Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta ; 1499(3): 191-8, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11341966

ABSTRACT

We cloned a novel inhibitor of apoptosis protein (IAP) family member, BmIAP, from Bombyx mori BmN cells. BmIAP contains two baculoviral IAP repeat (BIR) domains followed by a RING domain. BmIAP shares striking amino acid sequence similarity with lepidopteran IAPs, SfIAP and TnIAP, and with two baculoviral IAPs, CpIAP and OpIAP, suggesting evolutionary conservation. BmIAP blocks programmed cell death (apoptosis) in Spodoptera frugiperda Sf-21 cells induced by p35 deficient Autographa californica nucleopolyhedrovirus (AcMNPV). This anti-apoptotic function requires both the BIR domains and RING domain of BmIAP. In mammalian cells, BmIAP inhibits Bax induced but not Fas induced apoptosis. Further biochemical data suggest that BmIAP is a specific inhibitor of mammalian caspase-9, an initiator caspase in the mitochondria/cytochrome-c pathway, but not the downstream effector proteases, caspase-3 and caspase-7. These results suggest that suppression of apoptosis by lepidopteran IAPs in insect cells may involve inhibition of an upstream initiator caspase in the conserved mitochondria/cytochrome-c pathway for apoptosis.


Subject(s)
Bombyx/genetics , Drosophila Proteins , Insect Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , Amino Acid Sequence , Animals , Apoptosis , Baculoviridae/genetics , Base Sequence , Caspase 9 , Caspase Inhibitors , Caspases/metabolism , Cell Line , Cloning, Molecular , Cysteine Proteinase Inhibitors/metabolism , DNA Primers/genetics , Inhibitor of Apoptosis Proteins , Insect Proteins/chemistry , Molecular Sequence Data , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Spodoptera , bcl-2-Associated X Protein
2.
J Biol Chem ; 276(11): 8087-93, 2001 Mar 16.
Article in English | MEDLINE | ID: mdl-11098060

ABSTRACT

TRAF family proteins are signal-transducing adapter proteins that interact with the cytosolic domains of tumor necrosis factor (TNF) family receptors. Here we show that TRAF1 (but not TRAF2-6) is cleaved by certain caspases in vitro and during TNF-alpha- and Fas-induced apoptosis in vivo. (160)LEVD(163) was identified as the caspase cleavage site within TRAF1, generating two distinct fragments. Significant enhancement of TNF receptor-1 (CD120a)- and, to a lesser extent, Fas (CD95)-mediated apoptosis was observed when overexpressing the C-terminal TRAF1 fragment in HEK293T and HT1080 cells. The same fragment was capable of potently suppressing TNF receptor-1- and TRAF2-mediated nuclear factor-kappaB activation in reporter gene assays, providing a potential mechanism for the enhancement of TNF-mediated apoptosis. Cell death induced by other death receptor-independent stimuli such as cisplatin, staurosporine, and UV irradiation did not result in cleavage of TRAF1, and overexpression of the C-terminal TRAF1 fragment did not enhance cell death in these cases. TRAF1 cleavage was markedly reduced in cells that contain little procaspase-8 protein, suggesting that this apical protease in the TNF/Fas death receptor pathway is largely responsible. These data identify TRAF1 as a specific target of caspases activated during TNF- and Fas-induced apoptosis and illustrate differences in the repertoire of protease substrates cleaved during activation of different apoptotic pathways.


Subject(s)
Apoptosis , Caspases/metabolism , Proteins/metabolism , Tumor Necrosis Factor-alpha/physiology , Female , Humans , Lymphocyte Activation , Lymphocytes/metabolism , NF-kappa B/metabolism , TNF Receptor-Associated Factor 1 , Tumor Cells, Cultured , fas Receptor/physiology
3.
Cardiol Clin ; 19(1): 57-74, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11787814

ABSTRACT

The balance between pro- and antiapoptotic proteins can determine cellular fate. In this regard, the Bcl-2 and IAP protein families have evolved as highly conserved regulators of cell death. A further testament to their critical roles in maintaining balance between cell life and death may be the increasing implication of Bcl-2 and TAP proteins in the pathologies of human diseases. Although much has been learned about these families of proteins, future studies of the Bcl-2 and IAP families are sure to hold more exciting discoveries and will continue to reveal new strategies for combating human diseases.


Subject(s)
Apoptosis/physiology , Cardiovascular Diseases/physiopathology , Insect Proteins/physiology , Proteins , Proto-Oncogene Proteins c-bcl-2/physiology , Humans , Inhibitor of Apoptosis Proteins
4.
Cancer Res ; 60(16): 4386-90, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10969782

ABSTRACT

In this study, we sought to investigate in more detail the role of caspase-3 in apoptotic processes in cultured cells and in cell-free extracts of breast cancer cells. We present evidence that apoptosis of caspase-3-deficient MCF-7 breast cancer cells is defective in response to cisplatin treatment, as determined by chromatin condensation, nuclear fragmentation, DNA fragmentation, and release of cytochrome c from the mitochondria. Reconstitution of MCF-7 cells by stable transfection of CASP-3 cDNA restores all these defects and results in an extensive apoptosis after cisplatin treatment. We further show that in extracts from caspase-3-deficient MCF-7 cells, procaspase-9 processing is strongly impaired after stimulation with either cytochrome c or recombinant caspase-8. Reconstitution of MCF-7 cell extracts with procaspase-3 corrects this defect, resulting in an efficient and complete processing of procaspase-9. Together, our data define caspase-3 as an important integrator of the apoptotic process in MCF-7 breast cancer cells and reveal an essential function of caspase-3 for procaspase-9 processing.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Caspases/metabolism , Caspases/physiology , Cisplatin/pharmacology , Enzyme Precursors/metabolism , Apoptosis/physiology , Breast Neoplasms/drug therapy , Caspase 3 , Caspase 8 , Caspase 9 , Cell Extracts , Cytochrome c Group/physiology , Enzyme Activation , Humans , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Cells, Cultured
6.
Nat Cell Biol ; 2(6): 318-25, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10854321

ABSTRACT

Mitochondria trigger apoptosis by releasing caspase activators, including cytochrome c (cytC). Here we show, using a pH-sensitive green fluorescent protein (GFP), that mitochondria-dependent apoptotic stimuli (such as Bax, staurosporine and ultraviolet irradiation) induce rapid, Bcl-2-inhibitable mitochondrial alkalinization and cytosol acidification, followed by cytC release, caspase activation and mitochondrial swelling and depolarization. These events are not induced by mitochondria-independent apoptotic stimuli, such as Fas. Activation of cytosolic caspases by cytC in vitro is minimal at neutral pH, but maximal at acidic pH, indicating that mitochondria-induced acidification of the cytosol may be important for caspase activation; this finding is supported by results obtained from cells using protonophores. Cytosol acidification and cytC release are suppressed by oligomycin, a FoF1-ATPase/H +-pump inhibitor, but not by caspase inhibitors. Ectopic expression of Bax in wild-type, but not FoF1/H+-pump-deficient, yeast cells similarly results in mitochondrial matrix alkalinization, cytosol acidification and cell death. These findings indicate that mitochondria-mediated alteration of intracellular pH may be an early event that regulates caspase activation in the mitochondrial pathway for apoptosis.


Subject(s)
Apoptosis , Caspases/metabolism , Cytosol/metabolism , Mitochondria/metabolism , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Caspase Inhibitors , Cell Line , Cytochrome c Group/metabolism , Cytosol/drug effects , Cytosol/enzymology , Cytosol/radiation effects , Deoxyadenine Nucleotides/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/radiation effects , Mitochondrial Swelling/drug effects , Mitochondrial Swelling/radiation effects , Mutation , Oligomycins/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proton-Translocating ATPases/antagonists & inhibitors , Proton-Translocating ATPases/genetics , Proton-Translocating ATPases/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Staurosporine/antagonists & inhibitors , Staurosporine/pharmacology , Ultraviolet Rays , bcl-2-Associated X Protein , fas Receptor/physiology
7.
Proc Natl Acad Sci U S A ; 97(4): 1427-32, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10677478

ABSTRACT

We cloned a new inhibitor of apoptosis protein (IAP) homolog, SfIAP, from Spodoptera frugiperda Sf-21 cells, a host of insect baculoviruses. SfIAP contains two baculovirus IAP repeat domains followed by a RING domain. SfIAP has striking amino acid sequence similarity with baculoviral IAPs, CpIAP and OpIAP, suggesting that baculoviral IAPs may be host-derived genes. SfIAP and baculoviral CpIAP inhibit Bax but not Fas-induced apoptosis in human cells. Their apoptosis-suppressing activity in mammalian cells requires both baculovirus IAP repeat and RING domains. Further biochemical data suggest that SfIAP and CpIAP are specific inhibitors of mammalian caspase-9, the pinnacle caspase in the mitochondria/cytochrome c pathway for apoptosis, but are not inhibitors of downstream caspase-3 and caspase-7. Thus the mechanisms by which insect and baculoviral IAPs suppress apoptosis may involve inhibition of an insect caspase-9 homologue. Peptides representing the IAP-binding domain of the Drosophila cell death protein Grim abrogated human caspase suppression by SfIAP and CpIAP, implying evolutionary conservation of the functions of IAPs and their inhibitors.


Subject(s)
Apoptosis/drug effects , Bacterial Proteins/genetics , Caspase Inhibitors , Drosophila Proteins , Enzyme Inhibitors/chemistry , Insect Proteins/chemistry , Proteins , Spodoptera/chemistry , Amino Acid Sequence , Animals , Apoptosis/genetics , Bacterial Proteins/chemistry , Baculoviridae/chemistry , Caspase 9 , Cell Line , Cells, Cultured , Cloning, Molecular , Evolution, Molecular , Humans , Inhibitor of Apoptosis Proteins , Molecular Sequence Data , Neuropeptides/chemistry , Peptide Fragments/pharmacology , Sequence Alignment , Viral Proteins/chemistry , Viral Proteins/pharmacology
8.
EMBO J ; 18(19): 5242-51, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10508158

ABSTRACT

Several human inhibitor of apoptosis (IAP) family proteins function by directly inhibiting specific caspases in a mechanism that does not require IAP cleavage. In this study, however, we demonstrate that endogenous XIAP is cleaved into two fragments during apoptosis induced by the tumor necrosis factor family member Fas (CD95). The two fragments produced comprise the baculoviral inhibitory repeat (BIR) 1 and 2 domains (BIR1-2) and the BIR3 and RING (BIR3-Ring) domains of XIAP. Overexpression of the BIR1-2 fragment inhibits Fas-induced apoptosis, albeit at significantly reduced efficiency compared with full-length XIAP. In contrast, overexpression of the BIR3-Ring fragment results in a slight enhancement of Fas-directed apoptosis. Thus, cleavage of XIAP may be one mechanism by which cell death programs circumvent the anti-apoptotic barrier posed by XIAP. Interestingly, ectopic expression of the BIR3-Ring fragment resulted in nearly complete protection from Bax-induced apoptosis. Use of purified recombinant proteins revealed that BIR3-Ring is a specific inhibitor of caspase-9 whereas BIR1-2 is specific for caspases 3 and 7. Therefore XIAP possesses two different caspase inhibitory activities which can be attributed to distinct domains within XIAP. These data may provide an explanation for why IAPs have evolved with multiple BIR domains.


Subject(s)
Caspases/metabolism , Peptide Fragments/metabolism , Proteins/metabolism , Apoptosis/immunology , Base Sequence , Caspase Inhibitors , DNA Primers , Humans , Hydrolysis , Inhibitor of Apoptosis Proteins , Jurkat Cells , Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity , fas Receptor/immunology
9.
Proc Natl Acad Sci U S A ; 96(10): 5752-7, 1999 May 11.
Article in English | MEDLINE | ID: mdl-10318956

ABSTRACT

Caspase-9 is critical for cytochrome c (cyto-c)-dependent apoptosis and normal brain development. We determined that this apical protease in the cyto-c pathway for apoptosis resides inside mitochondria in several types of cells, including cardiomyocytes and many neurons. Caspase-9 is released from isolated mitochondria on treatment with Ca2+ or Bax, stimuli implicated in ischemic neuronal cell death that are known to induce cyto-c release from mitochondria. In neuronal cell culture models, apoptosis-inducing agents trigger translocation of caspase-9 from mitochondria to the nucleus, which is inhibitable by Bcl-2. Similarly, in an animal model of transient global cerebral ischemia, caspase-9 release from mitochondria and accumulation in nuclei was observed in hippocampal and other vulnerable neurons exhibiting early postischemic changes preceding apoptosis. Loss of mitochondrial barrier function during neuronal damage from ischemia or other insults therefore may play an important role in making certain caspases available to participate in apoptosis.


Subject(s)
Apoptosis , Brain Ischemia/metabolism , Brain/metabolism , Caspases/metabolism , Mitochondria/enzymology , Amino Acid Sequence , Animals , Calcium/pharmacology , Caspase 9 , Cell Nucleus/enzymology , Cytochrome c Group/metabolism , Enzyme Precursors/metabolism , Immunohistochemistry , Molecular Sequence Data , Myocardium/metabolism , Neurons/metabolism , PC12 Cells , Proto-Oncogene Proteins/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Reperfusion Injury/enzymology , bcl-2-Associated X Protein
10.
J Biol Chem ; 274(13): 8359-62, 1999 Mar 26.
Article in English | MEDLINE | ID: mdl-10085063

ABSTRACT

The recombinant form of the proapoptotic caspase-9 purified following expression in Escherichia coli is processed at Asp315, but largely inactive; however, when added to cytosolic extracts of human 293 cells it is activated 2000-fold in the presence of cytochrome c and dATP. Thus, the characteristic activities of caspase-9 are context-dependent, and its activation may not recapitulate conventional caspase activation mechanisms. To explore this hypothesis we produced recombinant forms of procaspase-9 containing mutations that disabled one or both of the interdomain processing sites of the zymogen. These mutants were able to activate downstream caspases, but only in the presence of cytosolic factors. The mutant with both processing sites abolished had 10% of the activity of wild-type, and was able to support apoptosis, with equal vigor to wild-type, when transiently expressed in 293 cells. Thus caspase-9 has an unusually active zymogen that does not require proteolytic processing, but instead is dependent on cytosolic factors for expression of its activity.


Subject(s)
Caspases/metabolism , Apoptosis/genetics , Caspase 9 , Caspases/genetics , Cell Line , Coumarins/metabolism , Cytochrome c Group/metabolism , Cytosol/metabolism , Deoxyadenine Nucleotides/metabolism , Enzyme Activation , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , Humans , Mutation/genetics , Oligopeptides/metabolism , Recombinant Proteins/metabolism
12.
J Clin Immunol ; 19(6): 388-98, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10634212

ABSTRACT

Caspases are cysteine proteases that are specific for aspastic acid residues. These enzymes have been extensively characterized as integral and highly conserved components of a variety of cell death programs. Cowpox and several insect viruses have evolved mechanisms that counter host cell suicide by encoding proteins that directly inhibit caspases-thereby allowing propagation of viral progeny within the host cell. It has only recently been elucidated, however, that endogenous cellular inhibitors of the caspases exist. To date five members of the inhibitor of apoptosis (IAP) family of proteins has been identified in humans and at least three of these have been shown directly to inhibit specific caspases. Thus, members of the IAP family of proteins are the only endogenous inhibitors of caspases known in mammals. Here we discuss the caspase and IAP families of proteins and review the data concerning their relationship.


Subject(s)
Apoptosis , Caspase Inhibitors , Cysteine Proteinase Inhibitors/physiology , Proteins/physiology , Viral Proteins/physiology , Animals , Caspases/physiology , Humans , Inhibitor of Apoptosis Proteins , Signal Transduction , X-Linked Inhibitor of Apoptosis Protein
13.
J Biol Chem ; 273(42): 27084-90, 1998 Oct 16.
Article in English | MEDLINE | ID: mdl-9765224

ABSTRACT

The apoptotic signal triggered by ligation of members of the death receptor family is promoted by sequential activation of caspase zymogens. We show here that in a purified system, the initiator caspases-8 and -10 directly process the executioner pro-caspase-3 with activation rates (kcat/Km) of 8.7 x 10(5) and 2.8 x 10(5) M-1 s-1, respectively. These rates are of sufficient magnitude to indicate direct processing in vivo. Differentially processed forms of caspase-3 that accumulate during its activation have similar rates of activation, activities, and specificities. The pattern and rate of caspase-8 induced activation of pro-caspase-3 in cytosolic extracts was the same as in a purified system. Moreover, immunodepletion of a putative intermediary in the pathway to activation, pro-caspase-9, was without consequence. Taken together these data demonstrate that the initiator caspase-8 can directly activate pro-caspase-3 without the requirement for an accelerator. The in vitro data thus help to deconvolute previous in vivo transfection studies which have debated the role of a direct versus indirect transmission of the apoptotic signal generated by ligation of death receptors.


Subject(s)
Caspases/metabolism , Enzyme Precursors/metabolism , Apoptosis , Caspase 10 , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/genetics , Cytosol/metabolism , Enzyme Activation , Enzyme Precursors/genetics , Granzymes , Kinetics , Models, Biological , Protein Processing, Post-Translational , Serine Endopeptidases/metabolism , Signal Transduction
14.
Proc Natl Acad Sci U S A ; 95(9): 4997-5002, 1998 Apr 28.
Article in English | MEDLINE | ID: mdl-9560217

ABSTRACT

Bax is a pro-apoptotic member of the Bcl-2 protein family that resides in the outer mitochondrial membrane. It is controversial whether Bax promotes cell death directly through its putative function as a channel protein versus indirectly by inhibiting cellular regulators of the cell death proteases (caspases). We show here that addition of submicromolar amounts of recombinant Bax protein to isolated mitochondria can induce cytochrome c (Cyt c) release, whereas a peptide representing the Bax BH3 domain was inactive. When placed into purified cytosol, neither mitochondria nor Bax individually induced proteolytic processing and activation of caspases. In contrast, the combination of Bax and mitochondria triggered release of Cyt c from mitochondria and induced caspase activation in cytosols. Supernatants from Bax-treated mitochondria also induced caspase processing and activation. Recombinant Bcl-XL protein abrogated Bax-induced release of Cyt c from isolated mitochondria and prevented caspase activation. In contrast, the broad-specificity caspase inhibitor benzyloxycarbonyl-valinyl-alaninyl-aspartyl-(0-methyl)- fluoromethylketone (zVAD-fmk) and the caspase-inhibiting protein X-IAP had no effect on Bax-induced release of Cyt c from mitochondria in vitro but prevented the subsequent activation of caspases in cytosolic extracts. Unlike Ca2+, a classical inducer of mitochondrial permeability transition, Bax did not induce swelling of mitochondria in vitro. Because the organellar swelling caused by permeability transition causes outer membrane rupture, the findings, therefore, dissociate these two events, implying that Bax uses an alternative mechanism for triggering release of Cyt c from mitochondria.


Subject(s)
Cytochrome c Group/metabolism , Mitochondria, Liver/drug effects , Proto-Oncogene Proteins/pharmacology , Animals , Apoptosis , Cell-Free System , Cyclosporine/pharmacology , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation , Female , Humans , Membrane Potentials/drug effects , Mice , Mitochondrial Swelling/drug effects , Permeability , Proto-Oncogene Proteins c-bcl-2/pharmacology , Rats , Recombinant Proteins , bcl-2-Associated X Protein , bcl-X Protein
15.
EMBO J ; 17(8): 2215-23, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9545235

ABSTRACT

Inhibitor of apoptosis (IAP) gene products play an evolutionarily conserved role in regulating programmed cell death in diverse species ranging from insects to humans. Human XIAP, cIAP1 and cIAP2 are direct inhibitors of at least two members of the caspase family of cell death proteases: caspase-3 and caspase-7. Here we compared the mechanism by which IAPs interfere with activation of caspase-3 and other effector caspases in cytosolic extracts where caspase activation was initiated by caspase-8, a proximal protease activated by ligation of TNF-family receptors, or by cytochrome c, which is released from mitochondria into the cytosol during apoptosis. These studies demonstrate that XIAP, cIAP1 and cIAP2 can prevent the proteolytic processing of pro-caspases -3, -6 and -7 by blocking the cytochrome c-induced activation of pro-caspase-9. In contrast, these IAP family proteins did not prevent caspase-8-induced proteolytic activation of pro-caspase-3; however, they subsequently inhibited active caspase-3 directly, thus blocking downstream apoptotic events such as further activation of caspases. These findings demonstrate that IAPs can suppress different apoptotic pathways by inhibiting distinct caspases and identify pro-caspase-9 as a new target for IAP-mediated inhibition of apoptosis.


Subject(s)
Apoptosis , Caspases , Cysteine Endopeptidases/metabolism , Cytochrome c Group/metabolism , Proteins/metabolism , Caspase 3 , Caspase 8 , Caspase 9 , Cell Extracts , Cell Line, Transformed , Cysteine Proteinase Inhibitors/metabolism , Cytosol , Enzyme Activation , Enzyme Precursors/metabolism , Humans , Inhibitor of Apoptosis Proteins , Protein Processing, Post-Translational , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured , Ubiquitin-Protein Ligases , X-Linked Inhibitor of Apoptosis Protein
16.
J Biol Chem ; 273(14): 7787-90, 1998 Apr 03.
Article in English | MEDLINE | ID: mdl-9525868

ABSTRACT

The inhibitor of apoptosis proteins (IAPs) constitute an evolutionarily conserved family of homologous proteins that suppress apoptosis induced by multiple stimuli. Some IAP family proteins, including XIAP, cIAP-1, and cIAP-2, can bind and directly inhibit selected caspases, a group of intracellular cell death proteases. These caspase-inhibiting IAP family proteins all contain three tandem BIR domains followed by a RING zinc finger domain. To determine the structural basis for caspase inhibition by XIAP, we analyzed the effects of various fragments of this IAP family protein on caspase activity in vitro and on apoptosis suppression in intact cells. The RING domain of XIAP failed to inhibit the activity of recombinant caspases-3 or -7, whereas a fragment of XIAP encompassing the three tandem BIR domains potently inhibited these caspases in vitro and blocked Fas (CD95)-induced apoptosis when expressed in cells. Further dissection of the XIAP protein demonstrated that only the second of the three BIR domains (BIR2) was capable of binding and inhibiting these caspases. The apparent inhibition constants (Ki) for BIR2-mediated inhibition of caspases-3 and -7 were 2-5 nM, indicating that this single BIR domain possesses potent anti-caspase activity. Expression of the BIR2 domain in cells also partially suppressed Fas-induced apoptosis and blocked cytochrome c-induced processing of caspase-9 in cytosolic extracts, whereas BIR1 and BIR3 did not. These findings identify BIR2 as the minimal caspase-inhibitory domain of XIAP and indicate that a single BIR domain can be sufficient for binding and inhibiting caspases.


Subject(s)
Apoptosis , Caspases , Cysteine Endopeptidases/metabolism , Peptide Fragments/metabolism , Proteins/genetics , Animals , Apoptosis/genetics , Caspase 3 , Caspase 7 , Cell Line , Cysteine Endopeptidases/genetics , Escherichia coli , Gene Expression Regulation , Humans , Peptide Fragments/genetics , Proteins/chemistry , Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , X-Linked Inhibitor of Apoptosis Protein
17.
J Biol Chem ; 273(10): 5461-7, 1998 Mar 06.
Article in English | MEDLINE | ID: mdl-9488668

ABSTRACT

Ubiquitylated proteins are degraded by the 26 S protease, an enzyme complex that contains 30 or more unique subunits. One of these proteins, subunit 5a (S5a), has been shown to bind ubiquitin-lysozyme conjugates and free polyubiquitin chains. Using deletional analysis, we have identified in the carboxyl-terminal half of human S5a, two independent polyubiquitin binding sites whose sequences are highly conserved among higher eukaryotic S5a homologs. The sites are approximately 30-amino acids long and are separated by 50 intervening residues. When expressed as small fragments or when present in full-length S5a molecules, the sites differ at least 10-fold in their apparent affinity for polyubiquitin chains. Each binding site contains 5 hydrophobic residues that form an alternating pattern of large and small side chains, e.g. Leu-Ala-Leu-Ala-Leu, and this pattern is essential for binding ubiquitin chains. Based on the importance of the alternating hydrophobic residues in the binding sites and previous studies showing that a hydrophobic patch on the surface of ubiquitin is essential for proteolytic targeting, we propose a model for molecular recognition of polyubiquitin chains by S5a.


Subject(s)
Biopolymers/metabolism , Peptide Hydrolases/chemistry , Proteasome Endopeptidase Complex , Ubiquitins/metabolism , Amino Acid Sequence , Binding Sites/physiology , Conserved Sequence/genetics , Humans , Molecular Sequence Data , Muramidase/metabolism , Mutagenesis, Site-Directed/genetics , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptide Hydrolases/physiology , Polyubiquitin , Protein Binding/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Sequence Deletion/genetics , Sequence Homology, Amino Acid
18.
Nature ; 388(6639): 300-4, 1997 Jul 17.
Article in English | MEDLINE | ID: mdl-9230442

ABSTRACT

The inhibitor-of-apoptosis (IAP) family of genes has an evolutionarily conserved role in regulating programmed cell death in animals ranging from insects to humans. Ectopic expression of human IAP proteins can suppress cell death induced by a variety of stimuli, but the mechanism of this inhibition was previously unknown. Here we show that human X-chromosome-linked IAP directly inhibits at least two members of the caspase family of cell-death proteases, caspase-3 and caspase-7. As the caspases are highly conserved throughout the animal kingdom and are the principal effectors of apoptosis, our findings suggest how IAPs might inhibit cell death, providing evidence for a mechanism of action for these mammalian cell-death suppressors.


Subject(s)
Apoptosis/physiology , Caspases , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/physiology , Proteins/physiology , Proto-Oncogene Proteins c-bcl-2 , X Chromosome , Amino Acid Sequence , Caspase 1 , Caspase 3 , Caspase 7 , Cell Nucleus/metabolism , Cell-Free System , Cytochrome c Group/metabolism , Cytosol/metabolism , Enzyme Activation , Genetic Linkage , Humans , Jurkat Cells , Molecular Sequence Data , Protein Processing, Post-Translational , Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Recombinant Proteins/metabolism , Transfection , X-Linked Inhibitor of Apoptosis Protein , bcl-2-Associated X Protein
19.
EMBO J ; 16(23): 6914-25, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9384571

ABSTRACT

The inhibitor of apoptosis (IAP) family of proteins are highly conserved through evolution. However, the mechanisms by which these proteins interfere with apoptotic cell death have been enigmatic. Recently, we showed that one of the human IAP family proteins, XIAP, can bind to and potently inhibit specific cell death proteases (caspases) that function in the distal portions of the proteolytic cascades involved in apoptosis. In this study, we investigated three of the other known members of the human IAP family, c-IAP-1, c-IAP-2 and NAIP. Similarly to XIAP, in vitro binding experiments indicated that c-IAP-1 and c-IAP-2 bound specifically to the terminal effector cell death proteases, caspases-3 and -7, but not to the proximal protease caspase-8, caspases-1 or -6. In contrast, NAIP failed to bind tightly to any of these proteases. Recombinant c-IAP-1 and c-IAP-2 also inhibited the activity of caspases-3 and -7 in vitro, with estimated Kis of <=0.1 microM, whereas NAIP did not. The BIR domain-containing region of c-IAP-1 and c-IAP-2 was sufficient for inhibition of these caspases, though proteins that retained the RING domain were somewhat more potent. Utilizing a cell-free system in which caspases were activated in cytosolic extracts by addition of cytochrome c, c-IAP-1 and c-IAP-2 inhibited both the generation of caspase activities and proteolytic processing of pro-caspase-3. Similar results were obtained in intact cells when c-IAP-1 and c-IAP-2 were overexpressed by gene transfection, and apoptosis was induced by the anticancer drug, etoposide. Cleavage of c-IAP-1 or c-IAP-2 was not observed when interacting with the caspases, implying a different mechanism from the baculovirus p35 protein, the broad spectrum suicide inactivator of caspases. Taken together, these findings suggest that c-IAP-1 and c-IAP-2 function similarly to XIAP by inhibiting the distal cell death proteases, caspases-3 and -7, whereas NAIP presumably inhibits apoptosis via other targets.


Subject(s)
Apoptosis , Caspases , Cysteine Endopeptidases/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Caspase 3 , Caspase 7 , Caspase 8 , Caspase 9 , Cytochrome c Group/pharmacology , Cytosol/drug effects , Cytosol/enzymology , Enzyme Activation , Etoposide/pharmacology , Humans , Inhibitor of Apoptosis Proteins , Protein Binding , Protein Processing, Post-Translational , Recombinant Proteins/pharmacology , Signal Transduction , Ubiquitin-Protein Ligases
20.
FEBS Lett ; 381(1-2): 143-8, 1996 Feb 26.
Article in English | MEDLINE | ID: mdl-8641424

ABSTRACT

S5a is a subunit of the 26S protease that binds and presumably selects multiubiquitinated proteins for destruction. We recently identified an Arabidopsis protein, MBP1, that is physically, immunologically and biochemically similar to S5a from the human erythrocyte 26S protease. Based upon the MBP1 cDNA sequence we have now isolated a HeLa cell cDNA coding for human S5a. The HeLa cDNA sequence is highly similar to MBP1 and it encodes peptides obtained directly from human erythrocyte S5a. Moreover, expression of the isolated cDNA in E. coli results in a recombinant protein with an apparent molecular mass and multiubiquitin binding properties that match those of human S5a obtained from the purified 26S enzyme.


Subject(s)
Arabidopsis Proteins , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex , Ubiquitins/metabolism , Amino Acid Sequence , Animals , Arabidopsis/enzymology , Base Sequence , Binding Sites , Carrier Proteins/chemistry , Cloning, Molecular , Drosophila , Erythrocytes/enzymology , Gene Expression , HeLa Cells , Humans , Macromolecular Substances , Molecular Sequence Data , Peptide Hydrolases/biosynthesis , Peptide Hydrolases/chemistry , RNA-Binding Proteins , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...