151
|
Abstract
Cellular caspase-8 (FLICE)-like inhibitory protein (cFLIP) was originally identified as an inhibitor of death-receptor signalling through competition with caspase-8 for recruitment to FAS-associated via death domain (FADD). More recently, it has been determined that both cFLIP and caspase-8 are required for the survival and proliferation of T cells following T-cell-receptor stimulation. This paradoxical finding launched new investigations of how these molecules might connect with signalling pathways that link to cell survival and growth following antigen-receptor activation. As discussed in this Review, insight gained from these studies indicates that cFLIP and caspase-8 form a heterodimer that ultimately links T-cell-receptor signalling to activation of nuclear factor-kappaB through a complex that includes B-cell lymphoma 10 (BCL-10), mucosa-associated-lymphoid-tissue lymphoma-translocation gene 1 (MALT1) and receptor-interacting protein 1 (RIP1).
Collapse
Affiliation(s)
- Ralph C Budd
- Immunobiology Program, Department of Medicine, The University of Vermont College of Medicine, Burlington, Vermont 50405, USA
| | | | | |
Collapse
|
152
|
Arechiga AF, Bell BD, Solomon JC, Chu IH, Dubois CL, Hall BE, George TC, Coder DM, Walsh CM. Cutting edge: FADD is not required for antigen receptor-mediated NF-kappaB activation. THE JOURNAL OF IMMUNOLOGY 2006; 175:7800-4. [PMID: 16339514 DOI: 10.4049/jimmunol.175.12.7800] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recently, it has been demonstrated that stimulated T cells bearing defects in caspase-8 fail to promote nuclear shuttling of NF-kappaB complexes. Such cells display strikingly similar proliferative and survival defects as T cells lacking Fas-associated death domain protein (FADD) function. We characterized NF-kappaB signaling in T cells bearing a dominant-negative FADD transgene (FADDdd). Whereas FADDdd T cells displayed proliferative defects following activation, these were not a consequence of aberrant NF-kappaB signaling, as measured by IKK/IkappaB phosphorylation and IkappaB degradation. There were no appreciable defects in nuclear translocation of p65/Rel using ImageStream, a flow-based imaging cytometer. Pretreatment with benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone, a potent caspase inhibitor, also failed to impede canonical NF-kappaB signaling. Secretion of IL-2 and up-regulation of various activation markers occurred normally. Thus, FADD does not play an essential role in NF-kappaB activation, suggesting an alternative route by which this adaptor promotes the clonal expansion of T cells.
Collapse
Affiliation(s)
- Adrian F Arechiga
- Center for Immunology and Department of Molecular Biology & Biochemistry, University of California, Irvine. CA 92697, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Kalamvoki M, Georgopoulou U, Mavromara P. The NS5A protein of the hepatitis C virus genotype 1a is cleaved by caspases to produce C-terminal-truncated forms of the protein that reside mainly in the cytosol. J Biol Chem 2006; 281:13449-13462. [PMID: 16517592 DOI: 10.1074/jbc.m601124200] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The nonstructural 5A (NS5A) protein of the hepatitis C virus (HCV) is a multifunctional protein that is implicated in viral replication and pathogenesis. We report here that NS5A of HCV-1a is cleaved at multiple sites by caspase proteases in transfected cells. Two cleavage sites at positions Asp154 and 248DXXD251 were mapped. Cleavage at Asp154 has been previously recognized as one of the caspase cleavage sites for the NS5A protein of HCV genotype 1b (1, 2) and results in the production of a 17-kDa fragment. The sequence 248DXXD251 is a novel caspase recognition motif for NS5A and is responsible for the production of a 31-kDa fragment. Furthermore, we show that Arg217 is implicated in the production of the previously described 24-kDa product, whose accumulation is affected by both calpain and caspase inhibitors. We also showed that caspase-mediated cleavage occurs in the absence of exogenous proapoptotic stimuli and is not related to the accumulation of the protein in the endoplasmic reticulum. Interestingly, our data indicate that NS5A is targeted by at least two different caspases and suggest that caspase 6 is implicated in the production of the 17-kDa fragment. Most importantly, we report that, all the detectable NS5A fragments following caspase-mediated cleavage are C-terminal-truncated forms of NS5A and are mainly localized in the cytosol. Thus, in sharp contrast to the current view we found no evidence supporting a role for caspase-mediated cleavage in the transport of the NS5A protein to the nucleus, which could lead to transcriptional activation.
Collapse
Affiliation(s)
- Maria Kalamvoki
- Molecular Virology Laboratory, Hellenic Pasteur Institute, 115 21 Athens, Greece
| | - Urania Georgopoulou
- Molecular Virology Laboratory, Hellenic Pasteur Institute, 115 21 Athens, Greece
| | - Penelope Mavromara
- Molecular Virology Laboratory, Hellenic Pasteur Institute, 115 21 Athens, Greece.
| |
Collapse
|
154
|
Sabbagh L, Bourbonnière M, Denis F, Sékaly RP. Cloning and Functional Characterization of the Murine Caspase-3 Gene Promoter. DNA Cell Biol 2006; 25:104-15. [PMID: 16460234 DOI: 10.1089/dna.2006.25.104] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Several studies have shown that the levels of caspase-3 are upregulated under different conditions of apoptosis. Previously, we have shown that activation of T cells through the TCR leads to the upregulation of caspase-3 levels. These findings highlight the importance of regulating the expression of caspase-3 in order to prevent premature cell death. To better understand the regulation of the caspase-3 gene, a portion of the 5'- untranslated region was cloned, sequenced, and characterized. The segment of the 5'-flanking region of the caspase-3 gene was also cloned upstream of a luciferase reporter gene, demonstrating that this fragment contains promoter activity. Higher luciferase expression was found with several of the promoter deletion constructs in Jurkat T cells but not the mouse Neuro-2A neuroblastoma cell line, suggesting the presence of a T-cell-specific regulated region. The importance of these sequences is further supported by the genomic organization of the human and mouse caspase-3 promoter regions. These findings demonstrated that the -2245/+14 region of the caspase-3 promoter shows constitutive levels of expression, and that several regions of the promoter play a role in basal regulation. Finally, some of the conserved transcription factor binding sites identified between the human and mouse promoters appear to play an important role in lymphoid cells.
Collapse
Affiliation(s)
- Laurent Sabbagh
- Laboratoire d'Immunologie, Centre de Recherche du CHUM, Campus St.-Luc, Pavillon Edouard-Asselin, 264 Boulevard René Lévesque Est, #1370D, Montreal, Quebec H3C 3J7, Canada
| | | | | | | |
Collapse
|
155
|
Maksimow M, Söderström TS, Jalkanen S, Eriksson JE, Hänninen A. Fas costimulation of naive CD4 T cells is controlled by NF-kappaB signaling and caspase activity. J Leukoc Biol 2005; 79:369-77. [PMID: 16330535 DOI: 10.1189/jlb.0505238] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Fas ligation induces apoptosis of activated T cells via the caspase cascade but can also mediate costimulatory signals to naïve T cells at the time of activation. We have previously shown that Fas ligation of naïve CD4 T cells activated by dendritic cells induces death or accelerates their proliferation and increases interferon-gamma (IFN-gamma) production. To understand this costimulation, we investigated the roles of caspases and nuclear factor (NF)-kappaB in survival and proliferation of responding T cells. Fas ligation increased caspase-3 and -8 activities during T cell activation, irrespective of cell fate. The accelerated proliferation induced by Fas ligation could be reduced by selective inhibition of both caspases. Inhibition of NF-kappaB simultaneously with Fas ligation inhibited the increased IFN-gamma production and caused uniform death of all responding T cells. Thus, Fas-mediated costimulation of naïve CD4 T cells is driven by active caspases, and NF-kappaB acts as a dominant survival-supporting factor of Fas-costimulated cells containing high levels of activated caspase-8 and -3.
Collapse
|
156
|
Abstract
Apoptosis is a highly regulated process of cell deletion and plays a fundamental role in the maintenance of tissue homeostasis in the adult organism. Numerous studies in recent years have revealed that apoptosis is a constitutive suicide programme expressed in most, if not all cells, and can be triggered by a variety of extrinsic and intrinsic signals. Many human diseases can be attributed directly or indirectly to a derangement of apoptosis, resulting in either cell accumulation, in which cell eradication or cell turnover is impaired, or cell loss, in which the apoptotic programme is inadvertently triggered. In addition, defective macrophage engulfment and degradation of cell corpses may also contribute to a dysregulation of tissue homeostasis. An increased understanding of the signalling pathways that govern the execution of apoptosis and the subsequent clearance of dying cells may thus yield novel targets for therapeutic intervention in a wide range of human maladies.
Collapse
Affiliation(s)
- B Fadeel
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | | |
Collapse
|
157
|
|
158
|
Oliver L, Vallette FM. The role of caspases in cell death and differentiation. Drug Resist Updat 2005; 8:163-70. [PMID: 15946892 DOI: 10.1016/j.drup.2005.05.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2005] [Revised: 05/21/2005] [Accepted: 05/24/2005] [Indexed: 12/19/2022]
Abstract
The complexity, redundancy and interdependence of the biological systems involved in tumour response to different treatments hamper progress towards developing specific and effective therapies. In addition, the many and even contradictory roles played by certain key proteins can significantly amend our view on tumourigenesis. The role of caspases in the modulation of cell death and differentiation is a prominent example of such a complexity. Here we focus on the role of caspases in apoptotic cell death, mainly in haematological malignancies, tumourigenesis, sepsis, T-cell proliferation and cell differentiation.
Collapse
Affiliation(s)
- Lisa Oliver
- Equipe 4 Labellisée Ligue contre le Cancer, UMR 601 INSERM/Université de Nantes, 9 Quai Moncousu, 44035 Nantes Cedex 01, France.
| | | |
Collapse
|
159
|
Krishnan S, Kiang JG, Fisher CU, Nambiar MP, Nguyen HT, Kyttaris VC, Chowdhury B, Rus V, Tsokos GC. Increased caspase-3 expression and activity contribute to reduced CD3zeta expression in systemic lupus erythematosus T cells. THE JOURNAL OF IMMUNOLOGY 2005; 175:3417-23. [PMID: 16116236 DOI: 10.4049/jimmunol.175.5.3417] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
T cells isolated from patients with systemic lupus erythematosus (SLE) express low levels of CD3zeta-chain, a critical molecule involved in TCR-mediated signaling, but the involved mechanisms are not fully understood. In this study we examined caspase-3 as a candidate for cleaving CD3zeta in SLE T cells. We demonstrate that SLE T cells display increased expression and activity of caspase-3. Treatment of SLE T cells with the caspase-3 inhibitor Z-Asp-Glu-Val-Asp-FMK reduced proteolysis of CD3zeta and enhanced its expression. In addition, Z-Asp-Glu-Val-Asp-FMK treatment increased the association of CD3zeta with lipid rafts and simultaneously reversed the abnormal lipid raft preclustering, heightened TCR-induced calcium responses, and reduced the expression of FcRgamma-chain exclusively in SLE T cells. We conclude that caspase-3 inhibitors can normalize SLE T cell function by limiting the excessive digestion of CD3zeta-chain and suggest that such molecules can be considered in the treatment of this disease.
Collapse
Affiliation(s)
- Sandeep Krishnan
- Department of Cellular Injury, Walter Reed Army Institute of Research, Silver Spring, MD 20910-7500, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Fischer H, Rossiter H, Ghannadan M, Jaeger K, Barresi C, Declercq W, Tschachler E, Eckhart L. Caspase-14 but not caspase-3 is processed during the development of fetal mouse epidermis. Differentiation 2005; 73:406-13. [PMID: 16316411 DOI: 10.1111/j.1432-0436.2005.00046.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The activation of caspases is a central step in apoptosis and may also be critical for terminal differentiation of epidermal keratinocytes (KC). In particular, caspase-3 has been implicated in the differentiation of embryonic KC as well as in programmed cell death of KC, and caspase-14 has been suggested to function in the formation or homeostasis of the stratum corneum (SC). To test the putative roles of these proteases, we determined their expression level and activation status during development of fetal mouse epidermis. The level of procaspase-3 did not change significantly during epidermal development, and enzyme activation was undetectable at any timepoint investigated. Despite the lack of active caspase-3, the newly formed stratum granulosum and the regressing periderm contained cells positive in the terminal deoxynucleotidyl transferase-mediated fluorescein-dUTP nick end labeling assay, indicating that nuclear DNA was degraded without activation of caspase-3, thereby arguing against a proteolytic function of caspase-3 in embryonic KC differentiation. By contrast, caspase-14 increased in abundance from embryonic day 14.5 (E14.5) onwards and consistently localized to the suprabasal layers of fetal epidermis. The caspase-14 pro-enzyme was processed into its catalytic subunits, a step required for enzyme activity, on day E17.5, coinciding with SC formation. Thus, processing of procaspase-14 is not confined to air-exposed mature skin but also occurs during epidermal development in utero. In summary, this study demonstrates that caspase-14, but not caspase-3 activation coincides temporally and spatially with embryonic KC differentiation, suggesting a role for caspase-14 in terminally differentiated KC.
Collapse
Affiliation(s)
- Heinz Fischer
- Department of Dermatology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
161
|
Bantel H, Beikler T, Flemmig TF, Schulze-Osthoff K. Caspase activation is involved in chronic periodontitis. FEBS Lett 2005; 579:5559-64. [PMID: 16213496 DOI: 10.1016/j.febslet.2005.09.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Revised: 07/12/2005] [Accepted: 09/13/2005] [Indexed: 11/26/2022]
Abstract
Periodontitis, a common infectious disease, is initiated by various gram-negative bacteria and characterized by the destruction of the periodontal tissue. Here, we investigated the role of caspases, intracellular proteases that are the key mediators of apoptosis. We show that activation of caspase-3 and caspase-7 is considerably enhanced in gingival tissue from patients with periodontitis. We also demonstrate in in vitro experiments that various periodontopathic bacteria exert a direct growth-suppressing effect and, moreover, can trigger a host-mediated cytotoxic activity involving the CD95 death receptor. Our data suggest that caspase activation is a prominent feature in periodontitis-associated tissue injury.
Collapse
Affiliation(s)
- Heike Bantel
- Institute of Molecular Medicine, University of Düsseldorf, Germany
| | | | | | | |
Collapse
|
162
|
Santambrogio L, Potolicchio I, Fessler SP, Wong SH, Raposo G, Strominger JL. Involvement of caspase-cleaved and intact adaptor protein 1 complex in endosomal remodeling in maturing dendritic cells. Nat Immunol 2005; 6:1020-8. [PMID: 16170319 DOI: 10.1038/ni1250] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2004] [Accepted: 08/09/2005] [Indexed: 01/01/2023]
Abstract
The involvement of the tetrameric adaptor protein 1 (AP-1) complex in protein sorting in intracellular compartments is not yet completely defined. Here we report that in immature dendritic cells, the beta1- and gamma-subunits of AP-1 underwent caspase 3-catalyzed cleavage in their hinge regions, resulting in removal of the C-terminal 'ear' domains. Cleavage was inhibited by lipopolysaccharide or caspase inhibitors, each of which led to maturation of the dendritic cells, demonstrated by endosomal remodeling and an increase in surface expression of peptide-loaded major histocompatibility complex class II. Overexpression of similarly truncated AP-1 together with 'silencing' of the endogenous genes in immature dendritic cells did not compromise delivery of major histocompatibility complex class II invariant chain to endosomal compartments. However, after lipopolysaccharide-induced maturation, overexpression of truncated AP-1 and 'silencing' of endogenous genes did result in the anomalous surface accumulation of invariant chain and the peptide-editing molecule H2-DM. Thus, at least one function for intact AP-1 is to retain some proteins in endosomes during the dendritic cell maturation process in which others are allowed to egress to the cell surface.
Collapse
Affiliation(s)
- Laura Santambrogio
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | |
Collapse
|
163
|
Siegmund D, Wicovsky A, Schmitz I, Schulze-Osthoff K, Kreuz S, Leverkus M, Dittrich-Breiholz O, Kracht M, Wajant H. Death receptor-induced signaling pathways are differentially regulated by gamma interferon upstream of caspase 8 processing. Mol Cell Biol 2005; 25:6363-79. [PMID: 16024776 PMCID: PMC1190324 DOI: 10.1128/mcb.25.15.6363-6379.2005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
FasL and gamma interferon (IFN-gamma) are produced by activated T cells and NK cells and synergistically induce apoptosis. Although both cytokines can also elicit proinflammatory responses, a possible cross talk of these ligands with respect to nonapoptotic signaling has been poorly addressed. Here, we show that IFN-gamma sensitizes KB cells for apoptosis induction by facilitating death-inducing signaling complex (DISC)-mediated caspase 8 processing. Moreover, after protection against death receptor-induced apoptosis by caspase inhibition or Bcl2 overexpression, IFN-gamma also sensitized for Fas- and TRAIL death receptor-mediated NF-kappaB activation leading to synergistic upregulation of a variety of proinflammatory genes. In contrast, Fas-mediated activation of JNK, p38, and p42/44 occurred essentially independent from IFN-gamma sensitization, indicating that the apoptosis- and NF-kappaB-related FasL-IFN-gamma cross talk was not due to a simple global enhancement of Fas signaling. Overexpression of FLIP(L) and FLIP(S) inhibited Fas- as well as TRAIL-mediated NF-kappaB activation and apoptosis induction in IFN-gamma-primed cells suggesting that both responses are coregulated at the level of the DISC.
Collapse
Affiliation(s)
- Daniela Siegmund
- Department of Molecular Internal Medicine, Medical Polyclinic, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Kim HS, Lee MS. Essential role of STAT1 in caspase-independent cell death of activated macrophages through the p38 mitogen-activated protein kinase/STAT1/reactive oxygen species pathway. Mol Cell Biol 2005; 25:6821-33. [PMID: 16024814 PMCID: PMC1190352 DOI: 10.1128/mcb.25.15.6821-6833.2005] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Unlike other immune cells, activation of macrophages by stimulating agents, such as lipopolysaccharide (LPS), confers significant resistance to many apoptotic stimuli, but the underlying mechanism of this phenomenon remains largely unknown. Here, we demonstrate that LPS-induced early caspase activation is essential for macrophage survival because blocking caspase activation with a pancaspase inhibitor (zVAD [benzyloxycarbonyl-Val-Ala-Asp]) rapidly induced death of activated macrophages. This type of death process by zVAD/LPS was principally mediated by intracellular generation of superoxide. STAT1 knockout macrophages demonstrated profoundly decreased superoxide production and were resistant to treatment with zVAD/LPS, indicating the crucial involvement of STAT1 in macrophage death by zVAD/LPS. STAT1 level and activity were reciprocally regulated by caspase activation and were associated with cell death. Activation of STAT1 was critically dependent upon serine phosphorylation induced by p38 mitogen-activated protein kinase (MAPK) because a p38 MAPK inhibitor nullified STAT1 serine phosphorylation, reactive oxygen species (ROS) production, and macrophage death by zVAD/LPS. Conversely, p38 MAPK activation was dependent upon superoxide and was also nullified in STAT1 knockout macrophages, probably due to impaired generation of superoxide. Our findings collectively indicate that STAT1 signaling modulates intracellular oxidative stress in activated macrophages through a positive-feedback mechanism involving the p38 MAPK/STAT1/ROS pathway, which is interrupted by caspase activation. Furthermore, our study may provide significant insights in regards to the unanticipated critical role of STAT1 in the caspase-independent death pathway.
Collapse
Affiliation(s)
- Hun Sik Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong, Kangnam-ku, Seoul 135-710, South Korea
| | | |
Collapse
|
165
|
Launay S, Hermine O, Fontenay M, Kroemer G, Solary E, Garrido C. Vital functions for lethal caspases. Oncogene 2005; 24:5137-48. [PMID: 16079910 DOI: 10.1038/sj.onc.1208524] [Citation(s) in RCA: 152] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Caspases are a family of cysteine proteases expressed as inactive zymogens in virtually all animal cells. These enzymes play a central role in most cell death pathways leading to apoptosis but growing evidences implicate caspases also in nonapoptotic functions. Several of these enzymes, activated in molecular platforms referred to as inflammasomes, play a role in innate immune response by processing some of the cytokines involved in inflammatory response. Caspases are requested for terminal differentiation of specific cell types, whether this differentiation process leads to enucleation or not. These enzymes play also a role in T and B lymphocyte proliferation and, in some circumstances, appear to be cytoprotective rather than cytotoxic. These pleiotropic functions implicate caspases in the control of life and death but the fine regulation of their dual effect remains poorly understood. The nonapoptotic functions of caspases implicate that cells can restrict the proteolytic activity of these enzymes to selected substrates. Deregulation of the pathways in which caspases exert these nonapoptotic functions is suspected to play a role in the pathophysiology of several human diseases.
Collapse
Affiliation(s)
- Sophie Launay
- INSERM U-517, IFR100, Faculty of Medicine, 7 Boulevard Jeanne d'Arc, 21033 Dijon, France
| | | | | | | | | | | |
Collapse
|
166
|
Paananen A, Järvinen K, Sareneva T, Salkinoja-Salonen MS, Timonen T, Hölttä E. Valinomycin-induced apoptosis of human NK cells is predominantly caspase independent. Toxicology 2005; 212:37-45. [PMID: 15876477 DOI: 10.1016/j.tox.2005.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2005] [Revised: 03/31/2005] [Accepted: 04/04/2005] [Indexed: 11/25/2022]
Abstract
Human NK cells are sensitive to the exogenous toxic compound valinomycin. This toxin, produced by Streptomyces griseus in moisture damaged buildings, induces apoptosis by dissipating the membrane potential in mitochondria. In this paper, we show that valinomycin-induced apoptosis involves two different pathways in human NK cells: the predominant one is caspase-3 independent and the other caspase-3 dependent. Resting human NK cells were found to contain high amounts of active caspase-3 as compared to the T cells in which high caspase-3 activity has been shown only after stimulation. Exposure to valinomycin did not alter the caspase-3 activity of human NK cells but induced nucleosomal fragmentation of DNA. General caspase inhibitor, Z-VAD-FMK, inhibited completely the caspase-3 activity, reduced DNA cleavage but did not prevent the spontaneous or valinomycin-induced apoptosis of NK cells. The endogenous high caspase-3 had only a slight effect on the major functions of human NK cells, i.e. cytotoxicity or gamma-IFN production, giving us a reason to suspect that the biological role of caspase-3 in NK cells could be the elimination of potentially harmful NK clones through apoptosis.
Collapse
Affiliation(s)
- Auli Paananen
- Department of Pathology, Haartman Institute, University of Helsinki, P.O. Box 21, FIN-00014 Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
167
|
Cohen LY, Bourbonnière M, Sabbagh L, Bouchard A, Chew T, Jeannequin P, Lazure C, Sékaly RP. Notch1 antiapoptotic activity is abrogated by caspase cleavage in dying T lymphocytes. Cell Death Differ 2005; 12:243-54. [PMID: 15650752 DOI: 10.1038/sj.cdd.4401568] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Excessive signaling via the Notch1 receptor inhibits apoptosis in T lymphocytes. Since several antiapoptotic proteins are cleaved by caspases during cell death, we investigated whether Notch1 was a caspase substrate. Results demonstrate that the intracellular domain of Notch1 (NICD) is cleaved into six fragments during apoptosis in Jurkat cells or peripheral T lymphocytes. Notch1 cleavage is prevented by the caspase inhibitors DEVD-fmk and VEID-fmk or by Bcl-2 expression. Caspase-3 and caspase-6 cleave the NICD into six fragments using sites located within the NF-kappaB binding domain, the ankyrin repeats and the transactivation domain. Notch1 cleavage correlates with the loss of HES-1 expression in apoptotic T cells. Notch1 fragments cannot inhibit activation-induced cell death in a T-cell hybridoma, confirming the abrogation of Notch1 antiapoptotic activity by caspases. The ability of the NICD but not the fragments to antagonize Nur77 activity supports a role for this factor in Notch1 antiapoptotic function.
Collapse
Affiliation(s)
- L Y Cohen
- Laboratoire d'Immunologie, CR-CHUM, campus St-Luc, Pavillon Edouard-Asselin, 264 Bd. René Lévesque E., Montréal, Québec, Canada H2X 1P1.
| | | | | | | | | | | | | | | |
Collapse
|
168
|
Silva EM, Guillermo LVC, Ribeiro-Gomes FL, De Meis J, Pereira RMS, Wu Z, Calegari-Silva TC, Seabra SH, Lopes UG, Siegel RM, Dosreis GA, Lopes MF. Caspase-8 activity prevents type 2 cytokine responses and is required for protective T cell-mediated immunity against Trypanosoma cruzi infection. THE JOURNAL OF IMMUNOLOGY 2005; 174:6314-21. [PMID: 15879131 DOI: 10.4049/jimmunol.174.10.6314] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
During Trypanosoma cruzi infection, T cells up-regulate caspase-8 activity. To assess the role of caspase-8 in T cell-mediated immunity, we investigated the effects of caspase-8 inhibition on T cells in viral FLIP (v-FLIP) transgenic mice. Compared with wild-type controls, increased parasitemia was observed in v-FLIP mice infected with T. cruzi. There was a profound decrease in expansion of both CD4 and CD8 T cell subsets in the spleens of infected v-FLIP mice. We did not find differences in activation ratios of T cells from transgenic or wild-type infected mice. However, the numbers of memory/activated CD4 and CD8 T cells were markedly reduced in v-FLIP mice, possibly due to defective survival. We also found decreased production of IL-2 and increased secretion of type 2 cytokines, IL-4 and IL-10, which could enhance susceptibility to infection. Similar, but less pronounced, alterations were observed in mice treated with the caspase-8 inhibitor, zIETD. Furthermore, blockade of caspase-8 by zIETD in vitro mimicked the effects observed on T. cruzi infection in vivo, affecting the generation of activated/memory T cells and T cell cytokine production. Caspase-8 is also required for NF-kappaB signaling upon T cell activation. Blockade of caspase-8 by either v-FLIP expression or treatment with zIETD peptide decreased NF-kappaB responses to TCR:CD3 engagement in T cell cultures. These results suggest a critical role for caspase-8 in the establishment of T cell memory, cell signaling, and regulation of cytokine responses during protozoan infection.
Collapse
Affiliation(s)
- Elisabeth M Silva
- Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Bloco G, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Sabbagh L, Bourbonnière M, Sékaly RP, Cohen LY. Selective up-regulation of caspase-3 gene expression following TCR engagement. Mol Immunol 2005; 42:1345-54. [PMID: 15950730 DOI: 10.1016/j.molimm.2004.12.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2004] [Accepted: 12/19/2004] [Indexed: 01/12/2023]
Abstract
Activation-induced cell death (AICD) in T lymphocytes depends on the expression of Fas-ligand, which triggers the apoptotic process after binding to its receptor Fas. This leads to the activation of cysteine proteases of the caspase family and especially of caspase-3, a critical effector protein during AICD. We have previously observed the up-regulation of caspase-3 expression in effector but not memory T cells stimulated in vivo. In this study, we further characterized the regulation of caspase expression following T cell receptor (TCR) signaling and demonstrate that a three-fold increase in caspase-3 mRNA levels was observed by semi-quantitative and real-time RT-PCR analysis. Caspase-3 expression was selectively increased among five different caspases following TCR stimulation, as assessed by RNase protection assay. Real-time RT-PCR analysis demonstrated that a three-fold up-regulation in caspase-3 mRNA levels was observed following TCR triggering, whereas caspase-8 mRNA levels remained unchanged. The increase in caspase-3 mRNA levels occurred before cleavage and activation of caspase-3 and in the absence of apoptosis. TCR-mediated induction in caspase-3 expression was not dependent on STAT1 activation, since following stimulation of KOX-14 cells the transcription factor was not phosphorylated. Together, these results show that TCR activation triggers the selective increase in caspase-3 mRNA levels, independently of caspase activity and the induction of apoptosis.
Collapse
Affiliation(s)
- Laurent Sabbagh
- Laboratoire d'Immunologie, Centre de Recherche du CHUM, Campus St. Luc, Pavillon Edouard-Asselin, 264 Boul. Rene Levesque Est #1307D, Montreal, Que., Canada H2X 1P1
| | | | | | | |
Collapse
|
170
|
Dohrman A, Russell JQ, Cuenin S, Fortner K, Tschopp J, Budd RC. Cellular FLIP Long Form Augments Caspase Activity and Death of T Cells through Heterodimerization with and Activation of Caspase-8. THE JOURNAL OF IMMUNOLOGY 2005; 175:311-8. [PMID: 15972663 DOI: 10.4049/jimmunol.175.1.311] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Caspase activity is required not only for the death of T cells, but also for their activation. A delicate balance of caspase activity is thus required during T cell activation at a level that will not drive cell death. How caspase activity is initiated and regulated during T cell activation is not known. One logical candidate for this process is cellular FLIP long form (c-FLIP(L)), because it can block caspase-8 recruitment after Fas (CD95) ligation as well as directly heterodimerize with and activate caspase-8. The current findings demonstrate that after T cell activation, caspase-8 and c-FLIP(L) associate in a complex enriched for active caspases. This occurs coincidently with the cleavage of two known caspase-8 substrates, c-FLIP(L) and receptor interacting protein 1. Caspase activity is higher in wild-type CD8(+) than CD4(+) effector T cells. Increased expression of c-FLIP(L) results in augmented caspase activity in resting and effector T cells to levels that provoke cell death, especially of the CD8 subset. c-FLIP(L) is thus not only an inhibitor of cell death by Fas, it can also act as a principal activator of caspases independently of Fas.
Collapse
Affiliation(s)
- Austin Dohrman
- Immunobiology Program, Department of Medicine, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | | | | | | | | | | |
Collapse
|
171
|
Dohrman A, Kataoka T, Cuenin S, Russell JQ, Tschopp J, Budd RC. Cellular FLIP (long form) regulates CD8+ T cell activation through caspase-8-dependent NF-kappa B activation. THE JOURNAL OF IMMUNOLOGY 2005; 174:5270-8. [PMID: 15843523 DOI: 10.4049/jimmunol.174.9.5270] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cellular FLIP long form (c-FLIP(L)) was originally identified as an inhibitor of Fas (CD95/Apo-1). Subsequently, additional functions of c-FLIP(L) were identified through its association with receptor-interacting protein (RIP)1 and TNFR-associated factor 2 to activate NF-kappaB, as well as by its association with and activation of caspase-8. T cells from c-FLIP(L)-transgenic (Tg) mice manifest hyperproliferation upon activation, although it was not clear which of the various functions of c-FLIP(L) was involved. We have further explored the effect of c-FLIP(L) on CD8(+) effector T cell function and its mechanism of action. c-FLIP(L)-Tg CD8(+) T cells have increased proliferation and IL-2 responsiveness to cognate Ags as well as to low-affinity Ag variants, due to increased CD25 expression. They also have a T cytotoxic 2 cytokine phenotype. c-FLIP(L)-Tg CD8(+) T cells manifest greater caspase activity and NF-kappaB activity upon activation. Both augmented proliferation and CD25 expression are blocked by caspase inhibition. c-FLIP(L) itself is a substrate of the caspase activity in effector T cells, being cleaved to a p43(FLIP) form. p43(FLIP) more efficiently recruits RIP1 than full-length c-FLIP(L) to activate NF-kappaB. c-FLIP(L) and RIP1 also coimmunoprecipitate with active caspase-8 in effector CD8(+) T cells. Thus, one mechanism by which c-FLIP(L) influences effector T cell function is through its activation of caspase-8, which in turn cleaves c-FLIP(L) to allow RIP1 recruitment and NF-kappaB activation. This provides a partial explanation of why caspase activity is required to initiate proliferation of resting T cells.
Collapse
Affiliation(s)
- Austin Dohrman
- Immunobiology Program, Department of Medicine, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | | | | | | | | | | |
Collapse
|
172
|
Vaisid T, Kosower NS, Barnoy S. Caspase-1 activity is required for neuronal differentiation of PC12 cells: cross-talk between the caspase and calpain systems. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1743:223-30. [PMID: 15843036 DOI: 10.1016/j.bbamcr.2005.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2004] [Revised: 01/11/2005] [Accepted: 01/11/2005] [Indexed: 01/29/2023]
Abstract
Previously, we have found that caspase-1 activity is increased during myoblast differentiation to myotubes. Here we show that caspase-1 activity is required for PC12 differentiation to neuronal-like cells. Caspase-1 is shown to be activated (by immunoblotting and by assessing activity in cell extracts) in the PC12 cells following the initial stage of differentiation. The inhibition of caspase-1 arrests PC12 cells at an intermediate stage of differentiation and prevents neurite outgrowth in these cells; the inhibition is reversed upon the removal of the inhibitor. Calpastatin (calpain endogenous specific inhibitor, and a known caspase substrate) is diminished at the later stages of PC12 cell differentiation, and diminution is prevented by caspase-1 inhibition. The degradation of fodrin (a known caspase and calpain substrate) is found in the advanced stage of differentiation. Caspase-1 has been implicated in the activation of proinflammatory cytokines, and in cell apoptosis. The involvement of caspase-1 in two distinct differentiation processes (myoblast fusion and neuronal differentiation of PC12 cells) indicates a function for this caspase in differentiation processes, and suggests some common mechanisms underlying caspase roles in such processes.
Collapse
Affiliation(s)
- T Vaisid
- Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel-Aviv University, Ramat-Aviv, Tel-Aviv 69978, Israel
| | | | | |
Collapse
|
173
|
Misra RS, Jelley-Gibbs DM, Russell JQ, Huston G, Swain SL, Budd RC. Effector CD4+ T cells generate intermediate caspase activity and cleavage of caspase-8 substrates. THE JOURNAL OF IMMUNOLOGY 2005; 174:3999-4009. [PMID: 15778357 PMCID: PMC4522921 DOI: 10.4049/jimmunol.174.7.3999] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Caspase-8 activation promotes cell apoptosis but is also essential for T cell activation. The extent of caspase activation and substrate cleavage in these divergent processes remains unclear. We show that murine effector CD4(+) T cells generated levels of caspase activity intermediate between unstimulated T cells and apoptotic populations. Both caspase-8 and caspase-3 were partially activated in effector T cells, which was reflected in cleavage of the caspase-8 substrates, c-FLIP(L), receptor interacting protein 1, and to a lesser extent Bid, but not the caspase-3 substrate inhibitor of caspase-activated DNase. Th2 effector CD4(+) T cells manifested more caspase activity than did Th1 effectors, and caspase blockade greatly decreased initiation of cell cycling. The current findings define the level of caspase activity and substrates during initiation of T cell cycling.
Collapse
Affiliation(s)
- Ravi S. Misra
- University of Vermont, Immunobiology Program, College of Medicine, Burlington, VT 05405
| | | | - Jennifer Q. Russell
- University of Vermont, Immunobiology Program, College of Medicine, Burlington, VT 05405
| | | | | | - Ralph C. Budd
- University of Vermont, Immunobiology Program, College of Medicine, Burlington, VT 05405
- Address correspondence and reprint requests to University of Vermont, Immunobiology Program, College of Medicine, 89 Beaumont Avenue, Given Building D305, Burlington, VT 05405.
| |
Collapse
|
174
|
Lemaire C, Godefroy N, Costina-Parvu I, Rincheval V, Renaud F, Trotot P, Bouleau S, Mignotte B, Vayssière JL. Caspase-9 can antagonize p53-induced apoptosis by generating a p76(Rb) truncated form of Rb. Oncogene 2005; 24:3297-308. [PMID: 15735701 DOI: 10.1038/sj.onc.1208493] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The tumor suppressor Rb (retinoblastoma protein) is known to regulate p53-dependent apoptosis, but the mechanisms involved are unclear. In a rat fibroblast model, we previously observed that caspase inhibition potentiates p53-dependent apoptosis and prevents the Rb cleavage associated with p53 activation. These results suggested that a caspase(s) can antagonize p53-mediated apoptosis via the production of a protective Rb truncated form. Here, we identify caspase-9 as the caspase that interferes, upstream of the mitochondrion, with p53-induced apoptosis in both immortalized and primary fibroblasts. This caspase can be detected as a p38 processed form in living cells, in the absence of apoptosome formation and apoptotic signal. We also provide evidence that the involvement of caspase-9 in a pre-mitochondrial protective pathway results from the previously undescribed cleavage of Rb, at a LExD site, into a p76(Rb) form, which antagonizes p53-induced apoptosis. These results establish that a truncated form of Rb can display an antiapoptotic activity, rather than just being a by-product of Rb degradation.
Collapse
Affiliation(s)
- Christophe Lemaire
- Laboratoire de Génétique et Biologie Cellulaire, CNRS FRE 2445, Université de Versailles/St Quentin-en-Yvelines, 45 avenue des Etats-Unis, 78035 Versailles cedex, France
| | | | | | | | | | | | | | | | | |
Collapse
|
175
|
Besnault-Mascard L, Leprince C, Auffredou MT, Meunier B, Bourgeade MF, Camonis J, Lorenzo HK, Vazquez A. Caspase-8 sumoylation is associated with nuclear localization. Oncogene 2005; 24:3268-73. [PMID: 15782135 DOI: 10.1038/sj.onc.1208448] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The cysteine protease caspase-8 plays a pivotal role in the initiation of different apoptotic pathways and controls the maturation and differentiation of various cell types including neurons, fibroblasts and lymphocytes. Specific substrates of caspase-8 are present in both the cytoplasm and the nucleus, which may determine the ultimate biological effect of caspase-8. However, the mechanisms regulating the cellular localization of caspase-8 are still unknown. We show here that, in contrast to other caspases such as caspase-9 and -3, caspase-8 can be sumoylated at lysine 156. This sumoylation (i) is associated with the nuclear localization of caspase-8 and (ii) did not impair caspase-8 activation.
Collapse
Affiliation(s)
- Laurence Besnault-Mascard
- INSERM U542, Hôpital Paul Brousse, Bâtiment Lavoisier, 14 Avenue Paul Vaillant Couturier, 94807 Villejuif cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
176
|
Sehra S, Patel D, Kusam S, Wang ZY, Chang CH, Dent AL. A role for caspases in controlling IL-4 expression in T cells. THE JOURNAL OF IMMUNOLOGY 2005; 174:3440-6. [PMID: 15749878 DOI: 10.4049/jimmunol.174.6.3440] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Although caspase activation is critical for T cell proliferation following activation, the role of caspases in T cell differentiation is unclear. In this study, we have examined the effect of inhibition of caspases on the process of Th1/Th2 differentiation. Naive CD4+ T cells activated under neutral differentiation conditions in the presence of the pan caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp (Z-VAD) fluoromethylketone showed increased Th2 cell differentiation concomitant with an up-regulation of GATA-3. Z-VAD induced optimal Th2 differentiation when T cells were stimulated under strong primary activation conditions. Treatment of naive CD4+ T cells with Z-VAD under strong activation conditions led to a 6-fold increase in IL-4 mRNA compared with control-treated T cells. The Z-VAD-induced increase in IL-4 transcription occurred within 24 h of activation and was independent of Stat6. IFN-gamma mRNA expression was not affected by Z-VAD at the 24-h time point. Z-VAD did not augment IL-4 expression from a committed Th2 cell, suggesting that caspases regulate IL-4 expression specifically during primary T cell activation. Z-VAD did not augment IL-12-driven Th1 differentiation. Activation of T cells in the presence of Z-VAD led to a specific increase in the expression of the transcription factor c-fos. Lastly, retrovirus-mediated expression of the antiapoptotic protein Bcl-2 resulted in an enhancement of Th2 cytokine expression, suggesting that inhibition of caspase activation by Bcl-2 can also modulate IL-4 expression. These findings reveal a novel regulatory mechanism of cytokine expression by caspases, and may explain how signaling pathways that inhibit apoptosis tend to promote Th2 differentiation.
Collapse
Affiliation(s)
- Sarita Sehra
- Department of Microbiology and Immunology, The Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | |
Collapse
|
177
|
Vaughan AT, Betti CJ, Villalobos MJ, Premkumar K, Cline E, Jiang Q, Diaz MO. Surviving apoptosis: a possible mechanism of benzene-induced leukemia. Chem Biol Interact 2005; 153-154:179-85. [PMID: 15935815 DOI: 10.1016/j.cbi.2005.03.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The pathological consequences resulting from deregulation of the apoptotic program include cancer (too little apoptosis) or diseases of cell deprivation, such as Alzheimer's (too much apoptosis). We have identified an additional pathology whereby cells reaching the earliest stage of chromatin cleavage have the potential to suppress apoptotic execution and survive. One specific cleavage event associated with this process is restricted to a location within the mixed lineage leukemia (MLL) gene at 11q23. The site of cleavage is consistent with the location where large, approximately 50 kbp loops of supercoiled DNA are attached to the nuclear matrix. Cells modified by this process generate MLL translocations, as shown by inverse PCR, that survive for days to weeks but which have no known relationship with clinical disease. Using a specific approach, cells stimulated by anti-CD95 antibody, a potent stimulator of the apoptotic program, facilitated creation of the MLL-AF9 fusion gene. Further, this rearrangement, which is commonly observed in patients with AML linked to exposure to cytotoxic agents, was efficiently transcribed in cells that were able to undergo cell division. These data are discussed in the context of benzene and benzene metabolite toxicity that impacts the process of apoptosis and is known to lead to leukemic disease.
Collapse
Affiliation(s)
- Andrew T Vaughan
- Department of Radiation Oncology, University of California, Davis, 4501 X Street, Suite G 126, Sacramento, CA 95817, USA.
| | | | | | | | | | | | | |
Collapse
|
178
|
Su H, Bidère N, Zheng L, Cubre A, Sakai K, Dale J, Salmena L, Hakem R, Straus S, Lenardo M. Requirement for caspase-8 in NF-kappaB activation by antigen receptor. Science 2005; 307:1465-8. [PMID: 15746428 DOI: 10.1126/science.1104765] [Citation(s) in RCA: 351] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Caspase-8, a proapoptotic protease, has an essential role in lymphocyte activation and protective immunity. We show that caspase-8 deficiency (CED) in humans and mice specifically abolishes activation of the transcription factor nuclear factor kappaB (NF-kappaB) after stimulation through antigen receptors, Fc receptors, or Toll-like receptor 4 in T, B, and natural killer cells. Caspase-8 also causes the alphabeta complex of the inhibitor of NF-kappaB kinase (IKK) to associate with the upstream Bcl10-MALT1 (mucosa-associated lymphatic tissue) adapter complex. Recruitment of the IKKalpha, beta complex, its activation, and the nuclear translocation of NF-kappaB require enzyme activity of full-length caspase-8. These findings thus explain the paradoxical association of defective apoptosis and combined immunodeficiency in human CED.
Collapse
Affiliation(s)
- Helen Su
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Oehme I, Neumann F, Bösser S, Zörnig M. Transgenic overexpression of the Caspase-8 inhibitor FLIPshortleads to impaired T cell proliferation and an increased memory T cell pool after staphylococcal enterotoxin B injection. Eur J Immunol 2005; 35:1240-9. [PMID: 15761846 DOI: 10.1002/eji.200425564] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The cellular homologues of the viral anti-apoptotic v-FLIP proteins exist as a long (c-FLIP(L)) and a short (c-FLIP(S)) splice variant. While c-FLIP(S) and v-FLIP are composed solely of two death effector domains, c-FLIP(L) contains an (inactive) caspase-like domain in addition to these two death effector domains, thereby structurally resembling pro-Caspase-8. Both c-FLIP(L) and c-FLIP(S) suppress apoptosis by inhibiting Caspase-8 activation, although at different levels of pro-Caspase-8 processing. To analyze the consequences of deregulated c-FLIP(S) expression in vivo, we established lck FLIP(S)-transgenic mice overexpressing the transgene in thymocytes and in mature T cells. As expected, CD95L-induced apoptosis was impaired in lck FLIP(S)-transgenic T cells, indicating the functionality of the FLIP(S) transgene. Remarkably, activation-induced cell death of transgenic T cells was unaffected, despite the observed inhibition of CD95-induced T cell death. Thymic and splenic cell numbers as well as CD4/CD8 cellularity were normal in lck FLIP(S)-transgenic animals, which in contrast to CD95-deficient mice do not accumulate Thy1(+) B220(+) CD4(-) CD8(-) peripheral T cells. c-FLIP(S) overexpression leads to a significant decrease in activation-induced T cell proliferation in vitro. Despite the capacity of FLIP(S) to inhibit CD95-induced apoptosis, T cell lymphomagenesis is not observed in lck FLIP(S)-transgenic mice. Interestingly, the Vbeta8(+) memory T cell pool is enlarged upon staphylococcal enterotoxin B injections, suggesting a specific in vivo function for FLIP(S) in the maintenance of restimulated T cells.
Collapse
Affiliation(s)
- Ina Oehme
- Georg-Speyer-Haus, Frankfurt, Germany
| | | | | | | |
Collapse
|
180
|
Fischer ANM, Herrera B, Mikula M, Proell V, Fuchs E, Gotzmann J, Schulte-Hermann R, Beug H, Mikulits W. Integration of Ras subeffector signaling in TGF-beta mediated late stage hepatocarcinogenesis. Carcinogenesis 2005; 26:931-42. [PMID: 15705598 DOI: 10.1093/carcin/bgi043] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Immortalized p19(ARF) null hepatocytes (MIM) feature a high degree of functional differentiation and are susceptible to transforming growth factor (TGF)-beta driven growth arrest and apoptosis. In contrast, polarized MIM hepatocytes expressing hyperactive Ha-Ras continue proliferation in cooperation with TGF-beta, and adopt an invasive phenotype by executing an epithelial to mesenchymal transition (EMT). In this study, we analyzed the involvement of Ras subeffectors in TGF-beta mediated hepatocellular EMT by employing MIM hepatocytes, which express Ras mutants allowing selective activation of either mitogen-activated protein kinase (MAPK) signaling (V12-S35) or phosphoinositide 3-OH (PI3)3 kinase (PI3K) signaling (V12-C40). We found that MAPK signaling in MIM-S35 hepatocytes was necessary and sufficient to promote resistance to TGF-beta mediated inhibition of proliferation in vitro and in vivo. MIM-S35 hepatocytes showed also PI3K activation during EMT, however, MAPK signaling on its own protected hepatocytes from apoptosis. Yet, MIM-C40 hepatocytes failed to form tumors and required additional MAPK stimulation to overcome TGF-beta mediated growth arrest. In vivo, the collaboration of MAPK signaling and TGF-beta activity drastically accelerated the cell-cycle progression of the hepatocytes, leading to vast tumor formation. From these data we conclude that MAPK is crucial for the cooperation with TGF-beta to regulate the proliferation as well as the survival of hepatocytes during EMT, and causes the fatal increase in hepatocellular tumor progression.
Collapse
Affiliation(s)
- Alexandra N M Fischer
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Borschke-Gasse 8a, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Peter ME, Legembre P, Barnhart BC. Does CD95 have tumor promoting activities? Biochim Biophys Acta Rev Cancer 2005; 1755:25-36. [PMID: 15907590 DOI: 10.1016/j.bbcan.2005.01.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2004] [Revised: 11/14/2004] [Accepted: 01/04/2005] [Indexed: 01/12/2023]
Abstract
CD95 (APO-1/Fas) is an important inducer of the extrinsic apoptosis signaling pathway and therapy induced apoptosis of many tumor cells has been linked to the activity of CD95. Changes in the expression of CD95 and/or its ligand CD95L are frequently found in human cancer. The downregulation or mutation of CD95 has been proposed as a mechanism by which cancer cells avoid destruction by the immune system through reduced apoptosis sensitivity. CD95 has therefore been viewed as a tumor suppressor. Furthermore, increased CD95L concentration in tumor patients has been linked to tumor cells killing infiltrating lymphocytes in a process called "the tumor counter-attack". Recent data have illuminated unknown activities of CD95 in tumor cells with downregulated or mutated CD95 in the presence of increased CD95L. Under these conditions the stimulation of CD95 signals nonapoptotic pathways, activating NF-kappaB and MAP kinases for example, which may result in the induction of tumorigenic or prosurvival genes. A new model of CD95 functions is proposed in which CD95 is converted from a tumor suppressor to a tumor promotor by a single point mutation in one of the CD95 alleles, a situation frequently found in advanced human cancer, resulting in apoptosis resistance and activation of tumorigenic pathways.
Collapse
Affiliation(s)
- Marcus E Peter
- The Ben May Institute for Cancer Research, The University of Chicago, Chicago, IL 60637, USA.
| | | | | |
Collapse
|
182
|
Abstract
AbstractAttempts at inducing allograft immune privilege by enforced Fas ligand expression have shown accelerated rejection mediated by neutrophils. While it has been proposed that Fas ligand was directly chemotactic toward neutrophils, several lines of evidence argue for an indirect recruitment mechanism. This question was addressed by using in vitro migration assays that used highly purified human leukocyte subsets. Granulocytes did not migrate in response to Fas engagement and required the presence of T cells expressing several natural killer (NK) cell markers. These rare CD8 memory T cells expressed T and NK cell markers and were not restricted to CD1d, showing that they are distinct from conventional natural killer T (NKT) cells. These cells were able to kill both NK-sensitive and -insensitive targets and secreted several CC and CXC chemokines active toward granulocytes, monocytes, and NK cells upon Fas engagement. Chemotactic factor release depended on caspase activity, in the absence of NKT cell apoptosis. The ability of CD1d-unrestricted NKT cells to recruit innate immune system cells might play a role in cancer cell eradication and contribute to inflammatory diseases.
Collapse
Affiliation(s)
- Martin Giroux
- Institut National de Recherche et de Sécurité-Institut Armand-Frappier, Laval, QC, Canada
| | | |
Collapse
|
183
|
Krakauer T. Caspase inhibitors attenuate superantigen-induced inflammatory cytokines, chemokines, and T-cell proliferation. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2005; 11:621-4. [PMID: 15138192 PMCID: PMC404582 DOI: 10.1128/cdli.11.3.621-624.2004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Proinflammatory cytokines mediate the toxic effect of superantigenic staphylococcal exotoxins (SE). A pan-caspase inhibitor suppressed SE-stimulated T-cell proliferation and the production of cytokines and chemokines by human peripheral blood mononuclear cells. These data suggest that caspase inhibitors may represent a novel therapeutic modality for treating SE-induced toxic shock.
Collapse
Affiliation(s)
- Teresa Krakauer
- Department of Immunology and Molecular Biology, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland 21702-5011, USA.
| |
Collapse
|
184
|
Tseveleki V, Bauer J, Taoufik E, Ruan C, Leondiadis L, Haralambous S, Lassmann H, Probert L. Cellular FLIP (long isoform) overexpression in T cells drives Th2 effector responses and promotes immunoregulation in experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2005; 173:6619-26. [PMID: 15557152 DOI: 10.4049/jimmunol.173.11.6619] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Cellular FLIP (c-FLIP) is an endogenous inhibitor of death receptor-induced apoptosis through the caspase 8 pathway. It is an NF-kappaB-inducible protein thought to promote the survival of T cells upon activation, and its down-regulation has been implicated in activation-induced cell death. We have generated transgenic mice overexpressing human c-FLIP long form (c-FLIP(L)) specifically in T cells using the CD2 promoter (TgFLIP(L)). TgFLIP(L) mice exhibit increased IgG1 production upon stimulation by a T cell-dependent Ag and a markedly enhanced contact hypersensitivity response to allergen. In addition to showing augmented Th2-type responses, TgFLIP(L) mice are resistant to the development of myelin oligodendrocyte glycoprotein 35-55 peptide-induced experimental autoimmune encephalomyelitis, a Th1-driven autoimmune disease. In vitro analyses revealed that T cells of TgFLIP(L) mice proliferate normally, but produce higher levels of IL-2 and show preferential maturation of Th2 cytokine-producing cells in response to antigenic stimulation. After adoptive transfer, these (Th2) cells protected wild-type recipient mice from experimental autoimmune encephalomyelitis induction. Our results show that the constitutive overexpression of c-FLIP(L) in T cells is sufficient to drive Th2 polarization of effector T cell responses and indicate that it might function as a key regulator of Th cell differentiation.
Collapse
MESH Headings
- Adjuvants, Immunologic/biosynthesis
- Adjuvants, Immunologic/genetics
- Adjuvants, Immunologic/physiology
- Adoptive Transfer
- Animals
- Antibodies, Monoclonal/pharmacology
- Autoantibodies/biosynthesis
- CASP8 and FADD-Like Apoptosis Regulating Protein
- CD3 Complex/immunology
- Cell Death/genetics
- Cell Death/immunology
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Dermatitis, Contact/immunology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Glycoproteins/immunology
- Humans
- Immunity, Innate
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Intracellular Signaling Peptides and Proteins/physiology
- Lymphocyte Activation
- Mice
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Mice, Transgenic
- Myelin-Oligodendrocyte Glycoprotein
- Peptide Fragments/immunology
- Protein Isoforms/biosynthesis
- Protein Isoforms/genetics
- Protein Isoforms/physiology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Th2 Cells/cytology
- Th2 Cells/immunology
- Th2 Cells/metabolism
- Th2 Cells/transplantation
- fas Receptor/physiology
Collapse
Affiliation(s)
- Vivian Tseveleki
- Laboratory of Molecular Genetics, Hellenic Pasteur Institute, National Center for Scientific Research Demokritos, Athens, Greece
| | | | | | | | | | | | | | | |
Collapse
|
185
|
Tacconi S, Perri R, Balestrieri E, Grelli S, Bernardini S, Annichiarico R, Mastino A, Caltagirone C, Macchi B. Increased caspase activation in peripheral blood mononuclear cells of patients with Alzheimer's disease. Exp Neurol 2005; 190:254-62. [PMID: 15473998 DOI: 10.1016/j.expneurol.2004.07.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2003] [Revised: 07/21/2004] [Accepted: 07/23/2004] [Indexed: 12/22/2022]
Abstract
In this study, we investigated whether alterations in the pattern of caspase activation could be found at the level of peripheral blood mononuclear cells (PBMCs) in patients with Alzheimer's disease (AD). The results showed that in experimental conditions resembling a physiological stimulation, there was a statistically significant increase in the enzymatic activity of caspase-3, caspase-8, and caspase-9 in PBMCs from a small, but well-characterized, cohort of sporadic AD patients compared to those from a comparable control group of aged adults (AA). This was accompanied by a parallel, early increase in the cleavage activity of the same caspases. The higher level of caspase activity in PBMCs from AD compared to AA was not associated with quantitative differences in cell subset profiles. Moreover, no increase in apoptosis level, in the same experimental conditions, was found in PBMCs from this cohort of AD patients compared to those from AA. Conversely, the higher proneness to caspase activation in PBMCs from AD patients in comparison with that from AA was associated with a higher proliferative response to PHA or CD3. These data show a new dysfunction in AD patients at the PBMCs level and suggest that increased proneness to caspase activation in lymphocytes could reflect an ongoing systemic response in neurodegenerative disease with pathogenetic implications.
Collapse
Affiliation(s)
- Santina Tacconi
- Department of Experimental Medicine and Biochemical Sciences, University Hospital of Tor Vergata, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
186
|
McLaughlin B. The kinder side of killer proteases: caspase activation contributes to neuroprotection and CNS remodeling. Apoptosis 2005; 9:111-21. [PMID: 15004508 PMCID: PMC2879070 DOI: 10.1023/b:appt.0000018793.10779.dc] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Caspases are a family of cysteine proteases that are expressed as inactive zymogens and undergo proteolytic maturation in a sequential manner in which initiator caspases cleave and activate the effector caspases 3, 6 and 7. Effector caspases cleave structural proteins, signaling molecules, DNA repair enzymes and proteins which inhibit apoptosis. Activation of effector, or executioner, caspases has historically been viewed as a terminal event in the process of programmed cell death. Emerging evidence now suggests a broader role for activated caspases in cellular maturation, differentiation and other non-lethal events. The importance of activated caspases in normal cell development and signaling has recently been extended to the CNS where these proteases have been shown to contribute to axon guidance, synaptic plasticity and neuroprotection. This review will focus on the adaptive roles activated caspases in maintaining viability, the mechanisms by which caspases are held in check so as not produce apoptotic cell death and the ramifications of these observations in the treatment of neurological disorders.
Collapse
Affiliation(s)
- B McLaughlin
- Department of Pharmacology, Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232-8548, USA.
| |
Collapse
|
187
|
Yang JY, Michod D, Walicki J, Murphy BM, Kasibhatla S, Martin SJ, Widmann C. Partial cleavage of RasGAP by caspases is required for cell survival in mild stress conditions. Mol Cell Biol 2005; 24:10425-36. [PMID: 15542850 PMCID: PMC529026 DOI: 10.1128/mcb.24.23.10425-10436.2004] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Tight control of apoptosis is required for proper development and maintenance of homeostasis in multicellular organisms. Cells can protect themselves from potentially lethal stimuli by expressing antiapoptotic factors, such as inhibitors of apoptosis, FLICE (caspase 8)-inhibitory proteins, and members of the Bcl2 family. Here, we describe a mechanism that allows cells to survive once executioner caspases have been activated. This mechanism relies on the partial cleavage of RasGAP by caspase 3 into an amino-terminal fragment called fragment N. Generation of this fragment leads to the activation of the antiapoptotic Akt kinase, preventing further amplification of caspase activity. Partial cleavage of RasGAP is required for cell survival under stress conditions because cells expressing an uncleavable RasGAP mutant cannot activate Akt, cannot prevent amplification of caspase 3 activity, and eventually undergo apoptosis. Executioner caspases therefore control the extent of their own activation by a feedback regulatory mechanism initiated by the partial cleavage of RasGAP that is crucial for cell survival under adverse conditions.
Collapse
Affiliation(s)
- Jiang-Yan Yang
- Department of Cellular Biology, Lausanne University, 1005 Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
188
|
Graham KL, Thibault DL, Steinman JB, Okeke L, Kao PN, Utz PJ. Granzyme B is dispensable for immunologic tolerance to self in a murine model of systemic lupus erythematosus. ACTA ACUST UNITED AC 2005; 52:1684-93. [PMID: 15934098 DOI: 10.1002/art.21092] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Proteolytic autoantigen cleavage by the serine protease granzyme B has been implicated in the development of systemic autoimmune disease; however, there has been no conclusive demonstration of a pathogenic role for granzyme B in autoimmunity. In this study, we evaluated the role of granzyme B in a murine model of autoimmunity. METHODS To identify potential novel granzyme B substrates, complementary DNAs encoding nuclear factor 45 (NF45) and NF90 were used to generate (35)S-methionine-labeled proteins by coupled in vitro transcription/translation. Radiolabeled proteins were then incubated with purified recombinant granzyme B or caspases, and the cleavage products were analyzed by autoradiography. We also immunized granzyme B-deficient and granzyme B-intact mice with the mineral oil pristane. Production of autoantibodies directed against granzyme B substrates in response to pristane was evaluated by Western blotting, immunoprecipitation, and enzyme-linked immunosorbent assay. RESULTS The double-stranded RNA-binding protein NF90 was identified as a novel substrate for caspases and granzyme B, both in vitro and in vivo. NF90 is uniquely cleaved by granzyme B in vitro; however, pristane immunization still induced anti-NF90 antibodies in granzyme B-deficient mice. Pristane-treated granzyme B-deficient mice also produced antibodies directed against the U1-70-kd antigen, a previously identified granzyme B substrate. Last, antibodies directed against U1-70 kd arose spontaneously in granzyme B-deficient mice. CONCLUSION These results demonstrate that granzyme B is not required for the production of autoantibodies directed against antigens that are granzyme B substrates in vitro. The data also suggest a protective role for this proapoptotic protease in systemic autoimmunity.
Collapse
Affiliation(s)
- Kareem L Graham
- Stanford University School of Medicine, Stanford, California, USA
| | | | | | | | | | | |
Collapse
|
189
|
Wong SH, Santambrogio L, Strominger JL. Caspases and nitric oxide broadly regulate dendritic cell maturation and surface expression of class II MHC proteins. Proc Natl Acad Sci U S A 2004; 101:17783-8. [PMID: 15598739 PMCID: PMC539763 DOI: 10.1073/pnas.0408229102] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The passage of dendritic cells (DC) from immature to terminally differentiated antigen-presenting cells is accompanied by numerous morphological, phenotypic, and functional changes. These changes include, for example, expression of "empty" class II MHC proteins (MHCII) at the surface in immature DC, whereas a much larger amount of peptide-loaded MHCII is expressed at the surface in mature DC. Here we show that, in cultured immature DC derived from murine bone-marrow precursors, a number of molecules involved in intracellular trafficking were present in a cleaved form, degraded by caspase-like proteases. Cleavage was either inhibited or reduced significantly during maturation of DC induced by either LPS and TNF-alpha or by peptides that inhibit caspase activities. Inducible nitric oxide (NO) synthetase up-regulated by LPS was essential for inhibiting the caspase-like activity during the maturation of DC. Moreover, treatment with LPS or caspase inhibitor resulted in expression of MHCII/peptide complexes at the cell surface. Thus, the alteration of the endosomal trafficking pathways during the development of DC that parallels the changes in surface expression of MHCII is regulated at least in part by the activities of caspases, inducible NO synthetase, and its product NO.
Collapse
Affiliation(s)
- Siew Heng Wong
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | |
Collapse
|
190
|
Weinberg A, Jesser RD, Edelstein CL, Bill JR, Wohl DA. Excess apoptosis of mononuclear cells contributes to the depressed cytomegalovirus-specific immunity in HIV-infected patients on HAART. Virology 2004; 330:313-21. [PMID: 15527856 DOI: 10.1016/j.virol.2004.09.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2004] [Revised: 09/01/2004] [Accepted: 09/15/2004] [Indexed: 12/21/2022]
Abstract
HIV-infected patients on highly active antiretroviral therapy (HAART) have persistently decreased cytomegalovirus (CMV)-specific proliferative responses [lymphocyte proliferation assay (LPA)] in spite of increases in CD4+ T cell counts. Here we demonstrate an association between apoptosis of unstimulated peripheral blood mononuclear cells (uPBMC) and decreased CMV-LPA. HAART recipients had more apoptosis of uPBMC than controls when measured by caspases 3, 8, and 9 activities and by annexin V binding. Patients with undetectable HIV replication maintained significantly higher apoptosis of CD4+ and CD14+ cells compared to controls. CMV-LPA decreased with higher apoptosis of uPBMC in patients only. This association was independent of CD4+ cell counts or HIV replication. Furthermore, rescuing PBMC from apoptosis with crmA, but not with TRAIL- or Fas-pathway blocking agents or with other caspase inhibitors, increased CMV-LPA in HAART recipients. This effect was not observed in uninfected controls, further indicating that the down regulatory effect of apoptosis on cell-mediated immunity (CMI) was specifically associated with the HIV-infected status.
Collapse
Affiliation(s)
- Adriana Weinberg
- Department of Pediatrics of the University of Colorado Health Sciences Center, Denver, CO 80220, USA.
| | | | | | | | | |
Collapse
|
191
|
Nasr R, El-Sabban ME, Karam JA, Dbaibo G, Kfoury Y, Arnulf B, Lepelletier Y, Bex F, de Thé H, Hermine O, Bazarbachi A. Efficacy and mechanism of action of the proteasome inhibitor PS-341 in T-cell lymphomas and HTLV-I associated adult T-cell leukemia/lymphoma. Oncogene 2004; 24:419-30. [PMID: 15543232 DOI: 10.1038/sj.onc.1208212] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
HTLV-I associated adult T-cell leukemia (ATL) and HTLV-I-negative peripheral T-cell lymphomas are associated with poor prognosis. Using pharmacological concentrations of the proteasome inhibitor PS-341, we demonstrate inhibition of cell proliferation and induction of apoptosis in fresh ATL cells, HTLV-I transformed and HTLV-I-negative malignant T cells, while normal resting or activated T lymphocytes were resistant. Combination of PS-341 and doxorubicin or etoposide resulted in an additive growth inhibition. In HTLV-I-negative malignant cells, PS-341 treatment significantly downregulated the antiapoptotic protein X-IAP and to a lesser extent c-IAP-1 and bcl-X(L) and resulted in caspase-dependent apoptosis. In HTLV-I transformed cells, the inhibition of the proteasomal degradation of Tax by PS-341 likely explains the relative protection of HTLV-I infected cells against caspase-dependent apoptosis. PS-341 treatment of these cells stabilized IkappaBalpha, IkappaBbeta, IkappaBvarepsilon, p21, p27 and p53 proteins and selectively inhibited Rel-A DNA binding NF-kappaB complexes. In both HTLV-I-positive and -negative cells, PS-341 treatment induced ceramide accumulation that correlated with apoptosis. We conclude that PS-341 affects multiple pathways critical for the survival of HTLV-I-positive and -negative malignant T cells supporting a potential therapeutic role for PS-341 in both ATL and HTLV-I-negative T-cell lymphomas, whether alone or in combination with chemotherapy.
Collapse
Affiliation(s)
- Rihab Nasr
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
192
|
Taylor SL, Weng SL, Fox P, Duran EH, Morshedi MS, Oehninger S, Beebe SJ. Somatic cell apoptosis markers and pathways in human ejaculated sperm: potential utility as indicators of sperm quality. ACTA ACUST UNITED AC 2004; 10:825-34. [PMID: 15465851 DOI: 10.1093/molehr/gah099] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In this study we extended earlier work to determine whether sperm respond to somatic cell apoptotic stimuli and whether apoptotic phenotypes are significant indicators of human sperm quality. We evaluated ejaculated sperm from fertile donors and subfertile patients following purification of fractions of high and low motility. In unstimulated conditions, caspase enzymatic activity was higher in motile fractions from subfertile patients than in donors, and was higher in low motility fractions from both groups. Staurosporine, but not a Fas ligand or H2O2, significantly increased caspase activity, but only in high motility fractions. Procaspase-3, -7 and -9 and low levels of active caspase-3, -7 and -9 were identified by immunoblot analysis. Apoptosis-inducing factor (AIF) was present in all samples but poly ADP-ribose polymerase-1 (PARP-1) was not detected. Phosphatidylserine translocation was significantly increased only with H2O2 treatment. In ejaculates of both subfertile and fertile men, we demonstrated the presence and activation of several proteins that are key constituents of apoptosis-related pathways in somatic cells, which may serve as markers for sperm quality.
Collapse
Affiliation(s)
- S L Taylor
- The Jones Institute for Reproductive Medicine, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | | | | | | | | | | | | |
Collapse
|
193
|
Sabbagh L, Kaech SM, Bourbonnière M, Woo M, Cohen LY, Haddad EK, Labrecque N, Ahmed R, Sékaly RP. The Selective Increase in Caspase-3 Expression in Effector but Not Memory T Cells Allows Susceptibility to Apoptosis. THE JOURNAL OF IMMUNOLOGY 2004; 173:5425-33. [PMID: 15494489 DOI: 10.4049/jimmunol.173.9.5425] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Caspases play a central role in T lymphocyte activation and death. We have demonstrated previously that caspase-3, an effector molecule for activation-induced cell death (AICD), is processed following T cell activation in the absence of apoptosis. We report in this study that caspase-3 mRNA levels were selectively increased in peripheral T cells, following Ag receptor-mediated activation. The up-regulation of caspase-3 mRNA was confined to cells in the early phases of the cell cycle (G0/G1) and was independent of IL-2 signaling. This increase led to the renewal of procaspase-3 as evidenced by a 6-fold up-regulation of the zymogen in nonapoptotic stimulated T cells. The increase of mRNA levels and of both the zymogen and the cleaved forms of caspase-3 was observed in in vivo stimulated Ag-specific effector, but not memory T cells, correlating with the enhanced susceptibility of effector T cells to AICD. Furthermore, we confirm that caspase-3 levels directly influence the sensitivity of activated T cells to apoptosis, as shown using T lymphocytes isolated from caspase-3 heterozygous and knockout mice. These findings indicate that the selective up-regulation of caspase-3 transcription is required to maintain the cytoplasmic levels of this protease, which control AICD and T cell homeostasis.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Apoptosis/immunology
- Caspase 3
- Caspases/biosynthesis
- Caspases/deficiency
- Caspases/genetics
- Caspases/metabolism
- Enzyme Activation/immunology
- Epitopes, T-Lymphocyte/immunology
- G1 Phase/immunology
- Immunity, Innate/genetics
- Immunologic Memory/genetics
- Interleukin-2/physiology
- Lymphocyte Activation/genetics
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- RNA, Messenger/biosynthesis
- Receptors, Antigen, T-Cell/immunology
- Resting Phase, Cell Cycle/immunology
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/enzymology
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/enzymology
- T-Lymphocytes, Regulatory/immunology
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Laurent Sabbagh
- Laboratory of Immunology, Université de Montréal, Montreal, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Dondi E, Roué G, Yuste VJ, Susin SA, Pellegrini S. A dual role of IFN-alpha in the balance between proliferation and death of human CD4+ T lymphocytes during primary response. THE JOURNAL OF IMMUNOLOGY 2004; 173:3740-7. [PMID: 15356120 DOI: 10.4049/jimmunol.173.6.3740] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Type I IFNs (IFN-alphabeta) enhance immune responses, notably T cell-mediated responses, in part by promoting the functional activities of dendritic cells. In this study, we analyzed the direct impact of IFN-alpha on proliferative and apoptotic signals upon in vitro activation of human naive CD4+ T lymphocytes. We demonstrate that IFN-alpha protects T cells from the intrinsic mitochondrial-dependent apoptosis early upon TCR/CD28 activation. IFN-alpha acts by delaying entry of cells into the G1 phase of the cell cycle, as well as by increasing Bcl-2 and limiting Bax activation. Later, upon activation, T cells that were exposed to IFN-alpha showed increased levels of surface Fas associated with partially processed caspase-8, a key component of the extrinsic apoptotic pathway. Caspase-8 processing was augmented furthermore by Fas ligation. Overall, these findings support a model whereby IFN-alpha favors an enhanced clonal expansion, yet it sensitizes cells to the Ag-induced cell death occurring at the end of an immune response. These observations point to a complex role of type I IFN in regulating the magnitude of proliferation and survival of naive CD4+ T cells during primary response and underline how crucial could be the timing of exposure to this cytokine.
Collapse
Affiliation(s)
- Elisabetta Dondi
- Unité de Signalisation des Cytokines, Institut Pasteur, Paris, France
| | | | | | | | | |
Collapse
|
195
|
Tawa P, Hell K, Giroux A, Grimm E, Han Y, Nicholson DW, Xanthoudakis S. Catalytic activity of caspase-3 is required for its degradation: stabilization of the active complex by synthetic inhibitors. Cell Death Differ 2004; 11:439-47. [PMID: 14713960 DOI: 10.1038/sj.cdd.4401360] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The activation of caspase-3 represents a critical step in the pathways leading to the biochemical and morphological changes that underlie apoptosis. Upon induction of apoptosis, the large (p17) and small (p12) subunits, comprising active caspase-3, are generated via proteolytic processing of a latent proenzyme dimer. Two copies of each individual subunit are generated to form an active heterotetramer. The tetrameric form of caspase-3 cleaves specific protein substrates within the cell, thereby producing the apoptotic phenotype. In contrast to the proenzyme, once activated in HeLa cells, caspase-3 is difficult to detect due to its rapid degradation. Interestingly, however, enzyme stability and therefore detection of active caspase-3 by immunoblot analysis can be restored by treatment of cells with a peptide-based caspase-3 selective inhibitor, suggesting that the active form can be stabilized through protein-inhibitor interaction. The heteromeric active enzyme complex is necessary for its stabilization by inhibitors, as expression of the large subunit alone is not stabilized by the presence of inhibitors. Our results show for the first time, that synthetic caspase inhibitors not only block caspase activity, but may also increase the stability of otherwise rapidly degraded mature caspase complexes. Consistent with these findings, experiments with a catalytically inactive mutant of caspase-3 show that rapid turnover is dependent on the activity of the mature enzyme. Furthermore, turnover of otherwise stable active site mutants of capase-3 is rescued by the presence of the active enzyme suggesting that turnover can be mediated in trans.
Collapse
Affiliation(s)
- P Tawa
- Department of Biochemistry & Molecular Biology, Merck Frosst Centre for Therapeutic Research, Kirkland, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
196
|
Falk M, Ussat S, Reiling N, Wesch D, Kabelitz D, Adam-Klages S. Caspase Inhibition Blocks Human T Cell Proliferation by Suppressing Appropriate Regulation of IL-2, CD25, and Cell Cycle-Associated Proteins. THE JOURNAL OF IMMUNOLOGY 2004; 173:5077-85. [PMID: 15470051 DOI: 10.4049/jimmunol.173.8.5077] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Caspases have been described as proteases essential for the release of certain cytokines and for initiation as well as execution of apoptosis. Increasing evidence indicates, however, that caspase activity is also required for activation-induced proliferation of mature T lymphocytes. The molecular mechanism, how caspase activity facilitates T cell proliferation, is still controversially discussed. In this study, we show that proliferation of human T cells in response to a specific antigenic stimulus is completely prevented by caspase inhibition. In addition, we demonstrate that this lack of proliferation is due to a failure to initiate cell cycle progression, but not the result of increased T cell death. Our results demonstrate that caspase inhibition leads to strongly reduced IL-2 release, failure to up-regulate CD25, and a lack of proper regulation of cell cycle-associated proteins. Furthermore, T cell proliferation was partially rescued by addition of exogenous IL-2. Using Jurkat cells, we show that in the absence of caspase-8, the mitogen-induced activation of the transcription factor NF-kappaB is moderately diminished, while the activity of the composite element CD28 response element and NF-IL-2B AP-1 sites is strongly reduced. Finally, we provide evidence that caspase inhibition suppresses the activation of purified monocytes by bacterial Ags.
Collapse
Affiliation(s)
- Markus Falk
- Institut für Immunologie, Universitätsklinikum Schleswig-Holstein Campus Kiel, Germany
| | | | | | | | | | | |
Collapse
|
197
|
Abstract
During their development, B-lineage cells are selected to mature, to die, to divide, or to survive and wait, ready to respond to external signals. The homeostatic balance between growth, death, and survival is mediated by signaling pathways through the B-cell antigen receptor (BCR) complex, cytokine and chemokine receptors or cell-cell coreceptor interactions. The BCR complex is a master regulator essential at key checkpoints during development. These checkpoints involve various processes, including negative selection (deletion), anergy, receptor editing, and positive selection. Without BCRs or downstream BCR-signaling components, B-lineage cells arrest during development. Removal of BCRs from mature B cells leads to their death. Here, we discuss signaling pathways in B cells that activate members of the caspase family of cysteine proteases. In some B-cell subsets, BCR signaling activates caspases, which in turn induce a program leading to cell death. However, in other contexts, caspases are involved in the proliferation of B cells. The outcome depends in part on the presence or absence of modifiers that affect signaling thresholds and on which caspases are activated. These mechanisms allow the coordinated regulation of proliferation and apoptosis that is essential for lymphoid homeostasis.
Collapse
Affiliation(s)
- Jonathan D Graves
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | | | | |
Collapse
|
198
|
Kang TB, Ben-Moshe T, Varfolomeev EE, Pewzner-Jung Y, Yogev N, Jurewicz A, Waisman A, Brenner O, Haffner R, Gustafsson E, Ramakrishnan P, Lapidot T, Wallach D. Caspase-8 serves both apoptotic and nonapoptotic roles. THE JOURNAL OF IMMUNOLOGY 2004; 173:2976-84. [PMID: 15322156 DOI: 10.4049/jimmunol.173.5.2976] [Citation(s) in RCA: 295] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Knockout of caspase-8, a cysteine protease that participates in the signaling for cell death by receptors of the TNF/nerve growth factor family, is lethal to mice in utero. To explore tissue-specific roles of this enzyme, we established its conditional knockout using the Cre/loxP recombination system. Consistent with its role in cell death induction, deletion of caspase-8 in hepatocytes protected them from Fas-induced caspase activation and death. However, application of the conditional knockout approach to investigate the cause of death of caspase-8 knockout embryos revealed that this enzyme also serves cellular functions that are nonapoptotic. Its deletion in endothelial cells resulted in degeneration of the yolk sac vasculature and embryonal death due to circulatory failure. Caspase-8 deletion in bone-marrow cells resulted in arrest of hemopoietic progenitor functioning, and in cells of the myelomonocytic lineage, its deletion led to arrest of differentiation into macrophages and to cell death. Thus, besides participating in cell death induction by receptors of the TNF/nerve growth factor family, caspase-8, apparently independently of these receptors, also mediates nonapoptotic and perhaps even antiapoptotic activities.
Collapse
Affiliation(s)
- Tae-Bong Kang
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
199
|
Chaneva S, Schneider G, Siegmund D, Wajant H, Mages J, Häcker G. Enhanced basal AP-1 activity and de-regulation of numerous genes in T cells transgenic for a dominant interfering mutant of FADD/MORT1. Eur J Immunol 2004; 34:3006-15. [PMID: 15376191 DOI: 10.1002/eji.200425381] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The essential role of FADD/MORT1 and caspase-8 in 'death-receptor'-induced apoptosis is well characterized. Surprisingly, these proteins also play a critical role in antigen- or mitogen-induced activation and proliferation of T cells. We report the results of gene expression profiling in T cells from mice defective in FADD-signaling due to transgenic expression of a dominant negative mutant of FADD (FADDdn T cells). Of the tested genes, 159 were differentially expressed in mutant cells prior to or, more often, after mitogenic stimulation for 20 h. No obvious regulator of proliferation was changed in expression. However, a number of T cell effector genes such as IL-2 and IL-9 were up-regulated upon stimulation. Analysis of IL-2 regulation showed enhanced mRNA induction but normal protein production in activated FADDdn T cells. Activation of the nuclear factor of activated T cells was normal upon stimulation but the activity of the activator protein 1 (AP-1) family was strongly increased in resting FADDdn T cells. Expression and transcriptional activity of classical AP-1 family members was unaltered in FADDdn cells, suggesting the involvement of other AP-1 family members. The constitutive activation of AP-1 may thus serve to precondition resting T cells for an enhanced expression of many immunologically relevant genes during activation.
Collapse
Affiliation(s)
- Svetla Chaneva
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, Munich, Germany
| | | | | | | | | | | |
Collapse
|
200
|
Medina-Palazon C, Bernard E, Frost V, Morley S, Sinclair AJ. KIPase activity is a novel caspase-like activity associated with cell proliferation. ACTA ACUST UNITED AC 2004; 271:2716-23. [PMID: 15206936 DOI: 10.1111/j.1432-1033.2004.04200.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A novel caspase-like activity, which is directly regulated with cell proliferation is a candidate to regulate the abundance of the cyclin-dependent kinase inhibitor, p27(KIP1), in human lymphoid cells. This activity, which we term KIPase activity, can also cleave a subset of caspase substrates. Here we demonstrate that KIPase is a novel enzyme distinct from any of the previously characterized human caspases. We show that KIPase is active in a variety of cell lineages, its activity is associated with the proliferation of the human T-cell line, Jurkat, and is not inhibited by the broad spectrum caspase inhibitor z-VAD-fmk. Gel filtration analysis revealed that KIPase has a native molecular mass of approximately 100-200 kDa. Furthermore, the activity of KIPase does not change during apoptosis induced by either ligation of FAS or exposure of cells to etoposide. The uniqueness of KIPase is demonstrated by the fact that none of the human caspases tested (1-10) are able to cleave a specific KIPase substrate (Ac-DPSD-AMC) and that an aldehyde modified derivative of the DPSD tetra peptide is unable to inhibit caspases, but is a good inhibitor of KIPase activity. This supports a hypothesis whereby KIPase is a currently unidentified caspase-like enzyme which regulates the abundance of p27(KIP1) in a proliferation-dependent manner.
Collapse
Affiliation(s)
- Cahora Medina-Palazon
- Biochemistry Department, School of Life Sciences, University of Sussex, Brighton, UK
| | | | | | | | | |
Collapse
|