introduced c-myc genes, it is not clear whether overexpression resulting from the amplification of the resident c-myc gene in tumor cells is sufficient to induce apoptosis. We have investigated the relationship between c-myc gene amplification and the propensity of tumor cells to undergo apoptosis, using the SW613-12A1 and SW613-B3 cell lines, which are representatives, respectively, of tumorigenic and non-tumorigenic clones isolated from the SW613-S human colon carcinoma cell line. Tumorigenic clones are characterized by a high level of amplification and expression of the c-myc gene, whereas cells of non-tumorigenic clones have a small number of copies and a lower level of expression of this gene. Analysis of c-myc mRNA level in cells cultured under low serum conditions indicated that the expression of the gene is tightly regulated by serum growth factors in non-tumorigenic B3 cells, whereas it is poorly regulated in tumorigenic 12A1 cells, the level of mRNAs remaining relatively high in serum-starved 12A1 cells. Under these conditions, 12A1 cells showed clear evidence of apoptosis, whereas B3 cells were completely refractory to the induction of apoptosis. Moreover, the study of cell lines derived from non-tumorigenic apoptosis-resistant clones following the introduction by transfection of exogenous c-myc gene copies showed that they have acquired an apoptosisprone phenotype. Altogether, our results strongly suggest that deregulated c-myc expression due to high-level amplification confers an apoptosis-prone phenotype to tumor cells. The possible consequences of these observations for cancer therapy are discussed.
Collapse
Affiliation(s)
- M Donzelli
- Istituto di Genetica Biochimica ed Evoluzionistica CNR, Pavia, Italy
| | | | | | | | | | | | | | | |
Collapse
|
225
|
Abstract
In the vertebrates, programmed cell death or apoptosis frequently involves the relocalization of mitochondrial cytochrome c to the cytoplasm. This prominent role in the regulation of apoptosis is in addition to the primary function of cytochrome c in the mitochondrial electron transport chain. These seemingly divergent roles become plausible when considering the symbiotic origin of the mitochondrion. Symbiosis involves conflicts between levels of selection, in this case between the primitive host cell and the protomitochondria. In an aerobic environment, selection on the protomitochondria may have favored routine manipulations of the host cell's phenotype using products and by-products of oxidative phosphorylation, in particular reactive oxygen species (ROS). Blocking the mitochondrial electron transport chain by removing cytochrome c enhances the production of ROS; thus cytochrome c release by protomitochondria may have altered the host cell's phenotype via enhanced ROS production. Subsequently, this signaling pathway may have been refined by selection so that cytochrome c itself became the trigger for changes in the host's phenotype. A mechanism of apoptosis in metazoans may thus be a vestige of evolutionary conflicts within the eukaryotic cell.
Collapse
Affiliation(s)
- N W Blackstone
- Department of Biological Sciences, Northern Illinois University, DeKalb 60115, USA
| | | |
Collapse
|
226
|
Wang W, Wykrzykowska J, Johnson T, Sen R, Sen J. A NF-κB/c- myc-Dependent Survival Pathway Is Targeted by Corticosteroids in Immature Thymocytes. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.1.314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Glucocorticoid hormones modulate T cell maturation in vivo. While low levels of hormones are required for appropriate T cell development, high levels of glucocorticoid hormones target immature developing thymocytes for cell death during systemic stress. In this report, we propose a molecular mechanism for the induction of apoptosis in CD4+CD8+ double-positive thymocytes by dexamethasone in vivo. Dexamethasone injection induced the expression of IκBα and IκBβ in thymocytes and down-regulated NF-κB DNA binding activated by intrathymic signals. Down-regulation of NF-κB DNA binding preceded cell death, suggesting that NF-κB may be important for the survival of immature thymocytes. In addition, ex vivo treatment of thymocyte single-cell suspension with dexamethasone accelerated p65/RelA down-regulation and cell death. Conversely, NF-κB induction diminished dexamethasone-induced death. Expression of the c-myc proto-oncogene, a NF-κB target, was also reduced in thymocytes of dexamethasone-treated animals, and ectopic transgenic expression of c-myc in mice provided partial rescue of double-positive thymocytes from dexamethasone mediated cell death. These observations suggest that viability of CD4+CD8+ thymocytes may be maintained by an NF-κB/c-myc-dependent pathway in vivo.
Collapse
Affiliation(s)
- Weihong Wang
- *Rosenstiel Research Center and Department of Biology, Brandeis University, Waltham, MA 02254; and
| | | | | | - Ranjan Sen
- *Rosenstiel Research Center and Department of Biology, Brandeis University, Waltham, MA 02254; and
| | - Jyoti Sen
- †Dana-Farber Cancer Institute, Boston, MA 02115
| |
Collapse
|
227
|
O'Connell J, Bennett MW, O'Sullivan GC, Collins JK, Shanahan F. The Fas counterattack: cancer as a site of immune privilege. IMMUNOLOGY TODAY 1999; 20:46-52. [PMID: 10081230 DOI: 10.1016/s0167-5699(98)01382-6] [Citation(s) in RCA: 174] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- J O'Connell
- Dept of Medicine, Cork University Hospital, Ireland.
| | | | | | | | | |
Collapse
|
228
|
Affiliation(s)
- C V Dang
- Department of Medicine, The Johns Hopkins Oncology Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
229
|
Shin EC, Shin JS, Park JH, Kim JJ, Kim H, Kim SJ. Expression of Fas-related genes in human hepatocellular carcinomas. Cancer Lett 1998; 134:155-62. [PMID: 10025875 DOI: 10.1016/s0304-3835(98)00251-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the tumors known to be resistant to Fas-mediated apoptosis. To elucidate the possible mechanisms of this resistance, we examined the mRNA transcripts of genes related to Fas in 10 hepatitis B virus (HBV)-associated HCC cell lines and six HBV-associated HCC tissues. Most of the HBV-associated HCC cell lines showed markedly decreased Fas expression. Some cell lines expressed FAP, a possible inhibitor of apoptosis, or showed downregulated expression of FADD or FLICE, molecules essential to Fas-mediated apoptosis. All six HBV-associated HCC tissues also showed decreased Fas expression compared with their non-malignant counterparts. Our data demonstrate that HBV-associated HCC showed a common defect in the expression of Fas, upregulation of FAP and downregulation of downstream molecules such as FADD and FLICE.
Collapse
Affiliation(s)
- E C Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
230
|
Clavería C, Albar JP, Serrano A, Buesa JM, Barbero JL, Martínez-A C, Torres M. Drosophila grim induces apoptosis in mammalian cells. EMBO J 1998; 17:7199-208. [PMID: 9857177 PMCID: PMC1171066 DOI: 10.1093/emboj/17.24.7199] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Genetic studies have shown that grim is a central genetic switch of programmed cell death in Drosophila; however, homologous genes have not been described in other species, nor has its mechanism of action been defined. We show here that grim expression induces apoptosis in mouse fibroblasts. Cell death induced by grim in mammalian cells involves membrane blebbing, cytoplasmic loss and nuclear DNA fragmentation. Grim-induced apoptosis is blocked by both natural and synthetic caspase inhibitors. We found that grim itself shows caspase-dependent proteolytic processing of its C-terminus in vitro. Grim-induced death is antagonized by bcl-2 in a dose-dependent manner, and neither Fas signalling nor p53 are required for grim pro-apoptotic activity. Grim protein localizes both in the cytosol and in the mitochondria of mouse fibroblasts, the latter location becoming predominant as apoptosis progresses. These results show that Drosophila grim induces death in mammalian cells by specifically acting on mitochondrial apoptotic pathways executed by endogenous caspases. These findings advance our knowledge of the mechanism by which grim induces apoptosis and show the conservation through evolution of this crucial programmed cell death pathway.
Collapse
Affiliation(s)
- C Clavería
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
231
|
Sata M, Walsh K. Endothelial cell apoptosis induced by oxidized LDL is associated with the down-regulation of the cellular caspase inhibitor FLIP. J Biol Chem 1998; 273:33103-6. [PMID: 9837872 DOI: 10.1074/jbc.273.50.33103] [Citation(s) in RCA: 119] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Fas (CD-95/APO-1) is a death receptor that initiates an apoptotic signal when activated by its ligand, FasL. Normal vascular endothelial cells are resistant to Fas-mediated apoptosis though they express both Fas and FasL. Oxidized low density lipoprotein (OxLDL) or lysophosphatidylcholine (LPC), a major component of OxLDL, induces endothelial cell suicide by sensitizing endothelial cells to Fas-mediated apoptosis. Here, we show that endothelial cell apoptosis by OxLDL and LPC-C16:0 was dose-dependent and correlated with down-regulation of FLICE-inhibitory protein (FLIP), an intracellular caspase inhibitor. FLIP down-regulation also occurred when endothelial cells were treated with toxic doses of LPC-C18:0 or minimally modified low density lipoprotein (LDL). In contrast, FLIP was not down-regulated by native LDL, acetylated LDL, LPC-C12:0, cholesterol, or 7-ketocholesterol, which are not toxic to endothelial cells. The cytotoxicity of oxidized lipids was reversed by transfecting endothelial cells with a FLIP expression plasmid. The results demonstrate, for the first time, FLIP regulation under conditions that lead to pathological tissue destruction.
Collapse
Affiliation(s)
- M Sata
- Division of Cardiovascular Research, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, Massachusetts 02135, USA
| | | |
Collapse
|
232
|
Nguyen M, Branton PE, Roy S, Nicholson DW, Alnemri ES, Yeh WC, Mak TW, Shore GC. E1A-induced processing of procaspase-8 can occur independently of FADD and is inhibited by Bcl-2. J Biol Chem 1998; 273:33099-102. [PMID: 9837871 DOI: 10.1074/jbc.273.50.33099] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Expression of the 243-residue form of the adenovirus E1A protein in the absence of other viral proteins triggers apoptosis by a pathway that requires p53. This pathway includes processing and activation of initiator procaspase-8, redistribution of cytochrome c, and activation of procaspase-3. Bcl-2 functions at or upstream of procaspase-8 processing to inhibit all of these events and prevent cell death. This contrasts with the anti-apoptotic influence of Bcl-2 family proteins in the cell death pathway induced by Fas ligand or tumor necrosis factor (TNF), in which Bcl-2 typically acts downstream of Fas/TNFR1-mediated activation of caspase-8. Moreover, E1A induces procaspase-8 processing and cell death in cells deleted of FADD, an adaptor protein critical for Fas/TNFR1 activation of caspase-8. The results indicate that E1A is capable of activating caspase-8 by a Bcl-2-inhibitable pathway that does not involve autocrine stimulation of FADD-dependent death receptor pathways.
Collapse
Affiliation(s)
- M Nguyen
- Department of Biochemistry, McIntyre Medical Sciences Building, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | | | | | | | | | | | | | | |
Collapse
|
233
|
He YW, Deftos ML, Ojala EW, Bevan MJ. RORgamma t, a novel isoform of an orphan receptor, negatively regulates Fas ligand expression and IL-2 production in T cells. Immunity 1998; 9:797-806. [PMID: 9881970 PMCID: PMC2776668 DOI: 10.1016/s1074-7613(00)80645-7] [Citation(s) in RCA: 207] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We have identified RORgamma t, a novel, thymus-specific isoform of the orphan nuclear receptor RORgamma that is expressed predominantly in CD4+ CD8+ double-positive thymocytes. Ectopic expression of RORgamma t protects T cell hybridomas from activation-induced cell death by inhibiting the upregulation of Fas ligand. Following hybridoma stimulation, RORgamma t also inhibits IL-2 production but does not affect the induction of Nur-77 and Egr-3 nor the upregulation of CD69. Both the ligand-binding and DNA-binding domains of RORgamma t are required for this effect. We propose that the role of RORgamma t expression in immature thymocytes is to inhibit Fas ligand expression and cytokine secretion following engagement of their TCR during positive or negative selection.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Cloning, Molecular
- DNA, Complementary
- Fas Ligand Protein
- Female
- Gene Expression Regulation
- Humans
- Hybridomas
- Interleukin-2/biosynthesis
- L Cells
- Membrane Glycoproteins/genetics
- Mice
- Mice, Inbred C57BL
- Molecular Sequence Data
- Nuclear Receptor Subfamily 1, Group F, Member 3
- Receptor-CD3 Complex, Antigen, T-Cell/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Retinoic Acid
- Receptors, Thyroid Hormone
- T-Lymphocytes/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- You-Wen He
- Department of Immunology and Howard Hughes Medical Institute University of Washington Seattle, Washington 98195
| | - Michael L. Deftos
- Department of Immunology and Howard Hughes Medical Institute University of Washington Seattle, Washington 98195
| | - Ethan W. Ojala
- Department of Immunology and Howard Hughes Medical Institute University of Washington Seattle, Washington 98195
| | - Michael J. Bevan
- Department of Immunology and Howard Hughes Medical Institute University of Washington Seattle, Washington 98195
| |
Collapse
|
234
|
Basu S, Bayoumy S, Zhang Y, Lozano J, Kolesnick R. BAD enables ceramide to signal apoptosis via Ras and Raf-1. J Biol Chem 1998; 273:30419-26. [PMID: 9804808 DOI: 10.1074/jbc.273.46.30419] [Citation(s) in RCA: 146] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Prior investigations document that proliferative signaling cascades, under some circumstances, initiate apoptosis, although mechanisms that dictate the final outcome are largely unknown. In COS-7 cells, ceramide signals Raf-1 activation through Ras (Zhang, Y., Yao, B., Delikat, S., Bayoumy, S., Lin, X. H., Basu, S., McGinley, M., Chan-Hui, P. Y., Lichenstein, H., and Kolesnick, R. (1997) Cell 89, 63-72), but not apoptosis. However, expression of small amounts of the pro-apoptotic Bcl-2 family member, BAD, conferred ceramide-induced apoptosis onto COS-7 cells. Ceramide signaled apoptosis in BAD-expressing cells by a pathway involving sequentially kinase suppressor of Ras (KSR)/ceramide-activated protein kinase, Ras, c-Raf-1, and MEK1. Downstream, this pathway linked to BAD dephosphorylation at serine 136 by prolonged inactivation of Akt/PKB. Further, mutation of BAD at serine 136 abrogated ceramide signaling of apoptosis. The present study indicates that when ceramide signals through the Ras/Raf cascade, the availability of a single target, BAD, may dictate an apoptotic outcome.
Collapse
Affiliation(s)
- S Basu
- Laboratory of Signal Transduction, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | | | | | | | | |
Collapse
|
235
|
Fearnhead HO, Rodriguez J, Govek EE, Guo W, Kobayashi R, Hannon G, Lazebnik YA. Oncogene-dependent apoptosis is mediated by caspase-9. Proc Natl Acad Sci U S A 1998; 95:13664-9. [PMID: 9811857 PMCID: PMC24876 DOI: 10.1073/pnas.95.23.13664] [Citation(s) in RCA: 142] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Understanding how oncogenic transformation sensitizes cells to apoptosis may provide a strategy to kill tumor cells selectively. We previously developed a cell-free system that recapitulates oncogene dependent apoptosis as reflected by activation of caspases, the core of the apoptotic machinery. Here, we show that this activation requires a previously identified apoptosis-promoting complex consisting of caspase-9, APAF-1, and cytochrome c. As predicted by the in vitro system, preventing caspase-9 activation blocked drug-induced apoptosis in cells sensitized by E1A, an adenoviral oncogene. Oncogenes, such as E1A, appear to facilitate caspase-9 activation by several mechanisms, including the control of cytochrome c release from the mitochondria.
Collapse
Affiliation(s)
- H O Fearnhead
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | | | | | | | | | | | | |
Collapse
|
236
|
ElShamy WM, Fridvall LK, Ernfors P. Growth arrest failure, G1 restriction point override, and S phase death of sensory precursor cells in the absence of neurotrophin-3. Neuron 1998; 21:1003-15. [PMID: 9856457 DOI: 10.1016/s0896-6273(00)80619-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
More than half of the dorsal root ganglion (DRG) neurons are lost by excessive cell death coinciding with precursor proliferation and cell cycle exit in neurotrophin-3 null mutant (NT-3-/-) mice. We find that in the absence of NT-3, sensory precursor cells fail to arrest the cell cycle, override the G1 phase restriction point, and die by apoptosis in S phase, which can be prevented in vivo by a cell cycle blocker. Uncoordinated cell cycle reentry is preceded by a failure of nuclear N-myc downregulation and is paralleled by the activation of the full repertoire of G1 and S phase cell cycle proteins required for cell cycle entry. Our results provide evidence for novel activity of neurotrophins in cell cycle control and point toward an N-myc sensitization to cell death in the nervous system that is under the control of NT-3.
Collapse
Affiliation(s)
- W M ElShamy
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | | | | |
Collapse
|
237
|
Abstract
Oxidized low density lipoproteins (OxLDL) promote chronic inflammatory responses in the vasculature that give rise to atherosclerotic plaques. Fas ligand (FasL) is naturally expressed on the vascular endothelium where it can induce apoptosis in Fas-expressing immune cells as they enter the vessel wall. Although vascular endothelial cells are normally resistant to Fas-mediated cell death, OxLDL were shown to induce apoptosis in cultured endothelial cells and endothelium of arterial explants by a process that could be inhibited with Fas L neutralizing antibodies. OxLDL-induced cell death was also reduced in the aortic endothelium cultured from gld (FasL-/-) and lpr (Fas-/-) mice as compared with wild-type mice. OxLDL acted by sensitizing endothelial cells to death signals from the Fas receptor. Thus, the ability of OxLDL to promote Fas-mediated endothelial cell suicide may be a feature that contributes to their atherogenicity.
Collapse
Affiliation(s)
- M Sata
- Division of Cardiovascular Research, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, Massachusetts 02135, USA
| | | |
Collapse
|
238
|
Soares MP, Muniappan A, Kaczmarek E, Koziak K, Wrighton CJ, Steinhäuslin F, Ferran C, Winkler H, Bach FH, Anrather J. Adenovirus-Mediated Expression of a Dominant Negative Mutant of p65/RelA Inhibits Proinflammatory Gene Expression in Endothelial Cells Without Sensitizing to Apoptosis. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.9.4572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
We hypothesized that blocking the induction of proinflammatory genes associated with endothelial cell (EC) activation, by inhibiting the transcription factor nuclear factor κB (NF-κB), would prolong survival of vascularized xenografts. Our previous studies have shown that inhibition of NF-κB by adenovirus-mediated overexpression of IκBα suppresses the induction of proinflammatory genes in EC. However, IκBα sensitizes EC to TNF-α-mediated apoptosis, presumably by suppressing the induction of the NF-κB-dependent anti-apoptotic genes A20, A1, manganese superoxide dismutase (MnSOD), and cellular inhibitor of apoptosis 2. We report here that adenovirus mediated expression of a dominant negative C-terminal truncation mutant of p65/RelA (p65RHD) inhibits the induction of proinflammatory genes, such as E-selectin, ICAM-1, VCAM-1, IL-8, and inducible nitric oxide synthase, in EC as efficiently as does IκBα. However, contrary to IκBα, p65RHD does not sensitize EC to TNF-α-mediated apoptosis although both inhibitors suppressed the induction of the anti-apoptotic genes A20, A1, and MnSOD equally well. We present evidence that this difference in sensitization of EC to apoptosis is due to the ability of p65RHD, but not IκBα, to inhibit the constitutive expression of c-myc, a gene involved in the regulation of TNF-α-mediated apoptosis. These data demonstrate that it is possible to block the expression of proinflammatory genes during EC activation by targeting NF-κB, without sensitizing EC to apoptosis and establishes the role of c-myc in controlling induction of apoptosis during EC activation. Finally, these data provide the basis for a potential approach to suppress EC activation in vivo in transgenic pigs to be used as donors for xenotransplantation.
Collapse
Affiliation(s)
- Miguel P. Soares
- Immunobiology Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Ashok Muniappan
- Immunobiology Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Elzbieta Kaczmarek
- Immunobiology Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Katarzyna Koziak
- Immunobiology Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Christopher J. Wrighton
- Immunobiology Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Florence Steinhäuslin
- Immunobiology Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Christiane Ferran
- Immunobiology Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Hans Winkler
- Immunobiology Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Fritz H. Bach
- Immunobiology Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Josef Anrather
- Immunobiology Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
239
|
Greil R, Egle A, Villunger A. On the role and significance of Fas (Apo-1/CD95) ligand (FasL) expression in immune privileged tissues and cancer cells using multiple myeloma as a model. Leuk Lymphoma 1998; 31:477-90. [PMID: 9922038 DOI: 10.3109/10428199809057607] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Our knowledge in immunology has been dramatically increased by several excellent investigations elucidating the role of the Fas (Apo-1/CD95) receptor/ligand (FasL) system in complex immunological processes such as the acquisition of self tolerance in T cells, progression of autoimmunity, clonal deletion of activated T cells, B-cell regulation and the establishment of "immune privileged" sites such as testis or retina. In addition to these regulatory immunological activities, Fas/FasL interaction was also shown to participate in active defense mechanisms of the host against infected or transformed cells thereby inducing apoptosis in target cells. However, the same mechanism seems also to be part of an escape strategy utilized by tumor cells in various neoplastic malignancies of both hematopoetic as also non-hematopoetic origin. We ourselves were able to demonstrate that neoplastic plasma cell lines, as well as native malignant myeloma cells constitutively express FasL mRNA and protein. The FasL molecule is functionally active and able to induce programmed cell death in Fas sensitive target T cells in vitro. These target T cells were protected from programmed cell death by preincubation of T cells with a Fas-blocking monoclonal antibody (mAb) or of myeloma cells with a FasL-neutralizing mAb. respectively. Furthermore, overexpression of the caspase inhibitor, cowpoxvirus protein CrmA, also protected target T cells from being killed by myeloma cells, identifying Fas/FasL mediated signaling as the effector pathway utilized by malignant plasma cells. Our observations strongly suggest the engagement of Fas/FasL interaction in the escape strategy of this malignancy. The molecular basis of this evasive mechanism differs in essential respects from those described in melanoma, lung cancer, hepatocellular carcinoma, or astrocytoma, since downregulation of Fas or instrinsic insensitivity towards Fas-mediated signaling were not prerequisites for the occurrence of this phenomenon in Fas-sensitive multiple myeloma cell lines. However, myeloma cell lines resisted cocultivation with FasL-expressing target T cells in vitro. The aim of this review is to discuss the role of Fas/FasL interaction in the establishment of malignant disease, in the light of our findings on myeloma cells and also by drawing upon similar observations of other investigators on different kinds of tumor cells and cell lines and further to consider its possible relevance in formulating novel approaches to cancer therapy.
Collapse
Affiliation(s)
- R Greil
- Dept. of Internal Medicine, University of Innsbruck Medical School, Austria
| | | | | |
Collapse
|
240
|
Rayet B, Lopez-Guerrero JA, Rommelaere J, Dinsart C. Induction of programmed cell death by parvovirus H-1 in U937 cells: connection with the tumor necrosis factor alpha signalling pathway. J Virol 1998; 72:8893-903. [PMID: 9765434 PMCID: PMC110306 DOI: 10.1128/jvi.72.11.8893-8903.1998] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/1998] [Accepted: 07/24/1998] [Indexed: 11/20/2022] Open
Abstract
The human promonocytic cell line U937 undergoes apoptosis upon treatment with tumor necrosis factor alpha (TNF-alpha). This cell line has previously been shown to be very sensitive to the lytic effect of the autonomous parvovirus H-1. Parvovirus infection leads to the activation of the CPP32 ICE-like cysteine protease which cleaves the enzyme poly(ADP-ribose)polymerase and induces morphologic changes that are characteristic of apoptosis in a way that is similar to TNF-alpha treatment. This effect is also observed when the U937 cells are infected with a recombinant H-1 virus which expresses the nonstructural (NS) proteins but in which the capsid genes are replaced by a reporter gene, indicating that the induction of apoptosis can be assigned to the cytotoxic nonstructural proteins in this cell system. The c-Myc protein, which is overexpressed in U937 cells, is rapidly downregulated during infection, in keeping with a possible role of this product in mediating the apoptotic cell death induced by H-1 virus infection. Interestingly, four clones (designated RU) derived from the U937 cell line and selected for their resistance to H-1 virus (J. A. Lopez-Guerrero et al., Blood 89:1642-1653, 1997) failed to decrease c-Myc expression upon treatment with differentiation agents and also resisted the induction of cell death after TNF-alpha treatment. Our data suggest that the RU clones have developed defense strategies against apoptosis, either by their failure to downregulate c-Myc and/or by activating antiapoptotic factors.
Collapse
Affiliation(s)
- B Rayet
- Angewandte Tumorvirologie, Abteilung F0100, Deutsches Krebsforschungszentrum, and Virologie Appliquée à l'Oncologie (Unité INSERM 375), D-69009 Heidelberg, Germany
| | | | | | | |
Collapse
|
241
|
Bennett M, Macdonald K, Chan SW, Luzio JP, Simari R, Weissberg P. Cell surface trafficking of Fas: a rapid mechanism of p53-mediated apoptosis. Science 1998; 282:290-3. [PMID: 9765154 DOI: 10.1126/science.282.5387.290] [Citation(s) in RCA: 515] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
p53 acts as a tumor suppressor by inducing both growth arrest and apoptosis. p53-induced apoptosis can occur without new RNA synthesis through an unknown mechanism. In human vascular smooth muscle cells, p53 activation transiently increased surface Fas (CD95) expression by transport from the Golgi complex. Golgi disruption blocked both p53-induced surface Fas expression and apoptosis. p53 also induced Fas-FADD binding and transiently sensitized cells to Fas-induced apoptosis. In contrast, lpr and gld fibroblasts were resistant to p53-induced apoptosis. Thus, p53 can mediate apoptosis through Fas transport from cytoplasmic stores.
Collapse
Affiliation(s)
- M Bennett
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK
| | | | | | | | | | | |
Collapse
|
242
|
Abstract
Studies of the biology of the IL-2 receptor have played a major part in establishing several of the fundamental principles that govern our current understanding of immunology. Chief among these is the contribution made by lymphokines to regulation of the interactions among vast numbers of lymphocytes, comprising a number of functionally distinct lineages. These soluble mediators likely act locally, within the context of the microanatomic organization of the primary and secondary lymphoid organs, where, in combination with signals generated by direct membrane-membrane interactions, a wide spectrum of cell fate decisions is influenced. The properties of IL-2 as a T-cell growth factor spawned the view that IL-2 worked in vivo to promote clonal T-cell expansion during immune responses. Over time, this singular view has suffered from increasing appreciation that the biologic effects of IL-2R signals are much more complex than simply mediating T-cell growth: depending on the set of conditions, IL-2R signals may also promote cell survival, effector function, and apoptosis. These sometimes contradictory effects underscore the fact that a diversity of intracellular signaling pathways are potentially activated by IL-2R. Furthermore, cell fate decisions are based on the integration of multiple signals received by a lymphocyte from the environment; IL-2R signals can thus be regarded as one input to this integration process. In part because IL-2 was first identified as a T-cell growth factor, the major focus of investigation in IL-R2 signaling has been on the mechanism of mitogenic effects in cultured cell lines. Three critical events have been identified in the generation of the IL-2R signal for cell cycle progression, including heterodimerization of the cytoplasmic domains of the IL-2R beta and gamma(c) chains, activation of the tyrosine kinase Jak3, and phosphorylation of tyrosine residues on the IL-2R beta chain. These proximal events led to the creation of an activated receptor complex, to which various cytoplasmic signaling molecules are recruited and become substrates for regulatory enzymes (especially tyrosine kinases) that are associated with the receptor. One intriguing outcome of the IL-2R signaling studies performed in cell lines is the apparent functional redundancy of the A and H regions of IL-2R beta, and their corresponding downstream pathways, with respect to the proliferative response. Why should the receptor complex induce cell proliferation through more than one mechanism or pathway? One possibility is that this redundancy is an unusual property of cultured cell lines and that primary lymphocytes require signals from both the A and the H regions of IL-2R beta for optimal proliferative responses in vivo. An alternative possibility is that the A and H regions of IL-2R beta are only redundant with respect to proliferation and that each region plays a unique and essential role in regulating other aspects of lymphocyte physiology. As examples, the A or H region could prove to be important for regulating the sensitivity of lymphocytes to AICD or for promoting the development of NK cells. These issues may be resolved by reconstituting IL-2R beta-/-mice with A-and H-deleted forms of the receptor chain and analyzing the effect on lymphocyte development and function in vivo. In addition to the redundant nature of the A and H regions, there remains a large number of biochemical activities mediated by the IL-2R for which no clear physiological role has been identified. Therefore, the circumstances are ripe for discovering new connections between molecular signaling events activated by the IL-2R and the regulation of immune physiology. Translating biochemical studies of Il-2R function into an understanding of how these signals regulate the immune system has been facilitated by the identification of natural mutations in IL-2R components in humans with immunodeficiency and by the generation of mice with targeted mutations in these gen
Collapse
Affiliation(s)
- B H Nelson
- Virginia Mason Research Center, Seattle, Washington 98101, USA
| | | |
Collapse
|
243
|
Abstract
Fas (Apo-1/CD95) is a cell-surface protein that is responsible for initiating a cascade of proteases (caspases) culminating in apoptotic cell death in a variety of cell types. The function of the Fas/FasL system in the dampening of immune responses to infectious agents through the autocrine deletion of activated T cells has been well documented. More recently, it has been proposed that tumor cells express FasL, presumably to avoid immune detection. In this review, we focus on the role of the interaction of Fas and FasL in the modulation of antitumor responses. We critically examine the evidence that FasL is expressed by tumor cells and explore alternative explanations for the observed phenomena in vitro and in vivo. By reviewing data that we have generated in our laboratory as well as reports from the literature, we will argue that the Fas/FasL system is a generalized mechanism used in an autocrine fashion to regulate cell survival and expansion in response to environmental and cellular cues. We propose that FasL expression by tumor cells, when present, is indicative of a perturbed balance in the control of proliferation while "immune privilege" is established by "suicide" of activated antitumor T cells, a form of activation-induced cell death.
Collapse
Affiliation(s)
- D B Chappell
- The Howard Hughes Medical Institute-National Institutes of Health Research Scholars Program, Bethesda MD 20814, USA
| | | |
Collapse
|
244
|
Fanidi A, Hancock DC, Littlewood TD. Suppression of c-Myc-induced apoptosis by the Epstein-Barr virus gene product BHRF1. J Virol 1998; 72:8392-5. [PMID: 9733891 PMCID: PMC110225 DOI: 10.1128/jvi.72.10.8392-8395.1998] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/1998] [Accepted: 07/13/1998] [Indexed: 11/20/2022] Open
Abstract
Constitutive expression of the c-myc proto-oncogene in growth factor-deprived fibroblasts promotes proliferation and induces apoptosis. In these cells, apoptosis can be inhibited by survival factors such as insulin-like growth factor I or the bcl-2 proto-oncogene product. Deregulated c-Myc expression is a common feature in Epstein-Barr virus-positive Burkitt's lymphoma in which the c-myc gene is reciprocally translocated and placed under the control of one of the immunoglobulin loci. BHRF1 is an Epstein-Barr virus protein expressed early in the lytic cycle. BHRF1 is a member of the Bcl-2 family and has been shown to suppress apoptosis and to increase cell survival in different settings. In the present study, we report that BHRF1 inhibits c-Myc-induced apoptosis which occurs in the absence of survival factors. It does not, however, affect the capacity of c-Myc to promote cell growth. These findings demonstrate that BHRF1 has not only structural but also functional similarities to Bcl-2.
Collapse
Affiliation(s)
- A Fanidi
- Biochemistry of the Cell Nucleus, Imperial Cancer Research Fund Laboratories, London WC2A 3PX, United Kingdom.
| | | | | |
Collapse
|
245
|
Abstract
Wound healing involves a series of rapid increases in specific cell populations that prepare the wound for repair, deposit new matrices and finally, mature the wound. Upon completing their tasks, these specific cell types must be eliminated from the wound prior to the progression to the next phase of healing. The most logical method of cellular down-regulation is through apoptosis. Apoptosis allows for the eliminations of entire populations without tissue damage or an inflammatory response. This review discusses which cells dominate the various phases of tissue repair and how the cellular pattern may vary after differing types of injury. The potential mechanisms involved in the down-regulation of inflammation and fibrosis are also covered. The studies that support the hypothesis that apoptosis is involved in the regulation of wound healing are discussed. The evidence supporting potential cell signals involved in the induction of apoptosis in tissue repair are examined. Finally, the review ends with a presentation of how dysregulation of apoptosis can lead to pathologic forms of healing such as excessive scarring and fibrosis. By understanding the mechanisms controlling apoptosis and tissue repair, one may eventually develop therapeutic modalities to minimize scarring, a final pathway for many disease processes.
Collapse
Affiliation(s)
- D G Greenhalgh
- Department of Surgery, Shriners Hospitals for Children, Northern California, Sacramento, USA.
| |
Collapse
|
246
|
Abstract
The MYC proto-oncogene has long been implicated in the control of normal cell growth and its deregulation is associated with the development of neoplasia. The MYC protein has a well-established role as a component of signal-transduction pathways promoting both proliferation and apoptosis. Because signalling pathways that drive cell death and cell proliferation are so tightly coupled, a synergy between genetic lesions leading to suppression of cell death and those promoting cell proliferation is observed during carcinogenesis. We discuss such synergy with respect to the cooperating oncogenes MYC, RAS and BCL2.
Collapse
Affiliation(s)
- A O Hueber
- Biochemistry of the Cell Nucleus Laboratory, Imperial Cancer Research Fund, London, UK.
| | | |
Collapse
|
247
|
Güzey M, Sattler C, DeLuca HF. Combinational effects of vitamin D3 and retinoic acid (all trans and 9 cis) on proliferation, differentiation, and programmed cell death in two small cell lung carcinoma cell lines. Biochem Biophys Res Commun 1998; 249:735-44. [PMID: 9731207 DOI: 10.1006/bbrc.1998.9186] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effects of a combination of vitamin D3 [1,25(OH)2D3] and retinoic acid (RA) on proliferation, differentiation, and apoptosis of the human small cell lung carcinoma (SCLC) cell lines NCI-H82 and NCI-H209 were evaluated. Cell proliferation was inhibited by 1,25(OH)2D3 and RA alone. The combination of 1,25(OH)2D3 and the cis form of retinoic acid resulted in an additive decrease in cell proliferation and the induction of apoptosis in various concentrations. Moreover, 3H-thymidine incorporation was inhibited and the number of viable cells was decreased. The characteristics of the apoptotic cells were examined and confirmed by morphologic analysis, light and electron microscopy, and fluorescence detection. It was concluded that 1,25(OH)2D3 and RA exert additive effects on the inhibition of proliferation and the induction of apoptosis in both the NCI-H82 and the NCI-H209 SCLC cell lines. This finding has important implications for the use of retinoids and 1,25(OH)2D3 in cancer prevention and in the therapy of small cell lung carcinoma.
Collapse
Affiliation(s)
- M Güzey
- TUBITAK-MAM, Research Institute for Genetic Engineering and Biotechnology, Gebze, Turkey.
| | | | | |
Collapse
|
248
|
Abstract
In multicellular organisms, mutations in somatic cells affecting critical genes that regulate cell proliferation and survival cause fatal cancers. Repair of the damage is one obvious option, although the relative inconsequence of individual cells in metazoans means that it is often a "safer" strategy to ablate the offending cell. Not surprisingly, corruption of the machinery that senses or implements DNA damage greatly predisposes to cancer. Nonetheless, even when oncogenic mutations do occur, there exist potent mechanisms that limit the expansion of affected cells by suppressing their proliferation or triggering their suicide. Growing understanding of these innate mechanisms is suggesting novel therapeutic strategies for cancer.
Collapse
Affiliation(s)
- G Evan
- Imperial Cancer Research Fund Laboratories, 44, Lincoln's Inn Fields, London WC2A 3PX, UK
| | | |
Collapse
|
249
|
Marthyn P, Beuscart A, Coll J, Moreau-Gachelin F, Righi M. DMSO reduces CSF-1 receptor levels and causes apoptosis in v-myc immortalized mouse macrophages. Exp Cell Res 1998; 243:94-100. [PMID: 9716453 DOI: 10.1006/excr.1998.4149] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have investigated the antiproliferative potentialof dimethyl sulfoxide (DMSO) on v-myc immortalized mouse macrophages on account of the cytotoxic effect induced by DMSO on myeloid cells. DMSO caused significant apoptosis in two immortalized macrophage celllines constitutively secreting colony-stimulating factor 1 (CSF-1). In contrast to the results described for mouse erythroleukemia cells, DMSO did not markedly decrease the level of the Spi-1/PU.1 transcription factor. However, DMSO caused a specific reduction in the protein level of the CSF-1 receptor (CSF-1R) compared to the FcgammaRIIIA immunoglobulin receptor, v-myc, and beta-actin proteins. To investigate if the level of CSF-1R might inversely correlate with DMSO-induced cell death, we derived a macrophage culture (named DN-11) that could be cultured in the presence of DMSO. Immunoblot analysis of DN-11, grown with or without DMSO, revealed significant amounts of CSF-1R under both conditions, suggesting a pivotal role for CSF-1R in the survival of DMSO-treated macrophages. Therefore, in these cells, DMSO seems to trigger apoptosis by interrupting an autocrine survival loop involving the CSF-1 receptor.
Collapse
Affiliation(s)
- P Marthyn
- Centre of Cellular and Molecular Pharmacology, Milan, I-20129, Italy
| | | | | | | | | |
Collapse
|
250
|
Fecho K, Bentley SA, Cohen PL. Mice deficient in fas ligand (gld) or fas (lpr) show few alterations in granulopoiesis. Cell Immunol 1998; 188:19-32. [PMID: 9743554 DOI: 10.1006/cimm.1998.1339] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Evidence exists that the life span of mature, circulating neutrophils is influenced by apoptosis induced by interactions between Fas ligand (FasL) and Fas (CD95). However, the role of FasL/Fas-mediated apoptosis in granulopoiesis has not been explored. The present study assessed differences in granulopoiesis between normal (B6) mice and mice carrying mutations in the genes for FasL (B6/gld) or Fas (B6/lpr). Granulopoiesis was examined by quantitating mature granulocytes in the blood, committed myeloid progenitor cells (or colony-forming units; CFU) in the bone marrow (BM), and granulocyte lineage cells in the BM. The present study also characterized through flow cytometry the ability of GR-1(+) granulocyte lineage cells from B6, B6/gld, and B6/lpr mice to undergo spontaneous apoptosis in vitro. In comparison to B6 mice, B6/gld mice (but not B6/lpr mice) showed small, but significant increases in the number and percentage of blood granulocytes and in the number of myeloid CFU. However, the number and percentage of GR-1(+) granulocyte lineage cells in the BM were similar in the three strains. The rate of spontaneous apoptosis of GR-1(+) granulocyte lineage cells also did not differ between B6, B6/gld, and B6/lpr mice. In B6 and B6/gld mice, Fas expression on granulocyte lineage cells was downregulated in conjunction with a decrease in forward-angle light scatter (fsc) and externalization of phosphatidylserine (PS), two measures of apoptosis. These results suggest that FasL-Fas interactions play only a minor role in modulating numbers of committed myeloid progenitor cells and the size of the peripheral pool of mature granulocytes. Interactions between FasL and Fas do not influence the size of the BM pool of granulocyte lineage cells or the ability of those cells to undergo spontaneous apoptosis.
Collapse
Affiliation(s)
- K Fecho
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599-7280, USA
| | | | | |
Collapse
|