Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters








Publication year range
1.
Oncogene ; 26(33): 4842-9, 2007 Jul 19.
Article in English | MEDLINE | ID: mdl-17297443

ABSTRACT

We have isolated a novel interferon (IFN)-retinoid regulated cell death regulatory protein genes associated with retinoid-IFN-induced mortality (GRIM)-19 earlier. To understand its mechanism of action, we have employed a yeast-two-hybrid screen and identified serine protease HtrA2 as its binding partner. GRIM-19 physically interacts with HtrA2 and augments cell death in an IFN/all-trans retinoic acid (RA)-dependent manner. In the presence of GRIM-19, the HtrA2-driven destruction of the antiapoptotic protein X-linked inhibitor of apoptosis (XIAP) is augmented. These interactions were disrupted by an human herpes virus-8 (HHV-8)-coded oncoprotein, vIRF1, and conferred resistance to IFN/RA-induced cell death. These data show a critical role of HtrA2 in a cytokine-induced cell death response for the first time and its inhibition by a viral protein.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Mitochondrial Proteins/metabolism , NADH, NADPH Oxidoreductases/metabolism , Serine Endopeptidases/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/physiology , Drug Resistance , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , High-Temperature Requirement A Serine Peptidase 2 , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interferon-beta/pharmacology , Mitochondrial Proteins/genetics , NADH, NADPH Oxidoreductases/genetics , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine Endopeptidases/genetics , Transfection , Tretinoin/pharmacology , Two-Hybrid System Techniques , Viral Proteins/genetics , Viral Proteins/metabolism , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism
2.
Cell Death Differ ; 11(2): 143-53, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14576767

ABSTRACT

Mitochondria are known to combine life-supporting functions with participation in apoptosis by controlling caspase activity. Here, we report that in human blood neutrophils the mitochondria are different, because they preserve mainly death-mediating abilities. Neutrophil mitochondria hardly participate in ATP synthesis, and have a very low activity of the tested marker enzymes. The presence of mitochondria in neutrophils was confirmed by quantification of mitochondrial DNA copy number, by detection of mitochondrial porin, and by JC-1 measurement of Deltapsi(m). During neutrophilic differentiation, HL-60 cells demonstrated a profound cytochrome c depletion and mitochondrial shape change reminiscent of neutrophils. However, blood neutrophils containing extremely low amounts of cytochrome c displayed strong caspase-9 activation during apoptosis, which was also observed in apoptotic neutrophil-derived cytoplasts lacking any detectable cytochrome c. We suggest that other proapoptotic factors such as Smac/DIABLO and HtrA2/Omi, which are massively released from the mitochondria, have an important role in neutrophil apoptosis.


Subject(s)
Apoptosis , Mitochondria/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Proto-Oncogene Proteins c-bcl-2 , Adenosine Triphosphate/metabolism , Caspase 9 , Caspases/metabolism , Cell Differentiation , Cell Lineage , Cytochromes c/metabolism , Cytosol/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Fumarate Hydratase/metabolism , Glutamate Dehydrogenase/metabolism , HL-60 Cells , Humans , L-Lactate Dehydrogenase/metabolism , Membrane Potentials , Mitochondria/genetics , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins/metabolism , bcl-2-Associated X Protein
3.
Cell Death Differ ; 10(8): 905-13, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12867998

ABSTRACT

SCL/Tal-1 is a helix-loop-helix (HLH) transcription factor required for blood cell development, whose abnormal expression is responsible for induction of T-cell acute lymphoblastic leukemia. We show here that SCL/Tal-1 is a key target of caspases in developing erythroblasts. SCL/Tal-1 degradation occurred rapidly after caspase activation and preceded the cleavage of the major erythroid transcription factor GATA-1. Expression of a caspase-resistant SCL/Tal-1 in erythroid progenitors was able to prevent amplification of caspase activation, GATA-1 degradation and impaired erythropoiesis induced by growth factor deprivation or death receptor triggering. The potent proerythropoietic activity of uncleavable SCL/Tal-1 was clearly evident in the absence of erythropoietin, a condition that did not allow survival of normal erythroid cells or expansion of erythroblasts expressing caspase-resistant GATA-1. In the absence of erythropoietin, cells expressing caspase-resistant SCL/Tal-1 maintain high levels of Bcl-X(L), which inhibits amplification of the caspase cascade and mediates protection from apoptosis. Thus, SCL/TAL-1 is a survival factor for erythroid cells, whereas caspase-mediated cleavage of SCL/Tal-1 results in amplification of caspase activation, GATA-1 degradation and impaired erythropoiesis.


Subject(s)
Caspases/metabolism , DNA-Binding Proteins/metabolism , Erythropoiesis/physiology , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Antibodies/pharmacology , Apoptosis/drug effects , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors , Blotting, Western , Caspase 3 , Caspase 7 , Caspase 8 , Cell Division/drug effects , Cloning, Molecular , DNA-Binding Proteins/genetics , Down-Regulation , Enzyme Precursors/metabolism , Erythroblasts/cytology , Erythroblasts/drug effects , Erythroblasts/metabolism , Erythroid-Specific DNA-Binding Factors , Erythropoietin/deficiency , Erythropoietin/pharmacology , GATA1 Transcription Factor , GATA2 Transcription Factor , Gene Expression Regulation , Green Fluorescent Proteins , Helix-Loop-Helix Motifs/genetics , Helix-Loop-Helix Motifs/physiology , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription Factors/genetics , bcl-X Protein , fas Receptor/immunology , fas Receptor/physiology
4.
Cell Death Differ ; 10(2): 175-84, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12700645

ABSTRACT

Apoptosis control in adult testis is crucial to achieve normal spermatogenesis. In this study c-FLIP, an apoptosis-modulating protein, was investigated. In Western blot and immunohistochemical analyses, the 55 KDa c-FLIP long isoform (c-FLIP(L)) was found to be expressed strongly in spermatocytes and spermatids, at low levels in spermatogonia and at almost undetectable levels in Sertoli cells. This expression pattern was confirmed by Northern blot analyses. Further experiments carried out on GC-1spg germ cell line revealed that reducing c-FLIP(L) expression increases Fas-dependent apoptosis. Conversely, restoring c-FLIP(L) expression reduces this response to control levels. Caspase-10 expression was found to match c-FLIP(L) expression pattern; further, caspase-10 activation upon anti-Fas treatment inversely correlated with c-FLIP(L) expression. Finally, TUNEL staining of seminiferous tubules incubated with anti-Fas antibody showed that apoptosis occurs mostly in basally located germ cells, indicating that such cells, expressing low levels of c-FLIP(L), are sensitive to Fas-mediated apoptosis. These data indicate for the first time that c-FLIP(L) might control germ cell apoptosis and caspase activity in the adult testis.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Testis/metabolism , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspases/metabolism , Cell Line/drug effects , Cells, Cultured , Enzyme Activation , Germ Cells/cytology , Male , Mice , Mice, Inbred Strains , Oligonucleotides, Antisense/pharmacology , Protein Isoforms/metabolism , Seminiferous Tubules , Sertoli Cells/cytology , Sertoli Cells/metabolism , Spermatids/cytology , Spermatids/metabolism , Spermatocytes/cytology , Spermatocytes/metabolism , Spermatogonia/cytology , Spermatogonia/metabolism , Testis/chemistry , Testis/cytology , fas Receptor/metabolism
5.
Cell Death Differ ; 9(9): 995-1006, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12181750

ABSTRACT

Caspase-14 is a recent addition to the caspase family of aspartate proteases involved in apoptotic processes. Human caspase-14 appears to be only weakly processed during apoptosis, and it does not cleave classical caspase substrates. Post partum, caspase-14 is prominently expressed by human keratinocytes and reportedly participates in terminal differentiation of complex epithelia. Here we provide evidence challenging the view that caspase-14 expression or processing is linked exclusively to terminal keratinocyte differentiation. We demonstrate that caspase-14 expression extended to multiple cell lines derived from simple epithelia of the breast, prostate, and stomach. In keratinocytes and breast epithelial cells, caspase-14 expression was upregulated in high-density cultures and during forced suspension culture. These effects were primarily due to transcriptional activation as indicated by reporter gene assays using a 2 kb caspase-14 promoter fragment. Importantly, caspase-14 was not cleaved during forced suspension culture of either cell type although this treatment induced caspase-dependent apoptosis (anoikis). Forced expression of caspase-14 in immortalized human keratinocytes had no effect on cell death in forced suspension nor was the transfected caspase-14 processed in this setting. In contrast to postconfluent and forced suspension culture, terminal differentiation of keratinocytes induced in vitro by Ca2+ treatment was not associated with increased caspase-14 expression or promoter activity. Our results indicate that (1) caspase-14 is expressed not only in complex but also simple epithelia; (2) cells derived from complex and simple epithelia upregulate caspase-14 expression in conditions of high cell density or lack of matrix interaction and; (3) in both cell types this phenomenon is due to transcriptional regulation.


Subject(s)
Caspases/genetics , Cell Differentiation/genetics , Epithelial Cells/enzymology , Epithelium/enzymology , Gene Expression Regulation, Enzymologic/genetics , Genes, Regulator/genetics , Antibody Specificity/immunology , Breast/cytology , Breast/enzymology , Breast/growth & development , Caspase 14 , Cell Adhesion/physiology , Cell Compartmentation/physiology , Cell Cycle/physiology , Cells, Cultured , Epidermal Cells , Epidermis/enzymology , Epidermis/growth & development , Epithelial Cells/cytology , Epithelium/growth & development , Extracellular Matrix/enzymology , Humans , Infant, Newborn , Keratinocytes/cytology , Keratinocytes/enzymology , Male , Promoter Regions, Genetic/genetics , Prostate/cytology , Prostate/enzymology , Prostate/growth & development
6.
Cell Death Differ ; 9(4): 439-47, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11965497

ABSTRACT

Death effector domain-containing proteins are involved in important cellular processes such as death-receptor induced apoptosis, NF-kappaB activation and ERK activation. Here we report the identification of a novel nuclear DED-containing protein, FLAME-3. FLAME-3 shares significant sequence (46.6% identical) and structural homology to another DED-containing protein, DEDD. FLAME-3 interacts with DEDD and c-FLIP (FLAME-1) but not with the other DED-containing proteins FADD, caspase-8 or caspase-10. FLAME-3 translocates to, and sequesters c-FLIP in the nucleus upon overexpression in human cell lines. Using the yeast two-hybrid system to identify DEDD-interacting proteins, the TFIIIC102 subunit of human transcription factor TFIIIC was identified as a DEDD- and FLAME-3-specific interacting protein. Co-expression of either DEDD or FLAME-3 with hTFIIIC102 in MCF-7 cells induces the translocation from the cytoplasm and sequestration of hTFIIIC102 in the nucleus, indicating that DEDD and FLAME-3 form strong heterocomplexes with hTFIIIC102 and might be important regulators of the activity of the hTFIIIC transcriptional complex. Consistent with this, overexpression of DEDD or FLAME-3 in 293 cells inhibited the expression of a luciferase-reporter gene under the control of the NF-kappaB promoter. Our data provide the first direct evidence for the involvement of DED-containing proteins in the regulation of components of the general transcription machinery in the nucleus.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Nuclear Proteins/metabolism , Transcription Factors, TFIII/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Cells, Cultured , Cloning, Molecular , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins , Humans , Macromolecular Substances , Mice , Molecular Sequence Data , NF-kappa B/antagonists & inhibitors , Nuclear Proteins/genetics , Protein Structure, Tertiary , Sequence Alignment , Sequence Analysis , Sequence Analysis, Protein , Transcription Factors, TFIII/genetics , Transfection
7.
Cell Death Differ ; 9(1): 20-6, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11803371

ABSTRACT

Proteome analysis of supernatant of isolated mitochondria exposed to recombinant tBid, a proapoptotic Bcl-2 member, revealed the presence of the serine protease Omi, also called HtrA2. This release was prevented in mitochondria derived from Bcl-2-transgenic mice. Release of Omi under apoptotic conditions was confirmed in vivo in livers from mice injected with agonistic anti-Fas antibodies and was prevented in livers from Bcl-2 transgenic mice. Omi release also occurs in apoptotic dying but not in necrotic dying fibrosarcoma L929 cells, treated with anti-Fas antibodies and TNF, respectively. The amino acid sequence reveals the presence of an XIAP interaction motif at the N-terminus of mature Omi. We demonstrate an interaction between endogeneous Omi and recombinant XIAP. Furthermore we show that endogenous Omi is involved in enhanced activation of caspases in cytosolic extracts.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Mitochondria/metabolism , Proteins/metabolism , Serine Endopeptidases/metabolism , Amino Acid Sequence , Animals , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/pharmacology , Cells, Cultured , Cytosol/metabolism , Enzyme Activation , High-Temperature Requirement A Serine Peptidase 2 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondrial Proteins , Molecular Sequence Data , Translocation, Genetic/drug effects , X-Linked Inhibitor of Apoptosis Protein
8.
Cell ; 107(3): 399-407, 2001 Nov 02.
Article in English | MEDLINE | ID: mdl-11701129

ABSTRACT

Apoptosis is primarily executed by active caspases, which are derived from the inactive procaspase zymogens through proteolytic cleavage. Here we report the crystal structures of a caspase zymogen, procaspase-7, and an active caspase-7 without any bound inhibitors. Compared to the inhibitor-bound caspase-7, procaspase-7 zymogen exhibits significant structural differences surrounding the catalytic cleft, which precludes the formation of a productive conformation. Proteolytic cleavage between the large and small subunits allows rearrangement of essential loops in the active site, priming active caspase-7 for inhibitor/substrate binding. Strikingly, binding by inhibitors causes a 180 degrees flipping of the N terminus in the small subunit, which interacts with and stabilizes the catalytic cleft. These analyses reveal the structural mechanisms of caspase activation and demonstrate that the inhibitor/substrate binding is a process of induced fit.


Subject(s)
Caspases/chemistry , Enzyme Precursors/chemistry , Amino Acid Sequence , Caspase 7 , Caspases/genetics , Caspases/metabolism , Crystallography, X-Ray , Enzyme Activation , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , Models, Molecular , Molecular Sequence Data , Protein Processing, Post-Translational , Protein Structure, Tertiary , Substrate Specificity
9.
Biochem Biophys Res Commun ; 287(1): 181-9, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11549272

ABSTRACT

Tumor necrosis factor-alpha (TNFalpha) mediates cytochrome c release from mitochondria, loss of mitochondrial membrane potential (DeltaPsim) and apoptosis in sensitive leukemic cells. In the present study, by using the human leukemic U937 cell line, we demonstrate that the cytochrome c release is caspase-8-dependent and can be blocked by an inhibitor of caspase-8, Z-Ile-Glu (OMe)-Thr-Asp(OMe)-fluoromethyl ketone (Z-IETD.fmk), or a pan caspase inhibitor, benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone (Z-VAD.fmk). However, TNFalpha-mediated loss of DeltaPsim was not inhibited by caspase inhibitors. The apoptotic process was blocked by either Z-IETD.fmk or Z-VAD.fmk in cells with lower DeltaPsim. U937 cells with stable transfection of the cellular inhibitor of apoptosis protein 1 (c-IAP1) are resistant to TNFalpha-induced activation of caspases, Bid cleavage, cytochrome c release and DeltaPsim collapse. In addition, both c-IAP1 and XIAP were not up-regulated upon prolonged exposure to TNFalpha. In contrast, there was a caspase-dependent cleavage of XIAP, but not c-IAP1, during treatment with TNFalpha for 7 days. These results demonstrate that c-IAP1 blocks TNFalpha signaling at a level controlling both activation of caspase-8 and a signal to cause loss of DeltaPsim. The sensitive U937 cell line failed to acquire resistance and gain a self-protecting advantage against apoptosis, upon induction of c-IAP1 expression.


Subject(s)
Apoptosis , Caspases/metabolism , Mitochondria/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Viral Proteins/pharmacology , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/metabolism , Caspase 3 , Caspase 8 , Caspase 9 , Caspase Inhibitors , Cytochrome c Group/metabolism , Drug Interactions , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Inhibitor of Apoptosis Proteins , Leukemia/pathology , Membrane Potentials/drug effects , Mitochondria/enzymology , Mitochondria/physiology , Proteins/metabolism , Signal Transduction/drug effects , Transfection , Tumor Cells, Cultured , U937 Cells , Ultraviolet Rays , Viral Proteins/genetics , X-Linked Inhibitor of Apoptosis Protein
10.
Cell Death Differ ; 8(6): 649-57, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11536016

ABSTRACT

We report here the identification and functional characterization of two new human caspase recruitment domain (CARD) molecules, termed Pseudo-interleukin-1beta converting enzyme (ICE) and ICEBERG. Both proteins share a high degree of homology, reaching 92% and 53% identity, respectively, to the prodomain of caspase-1/ICE. Interestingly, both Pseudo-ICE and ICEBERG are mapped to chromosome 11q22 that bears caspases-1, -4- and -5 genes, all involved in cytokine production rather than in apoptosis. We demonstrate that Pseudo-ICE and ICEBERG interact physically with caspase-1 and block, in a monocytic cell line, the interferon-gamma and lipopolysaccharide-induced secretion of interleukin-1beta which is a well-known consequence of caspase-1 activation. Moreover, Pseudo-ICE, but not ICEBERG, interacts with the CARD-containing kinase RICK/RIP2/CARDIAK and activates NF-kappaB. Our data suggest that Pseudo-ICE and ICEBERG are intracellular regulators of caspase-1 activation and could play a role in the regulation of IL-1beta secretion and NF-kappaB activation during the pro-inflammatory cytokine response.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Caspases/chemistry , Caspases/metabolism , Interleukin-1/biosynthesis , Intracellular Signaling Peptides and Proteins , Amino Acid Sequence , Apoptosis/drug effects , Base Sequence , Carrier Proteins/genetics , Caspase 1/chemistry , Caspase 1/metabolism , Caspase Inhibitors , Caspases/genetics , Cell Line , Cloning, Molecular , Enzyme Activation , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/pharmacology , Interleukin-1/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Molecular Sequence Data , NF-kappa B/metabolism , Protein Binding , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptors, Interleukin-1/antagonists & inhibitors , Sequence Alignment , Tumor Necrosis Factor-alpha/pharmacology
11.
Oncogene ; 20(35): 4817-26, 2001 Aug 09.
Article in English | MEDLINE | ID: mdl-11521193

ABSTRACT

Bax translocation from cytosol to mitochondria is believed to be a crucial step for triggering cytochrome c release from mitochondria. However, it is unclear whether Bax translocation is associated with Bax induction by DNA damaging agents. The induction of Bax in response to DNA damaging agents has been considered to be linked with p53. In this study, we used the p53 negative human chronic myeloid leukaemia K562 cell line. Bax up-regulation occurred at the whole cell level after DNA damage induced by etoposide. However, after incubation with etoposide, Bax failed to translocate to mitochondria and as a result, the apoptotic process was blocked. A Bax stable transfectant, the K/Bax cell line, expressed more Bax protein in the cytosol, mitochondria and nuclei. This Bax overexpression induced cytochrome c release, a reduction of cytochrome c oxidase activity and mitochondrial membrane potential (Delta(Psi)m). However, Bax-induced apoptosis was blocked downstream of mitochondria in K562 cells. The increased levels of mitochondrial Bax sensitized cells to etoposide-induced activation of caspases-2, -3 and -9 and apoptosis. However, after transient transfection with the Apaf-1 gene, K/Bax cells were sensitized to etoposide-induced caspase activation and apoptosis to a larger extent compared with Bax or Apaf-1 transfection alone. We therefore conclude that two mechanisms contribute to the resistance of K562 cells to etoposide-induced apoptosis; firstly failure of Bax targeting to mitochondria and, secondly, deficiency of Apaf-1. Uncoupling of Bax translocation from Bax induction can occur in response to etoposide-induced DNA damage.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Etoposide/pharmacology , Leukemia/drug therapy , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Apoptotic Protease-Activating Factor 1 , Biological Transport , Caspases/physiology , DNA Damage , Humans , K562 Cells , Leukemia/pathology , Proteins/physiology , bcl-2-Associated X Protein
12.
Cancer Res ; 61(15): 5796-802, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11479218

ABSTRACT

Because the apoptotic pathway is often disrupted in tumor cells, its genetic restoration is a very attractive approach for the treatment of tumors. To treat malignant gliomas with this approach, it would be preferred to restrict induction of apoptosis to tumor cells by establishing a tumor-specific expression system. Telomerase is an attractive target because the vast majority of malignant gliomas have telomerase activity whereas normal brain cells do not. Activation of telomerase is tightly regulated at the transcriptional level of the telomerase catalytic subunit [human telomerase reverse transcriptase, (hTERT)]. Therefore, we hypothesized that using a hTERT promoter-driven vector system, an apoptosis-inducible gene may be preferentially restricted to telomerase- or hTERT-positive tumor cells. In this study, we constructed an expression vector consisting of the constitutively active caspase-6 (rev-caspase-6) under the hTERT promoter (hTERT/rev-caspase-6) and then investigated its antitumor effect on malignant glioma cells. The rationale for using the rev-caspase-6 gene is because it induces apoptosis independent of the initiator caspases. We demonstrated that the hTERT/rev-caspase-6 construct induced apoptosis in hTERT-positive malignant glioma cells, but not in hTERT-negative astrocytes, fibroblasts, and alternative lengthening of telomeres cells. In addition, the growth of s.c. tumors in nude mice was significantly suppressed by the treatment with hTERT/rev-caspase-6 construct. The present results strongly suggest that the telomerase-specific transfer of the rev-caspase-6 gene under the hTERT promoter is a novel targeting approach for the treatment of malignant gliomas.


Subject(s)
Caspases/genetics , Genetic Therapy/methods , Glioma/therapy , Promoter Regions, Genetic/genetics , RNA , Telomerase/genetics , Animals , Apoptosis/genetics , Caspase 6 , Caspases/biosynthesis , Caspases/metabolism , DNA-Binding Proteins , Gene Transfer Techniques , Genetic Vectors/genetics , Glioma/enzymology , Glioma/genetics , Glioma/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Telomerase/biosynthesis , Transcriptional Activation , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Blood ; 98(2): 414-21, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11435311

ABSTRACT

The human leukemia cell lines K562, CEM, CEM/VLB(100), human leukemic blasts, and the bladder cancer J82 cell line have different sensitivities to UV light-induced apoptosis. It is reported that resistance to UV light-induced apoptosis occurs at a point in the apoptotic pathway upstream of caspase-3 but downstream of mitochondrial cytochrome c release. It is demonstrated that the block is due to deficiency of Apaf-1, a critical member of the apoptosome. Sensitivity to apoptosis was independent of caspase-9b or XIAP (inhibitors of apoptosis proteins) expression or levels of procaspase-9. Transfection of Apaf-1 conferred sensitivity to apoptosis in resistant cells. Apaf-1 deficiency may constitute a significant mode of resistance to apoptosis in human leukemia.


Subject(s)
Apoptosis , Cytochrome c Group/metabolism , Proteins/analysis , Antibodies/pharmacology , Apoptosis/drug effects , Apoptotic Protease-Activating Factor 1 , Caspase 3 , Caspase 9 , Caspases/analysis , Caspases/metabolism , Cytochrome c Group/pharmacology , Enzyme Activation/drug effects , Granzymes , Humans , Leukemia, Myeloid, Acute , Leukemia-Lymphoma, Adult T-Cell , Protein Precursors/metabolism , Proteins/genetics , Proteins/immunology , Proteins/metabolism , Serine Endopeptidases/pharmacology , Transfection , Tumor Cells, Cultured , Ultraviolet Rays , Urinary Bladder Neoplasms , X-Linked Inhibitor of Apoptosis Protein
15.
J Biol Chem ; 276(30): 28309-13, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11390368

ABSTRACT

Procaspase-9 contains an NH2-terminal caspase-associated recruitment domain (CARD), which is essential for direct association with Apaf-1 and activation. Procaspase-1 also contains an NH2-terminal CARD domain, suggesting that its mechanism of activation, like that of procaspase-9, involves association with an Apaf-1-related molecule. Here we describe the identification of a human Apaf-1-related protein, named Ipaf that contains an NH2-terminal CARD domain, a central nucleotide-binding domain, and a COOH-terminal regulatory leucine-rich repeat domain (LRR). Ipaf associates directly and specifically with the CARD domain of procaspase-1 through CARD-CARD interaction. A constitutively active Ipaf lacking its COOH-terminal LRR domain can induce autocatalytic processing and activation of procaspase-1 and caspase-1-dependent apoptosis in transfected cells. Our results suggest that Ipaf is a specific and direct activator of procaspase-1 and could be involved in activation of caspase-1 in response to pro-inflammatory and apoptotic stimuli.


Subject(s)
Caspase 1/metabolism , Proteins/chemistry , Proteins/physiology , Amino Acid Sequence , Apoptosis , Apoptotic Protease-Activating Factor 1 , Caspase 9 , Caspases/metabolism , Cell Line , Cells, Cultured , DNA, Complementary/metabolism , Enzyme Activation , Enzyme Precursors/metabolism , Glutathione Transferase/metabolism , Humans , Immunoblotting , Microscopy, Confocal , Molecular Sequence Data , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Signal Transduction , Tissue Distribution , Transfection , Tumor Cells, Cultured
16.
Mol Cell Biol ; 21(9): 3025-36, 2001 May.
Article in English | MEDLINE | ID: mdl-11287608

ABSTRACT

The function of BAD, a proapoptotic member of the Bcl-2 family, is regulated primarily by rapid changes in phosphorylation that modulate its protein-protein interactions and subcellular localization. We show here that, during interleukin-3 (IL-3) deprivation-induced apoptosis of 32Dcl3 murine myeloid precursor cells, BAD is cleaved by a caspase(s) at its N terminus to generate a 15-kDa truncated protein. The 15-kDa truncated BAD is a more potent inducer of apoptosis than the wild-type protein, whereas a mutant BAD resistant to caspase 3 cleavage is a weak apoptosis inducer. Truncated BAD is detectable only in the mitochondrial fraction, interacts with BCL-X(L) at least as effectively as the wild-type protein, and is more potent than wild-type BAD in inducing cytochrome c release. Human BAD, which is 43 amino acids shorter than its mouse counterpart, is also cleaved by a caspase(s) upon exposure of Jurkat T cells to anti-FAS antibody, tumor necrosis factor alpha (TNF-alpha), or TRAIL. Moreover, a truncated form of human BAD lacking the N-terminal 28 amino acids is more potent than wild-type BAD in inducing apoptosis. The generation of truncated BAD was blocked by Bcl-2 in IL-3-deprived 32Dcl3 cells but not in Jurkat T cells exposed to anti-FAS antibody, TNF-alpha, or TRAIL. Together, these findings point to a novel and important role for BAD in maintaining the apoptotic phenotype in response to various apoptosis inducers.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Caspases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Binding Sites , Carrier Proteins/genetics , Caspase 3 , Caspases/physiology , Cell Survival , Culture Media , Cytochrome c Group/metabolism , Hematopoietic Stem Cells/cytology , Humans , Interleukin-3/metabolism , Jurkat Cells , Mice , Mitochondria/metabolism , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , Subcellular Fractions , bcl-Associated Death Protein , bcl-X Protein
17.
Nature ; 410(6824): 112-6, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11242052

ABSTRACT

X-linked inhibitor-of-apoptosis protein (XIAP) interacts with caspase-9 and inhibits its activity, whereas Smac (also known as DIABLO) relieves this inhibition through interaction with XIAP. Here we show that XIAP associates with the active caspase-9-Apaf-1 holoenzyme complex through binding to the amino terminus of the linker peptide on the small subunit of caspase-9, which becomes exposed after proteolytic processing of procaspase-9 at Asp315. Supporting this observation, point mutations that abrogate the proteolytic processing but not the catalytic activity of caspase-9, or deletion of the linker peptide, prevented caspase-9 association with XIAP and its concomitant inhibition. We note that the N-terminal four residues of caspase-9 linker peptide share significant homology with the N-terminal tetra-peptide in mature Smac and in the Drosophila proteins Hid/Grim/Reaper, defining a conserved class of IAP-binding motifs. Consistent with this finding, binding of the caspase-9 linker peptide and Smac to the BIR3 domain of XIAP is mutually exclusive, suggesting that Smac potentiates caspase-9 activity by disrupting the interaction of the linker peptide of caspase-9 with BIR3. Our studies reveal a mechanism in which binding to the BIR3 domain by two conserved peptides, one from Smac and the other one from caspase-9, has opposing effects on caspase activity and apoptosis.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Caspases/metabolism , Mitochondrial Proteins , Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Apoptotic Protease-Activating Factor 1 , Caspase 9 , Caspase Inhibitors , Catalysis , Cloning, Molecular , Crystallography, X-Ray , Drosophila , Enzyme Activation , Enzyme Inhibitors/metabolism , Escherichia coli , Humans , Intracellular Signaling Peptides and Proteins , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Processing, Post-Translational , Proteins/chemistry , Recombinant Proteins/metabolism , X-Linked Inhibitor of Apoptosis Protein
18.
Cell ; 104(5): 769-80, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11257230

ABSTRACT

The inhibitor of apoptosis (IAP) proteins suppress cell death by inhibiting the catalytic activity of caspases. Here we present the crystal structure of caspase-7 in complex with a potent inhibitory fragment from XIAP at 2.45 A resolution. An 18-residue XIAP peptide binds the catalytic groove of caspase-7, making extensive contacts to the residues that are essential for its catalytic activity. Strikingly, despite a reversal of relative orientation, a subset of interactions between caspase-7 and XIAP closely resemble those between caspase-7 and its tetrapeptide inhibitor DEVD-CHO. Our biochemical and structural analyses reveal that the BIR domains are dispensable for the inhibition of caspase-3 and -7. This study provides a structural basis for the design of the next-generation caspase inhibitors.


Subject(s)
Carrier Proteins , Caspases/chemistry , Caspases/metabolism , Mitochondrial Proteins , Proteins/chemistry , Proteins/metabolism , Apoptosis/physiology , Caspase 7 , Caspases/genetics , Catalytic Domain , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Proteins/genetics , Sequence Homology, Amino Acid , Structure-Activity Relationship , X-Linked Inhibitor of Apoptosis Protein
19.
J Cell Physiol ; 187(2): 176-87, 2001 May.
Article in English | MEDLINE | ID: mdl-11267997

ABSTRACT

The Tpl-2 proto-oncoprotein promotes cellular proliferation when overexpressed in a variety of tumor cell lines. Here, we present evidence that when overexpressed in immortalized non-transformed cells, Tpl-2 induces apoptosis by promoting the activation of caspase-3 via a caspase-9-dependent mechanism, and that apoptosis is enhanced when Tpl-2 is co-expressed with the newly identified ankyrin repeat protein Tvl-1. The activation of caspase-3 by caspase-9 is known to depend on the assembly of a multimolecular complex that includes Apaf-1 and caspase-9. Data presented here show that co-expression of Tpl-2 with Tvl-1 promotes the assembly of a complex that involves several proteins that bind Apaf-1 including Tvl-1, itself, Tpl-2 and phosphorylated procaspase-9. More important, procaspase-3, which under normal growth conditions is not associated with the complex, binds Tvl-1 conditionally in response to Tpl-2-generated apoptotic signals. The conditional association of procaspase-3 with Tvl-1 promotes the in vivo proteolytic maturation of procaspase-3 by caspase-9, a process casually linked to apoptosis.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/physiology , Carrier Proteins/metabolism , Caspases/metabolism , Enzyme Precursors/metabolism , MAP Kinase Kinase Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Amino Acid Sequence , Animals , Apoptotic Protease-Activating Factor 1 , Carrier Proteins/genetics , Caspase 3 , Caspase 9 , Cell Line , DNA-Binding Proteins , Enzyme Activation/physiology , Fibroblasts/cytology , Fibroblasts/enzymology , Gene Expression/physiology , Humans , Kidney/cytology , MAP Kinase Kinase Kinases/genetics , Molecular Sequence Data , Phosphorylation , Protein Binding/physiology , Proteins/metabolism , Proto-Oncogene Proteins/genetics , Rats , Transcription Factors
20.
EMBO J ; 20(5): 998-1009, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11230124

ABSTRACT

During apoptosis, release of cytochrome c initiates dATP-dependent oligomerization of Apaf-1 and formation of the apoptosome. In a cell-free system, we have addressed the order in which apical and effector caspases, caspases-9 and -3, respectively, are recruited to, activated and retained within the apoptosome. We propose a multi-step process, whereby catalytically active processed or unprocessed caspase-9 initially binds the Apaf-1 apoptosome in cytochrome c/dATP-activated lysates and consequently recruits caspase-3 via an interaction between the active site cysteine (C287) in caspase-9 and a critical aspartate (D175) in caspase-3. We demonstrate that XIAP, an inhibitor-of-apoptosis protein, is normally present in high molecular weight complexes in unactivated cell lysates, but directly interacts with the apoptosome in cytochrome c/dATP-activated lysates. XIAP associates with oligomerized Apaf-1 and/or processed caspase-9 and influences the activation of caspase-3, but also binds activated caspase-3 produced within the apoptosome and sequesters it within the complex. Thus, XIAP may regulate cell death by inhibiting the activation of caspase-3 within the apoptosome and by preventing release of active caspase-3 from the complex.


Subject(s)
Apoptosis , Caspases/metabolism , Proteins/metabolism , Apoptotic Protease-Activating Factor 1 , Aspartic Acid/metabolism , Caspase 3 , Caspase 9 , Cell Line , Deoxyadenine Nucleotides/pharmacology , Enzyme Activation , Humans , Macromolecular Substances , Models, Biological , Precipitin Tests , Protein Binding , Protein Processing, Post-Translational , Recombinant Proteins , X-Linked Inhibitor of Apoptosis Protein
SELECTION OF CITATIONS
SEARCH DETAIL