Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Biomolecules ; 11(2)2021 01 25.
Article in English | MEDLINE | ID: mdl-33503908

ABSTRACT

The necrosome is a large-molecular-weight complex in which the terminal effector of the necroptotic pathway, Mixed Lineage Kinase Domain-Like protein (MLKL), is activated to induce necroptotic cell death. The precise mechanism of MLKL activation by the upstream kinase, Receptor Interacting Serine/Threonine Protein Kinase 3 (RIPK3) and the role of Receptor Interacting Serine/Threonine Protein Kinase 1 (RIPK1) in mediating MLKL activation remain incompletely understood. Here, we reconstituted human necrosome interactions in yeast by inducible expression of these necrosome effectors. Functional interactions were reflected by the detection of phosphorylated MLKL, plasma membrane permeabilization, and reduced proliferative potential. Following overexpression of human necrosome effectors in yeast, MLKL aggregated in the periphery of the cell, permeabilized the plasma membrane and compromised clonogenic potential. RIPK1 had little impact on RIPK3/MLKL-mediated yeast lethality; however, it exacerbated the toxicity provoked by co-expression of MLKL with a RIPK3 variant bearing a mutated RHIM-domain. Small molecule necroptotic inhibitors necrostatin-1 and TC13172, and viral inhibitors M45 (residues 1-90) and BAV_Rmil, abated the yeast toxicity triggered by the reconstituted necrosome. This yeast model provides a convenient tool to study necrosome protein interactions and to screen for and characterize potential necroptotic inhibitors.


Subject(s)
Necrosis , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/metabolism , Apoptosis , Cell Death , Cell Membrane/metabolism , Cell Proliferation , Enzyme Inhibitors , Green Fluorescent Proteins/metabolism , Humans , Phosphorylation , Plasmids/metabolism , Polymerase Chain Reaction , Protein Domains
2.
Vet Comp Oncol ; 16(4): 544-553, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29998615

ABSTRACT

Osteosarcoma, a common malignancy in large dog breeds, typically metastasises from long bones to lungs and is usually fatal within 1 to 2 years of diagnosis. Better therapies are needed for canine patients and their human counterparts, a third of whom die within 5 years of diagnosis. We compared the in vitro sensitivity of canine osteosarcoma cells derived from 4 tumours to the currently used chemotherapy drugs doxorubicin and carboplatin, and 4 new anti-cancer drugs. Agents targeting histone deacetylases or PARP were ineffective. Two of the 4 cell lines were somewhat sensitive to the BH3-mimetic navitoclax. The proteasome inhibitor bortezomib potently induced caspase-dependent apoptosis, at concentrations substantially lower than levels detected in the bones and lungs of treated rodents. Co-treatment with bortezomib and either doxorubicin or carboplatin was more toxic to canine osteosarcoma cells than each agent alone. Newer proteasome inhibitors carfilzomib, ixazomib, oprozomib and delanzomib manifested similar activities to bortezomib. Human osteosarcoma cells were as sensitive to bortezomib as the canine cells, but slightly less sensitive to the newer drugs. Human osteoblasts were less sensitive to proteasome inhibition than osteosarcoma cells, but physiologically relevant concentrations were toxic. Such toxicity, if replicated in vivo, may impair bone growth and strength in adolescent human osteosarcoma patients, but may be tolerated by canine patients, which are usually diagnosed later in life. Proteasome inhibitors such as bortezomib may be useful for treating canine osteosarcoma, and ultimately may improve outcomes for human patients if their osteoblasts survive exposure in vivo, or if osteoblast toxicity can be managed.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Dog Diseases/drug therapy , Osteosarcoma/drug therapy , Proteasome Inhibitors/therapeutic use , Aniline Compounds/therapeutic use , Animals , Bone Neoplasms/veterinary , Boron Compounds/therapeutic use , Boronic Acids/therapeutic use , Bortezomib/therapeutic use , Carboplatin/therapeutic use , Cell Line, Tumor , Dogs , Doxorubicin/therapeutic use , Glycine/analogs & derivatives , Glycine/therapeutic use , Humans , Oligopeptides/therapeutic use , Osteosarcoma/veterinary , Sulfonamides/therapeutic use , Threonine/analogs & derivatives , Threonine/therapeutic use
3.
Int Rev Cell Mol Biol ; 332: 155-212, 2017.
Article in English | MEDLINE | ID: mdl-28526132

ABSTRACT

Although caspase-2 is a highly conserved protease that has received a lot of research attention, consensus about its roles and the molecular mechanisms that underpin them has been elusive. Recent improvements to our understanding of the activities of caspase-2 have been facilitated by the development and refinement of techniques allowing identification of cellular processes instigated by this caspase. Following DNA damage, caspase-2 can be activated in a molecular complex called the "PIDDosome"; however, other stimuli provoke caspase-2-dependent activities that do not appear to involve this complex. Further research is needed into the mechanisms that activate caspase-2, and the substrates that it cleaves to accomplish its functions. Apart from DNA damage, caspase-2 has also been implicated in responses to other cellular stresses including oxidative damage, endoplasmic reticulum stress, and aberrant mitotic signaling. Caspase-2 sensitized animals fed diets high in fat and sugar to glucose intolerance and liver disease, so drugs that target this protease may be useful to prevent or treat metabolic conditions. Caspase-2 loss enhanced the survival of retinal ganglion cells following optic nerve damage, prompting hope that caspase-2 inhibitors may help treat optic nerve injuries. Caspase-2 predisposed animals to neuroblastoma but tended to provide protection against oncogene-driven cancers. Intriguingly, caspase-2 facilitated host cell death following viral or bacterial infection, raising the possibility that its evolutionary retention may reflect its ability to induce defensive apoptosis following intracellular infection.


Subject(s)
Caspase 2/metabolism , Animals , DNA Damage , Endoplasmic Reticulum Stress , Enzyme Activation , Humans , Mitosis , Oxidative Stress
4.
Cell Death Dis ; 4: e619, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23640461

ABSTRACT

Development of drugs targeting Bcl-2 relatives and caspases, for treating diseases including cancer and inflammatory disorders, often involves measuring interactions with recombinant target molecules, and/or monitoring cancer cell killing in vitro. Here, we present yeast-based methods for evaluating drug-mediated inhibition of Bcl-2 relatives or caspases. Active Bax and caspases kill Saccharomyces cerevisiae, and pro-survival Bcl-2 proteins can inhibit Bax-induced yeast death. By measuring the growth or adenosine triphosphate content of transformants co-expressing Bax with pro-survival Bcl-2 relatives, we found that the Bcl-2 antagonist drugs ABT-737 or ABT-263 abolished Bcl-2 or Bcl-xL function and reduced Bcl-w activity, but failed to inhibit Mcl-1, A1 or the poxvirus orthologs DPV022 and SPPV14. Using this technique, we also demonstrated that adenoviral E1B19K was resistant to these agents. The caspase inhibitor Q-VD-OPh suppressed yeast death induced by caspases 1 and 3. Yeast engineered to express human apoptotic regulators enable simple, automatable assessment of the activity and specificity of candidate drugs targeting Bcl-2 relatives or caspases.


Subject(s)
Caspases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Caspase Inhibitors/pharmacology , Caspases/chemistry , Nitrophenols/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Quinolines/pharmacology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Sulfonamides/pharmacology , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
5.
Cell Death Dis ; 2: e242, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22170098

ABSTRACT

Many viruses express proteins which prevent the host cell death that their infection would otherwise provoke. Some insect viruses suppress host apoptosis through the expression of caspase inhibitors belonging to the P35 superfamily. Although a number of P35 relatives have been identified, Autographa californica (Ac) P35 and Spodoptera littoralis (Spli) P49 have been the most extensively characterized. AcP35 was found to inhibit caspases via a suicide substrate mechanism: the caspase cleaves AcP35 within its 'reactive site loop' then becomes trapped, irreversibly bound to the cleaved inhibitor. The Maruca vitrata multiple nucleopolyhedrovirus encodes a P35 family member (MaviP35) that exhibits 81% identity to AcP35. We found that this relative shared with AcP35 the ability to inhibit mammalian and insect cell death. Caspase-mediated cleavage within the MaviP35 reactive site loop occurred at a sequence distinct from that in AcP35, and the inhibitory profiles of the two P35 relatives differed. MaviP35 potently inhibited human caspases 2 and 3, DCP-1, DRICE and CED-3 in vitro, but (in contrast to AcP35) only weakly suppressed the proteolytic activity of the initiator human caspases 8, 9 and 10. Although MaviP35 inhibited the AcP35-resistant caspase DRONC in yeast, and was sensitive to cleavage by DRONC in vitro, MaviP35 failed to inhibit the proteolytic activity of bacterially produced DRONC in vitro.


Subject(s)
Baculoviridae/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Apoptosis , Caspase Inhibitors , Caspases/genetics , Caspases/metabolism , Lepidoptera/virology , Molecular Sequence Data , Protein Structure, Tertiary , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Spodoptera/virology , Viral Proteins/chemistry
6.
Oncogene ; 29(36): 5048-60, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20639907

ABSTRACT

Chemotherapy and radiotherapy commonly damage DNA and trigger p53-dependent apoptosis through intrinsic apoptotic pathways. Two unfortunate consequences of this mechanism are resistance due to blockade of p53 or intrinsic apoptosis pathways, and mutagenesis of non-malignant surviving cells which can impair cellular function or provoke second malignancies. Death ligand-based drugs, such as tumor necrosis factor-related apoptosis inducing ligand (TRAIL), stimulate extrinsic apoptotic signaling, and may overcome resistance to treatments that induce intrinsic apoptosis. As death receptor ligation does not damage DNA as a primary mechanism of pro-apoptotic action, we hypothesized that surviving cells would remain genetically unscathed, suggesting that death ligand-based therapies may avoid some of the adverse effects associated with traditional cancer treatments. Surprisingly, however, treatment with sub-lethal concentrations of TRAIL or FasL was mutagenic. Mutations arose in viable cells that contained active caspases, and overexpression of the caspase-8 inhibitor crmA or silencing of caspase-8 abolished TRAIL-mediated mutagenesis. Downregulation of the apoptotic nuclease caspase-activated DNAse (CAD)/DNA fragmentation factor 40 (DFF40) prevented the DNA damage associated with TRAIL treatment. Although death ligands do not need to damage DNA in order to induce apoptosis, surviving cells nevertheless incur DNA damage after treatment with these agents.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Mutation , Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Cisplatin/pharmacology , Deoxyribonucleases/antagonists & inhibitors , Deoxyribonucleases/genetics , Deoxyribonucleases/metabolism , Drug Resistance, Neoplasm/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Mice , Models, Biological , Mutation/drug effects , Neoplasms/genetics , Neoplasms/metabolism , RNA, Small Interfering/pharmacology
7.
Br J Cancer ; 99(2): 294-304, 2008 Jul 22.
Article in English | MEDLINE | ID: mdl-18594532

ABSTRACT

TRAIL/Apo-2L has shown promise as an anti-glioma drug, based on investigations of TRAIL sensitivity in established glioma cell lines, but it is not known how accurately TRAIL signalling pathways of glioma cells in vivo are reproduced in these cell lines in vitro. To replicate as closely as possible the in vivo behaviour of malignant glioma cells, 17 early passage glioma cell lines and 5 freshly resected gliomas were exposed to TRAIL-based agents and/or chemotherapeutic drugs. Normal human hepatocytes and astrocytes and established glioma cell lines were also tested. Cross-linked TRAIL, but not soluble TRAIL, killed both normal cell types and cells from three tumours. Cells from only one glioma were killed by soluble TRAIL, although only inefficiently. High concentrations of cisplatin were lethal to glioma cells, hepatocytes and astrocytes. Isolated combinations of TRAIL and chemotherapy drugs were more toxic to particular gliomas than normal cells, but no combination was generally selective for glioma cells. This study highlights the widespread resistance of glioma cells to TRAIL-based agents, but suggests that a minority of high-grade glioma patients may benefit from particular combinations of TRAIL and chemotherapy drugs. In vitro sensitivity assays may help identify effective drug combinations for individual glioma patients.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Glioma/drug therapy , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Adult , Aged , Antineoplastic Agents/administration & dosage , Astrocytes/drug effects , Carboplatin/administration & dosage , Cell Line, Tumor , Cisplatin/administration & dosage , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Drug Screening Assays, Antitumor , Etoposide/administration & dosage , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioma/pathology , Hepatocytes/drug effects , Humans , Lomustine/administration & dosage , Male , Membrane Glycoproteins/administration & dosage , Middle Aged , Procarbazine/administration & dosage , Recombinant Fusion Proteins/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Temozolomide , Tumor Necrosis Factor-alpha/administration & dosage , Vincristine/administration & dosage
8.
Cell Death Differ ; 14(2): 348-57, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16794601

ABSTRACT

Direct IAP binding protein with low pI/second mitochondrial activator of caspases, HtrA2/Omi and GstPT/eRF3 are mammalian proteins that bind via N-terminal inhibitor of apoptosis protein (IAP) binding motifs (IBMs) to the baculoviral IAP repeat (BIR) domains of IAPs. These interactions can prevent IAPs from inhibiting caspases, or displace active caspases, thereby promoting cell death. We have identified several additional potential IAP antagonists, including glutamate dehydrogenase (GdH), Nipsnap 3 and 4, CLPX, leucine-rich pentatricopeptide repeat motif-containing protein and 3-hydroxyisobutyrate dehydrogenase. All are mitochondrial proteins from which N-terminal import sequences are removed generating N-terminal IBMs. Whereas most of these proteins have alanine at the N-terminal position, as observed for previously described antagonists, GdH has an N-terminal serine residue that is essential for X-linked IAP (XIAP) interaction. These newly described IAP binding proteins interact with XIAP mainly via BIR2, with binding eliminated or significantly reduced by a single point mutation (D214S) within this domain. Through this interaction, many are able to antagonise XIAP inhibition of caspase 3 in vitro.


Subject(s)
Inhibitor of Apoptosis Proteins/metabolism , Mammals/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Alanine , Amino Acid Motifs , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Caspase Inhibitors , Enzyme Inhibitors/pharmacology , Glutamate Dehydrogenase/chemistry , Glutamate Dehydrogenase/metabolism , High-Temperature Requirement A Serine Peptidase 2 , Humans , Inhibitor of Apoptosis Proteins/chemistry , Leucine-Rich Repeat Proteins , Mice , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Proteins/chemistry , Proteins/metabolism , Proteomics , Serine , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , X-Linked Inhibitor of Apoptosis Protein/chemistry , X-Linked Inhibitor of Apoptosis Protein/metabolism
9.
Leukemia ; 20(7): 1270-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16673017

ABSTRACT

Telomerase is a complex ribonucleoprotein enzyme that exhibits elevated activity in the majority of cases of human leukemia. We have previously shown that retroviral expression of the catalytic subunit of telomerase, human telomerase reverse transcriptase (hTERT), in human cord blood CD34+ cells leads to an enhanced survival of mature hematopoietic cells. The mechanism for this pro-survival effect is not known. Here, we show that telomerase may play a role in leukemogenesis as a survival factor, independent of its role in maintaining telomere length. Retroviral expression of hTERT in the cytokine-dependent, human hematopoietic progenitor cell line, TF-1, resulted in the survival of cells following the withdrawal of cytokine, with protection from apoptosis, but did not promote unlimited replicative potential. This hTERT-mediated effect on cell survival does not involve Bcl-2 family members, results in accumulation of cells in G1 and appears to operate via autocrine expression of IL-3 and activation of the p53/p21 pathway. Survival in the absence of cytokine stimulation was also observed following retroviral expression of hTERT in normal cord blood CD34+ cells. This study demonstrates a novel pro-survival role for hTERT and may have important implications for the role of hTERT in the pathogenesis of leukemia and drug resistance.


Subject(s)
Apoptosis/physiology , Cytokines/pharmacology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Telomerase/metabolism , Apoptosis/drug effects , Cell Cycle/physiology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Clone Cells , Fetal Blood/cytology , Gene Expression Regulation, Enzymologic , Humans , Leukemia/metabolism , Leukemia/physiopathology , Retroviridae/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Telomerase/genetics , Transduction, Genetic
10.
Apoptosis ; 11(4): 509-17, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16538379

ABSTRACT

This study characterised the impact of active metazoan apoptotic proteases (caspases) on Saccharomyces cerevisiae viability. Expression of active caspase-3 or caspase-8 in yeast ruptured plasma and nuclear membranes and dramatically impaired clonogenic survival, but did not damage DNA. Deletion of the proposed yeast apoptosis regulators YCA1 or Aif1p did not affect the ability of human, insect or nematode caspases to kill yeast. These data indicate that expression of active metazoan caspases causes irreversible damage to yeast membranes and organelles, in a manner independent of YCA1 and Aif1p.


Subject(s)
Apoptosis , Caspases/metabolism , NADH, NADPH Oxidoreductases/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/cytology , Actins/chemistry , Actins/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans/enzymology , Caspase 3 , Caspase 8 , Caspases/genetics , Caspases/physiology , Cell Survival , Drosophila/enzymology , Gene Deletion , Humans , Molecular Sequence Data , NADH, NADPH Oxidoreductases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/genetics , Transgenes
11.
Cell Death Differ ; 11(12): 1309-16, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15543163

ABSTRACT

A genetically defined pathway orchestrates the removal of 131 of the 1090 somatic cells generated during the development of the hermaphrodite nematode Caenorhabditis elegans. Regulation of apoptosis is highly evolutionarily conserved and the nematode cell death pathway is a valuable model for studying mammalian apoptotic pathways, the dysregulation of which can contribute to numerous diseases. The nematode caspase CED-3 is ultimately responsible for the destruction of worm cells in response to apoptotic signals, but it must first be activated by CED-4. CED-9 inhibits programmed cell death and considerable data have demonstrated that CED-9 can directly bind and inhibit CED-4. However, it has been suggested that CED-9 may also directly inhibit CED-3. In this study, we used a yeast-based system and biochemical approaches to explore this second potential mechanism of action. While we confirmed the ability of CED-9 to inhibit CED-4, our data argue that CED-9 can not directly inhibit CED-3.


Subject(s)
Apoptosis/physiology , Caenorhabditis elegans Proteins/metabolism , Caspases/metabolism , Proto-Oncogene Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Signal Transduction/physiology , Animals , Apoptosis Regulatory Proteins , Caenorhabditis elegans , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Caspases/chemistry , Caspases/genetics , Enzyme Activation/physiology , Feedback, Physiological/physiology , Gene Expression Regulation, Fungal/physiology , In Vitro Techniques , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2
12.
Cell Death Differ ; 9(12): 1311-20, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12478468

ABSTRACT

This study characterized the ability of a new member of the p35 family, p49, to inhibit a number of mammalian and insect caspases. p49 blocked apoptosis triggered by treatment with Fas ligand (FasL), Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) or ultraviolet (UV) radiation but provided negligible protection against apoptosis induced by the chemotherapeutic drug cisplatin. The caspase cleavage site in p49 was determined, and mutation of the P1 residue of this site abolished the ability of p49 to inhibit caspases, implying that p49 inhibits caspases through an analogous suicide-substrate mechanism to p35. Unlike p35, p49 inhibited the upstream insect caspase DRONC.


Subject(s)
Apoptosis/genetics , Drosophila Proteins , Eukaryotic Cells/metabolism , Immediate-Early Proteins/metabolism , Trans-Activators/metabolism , Viral Proteins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Apoptosis Regulatory Proteins , Caspase Inhibitors , Caspases/genetics , Caspases/metabolism , Cells, Cultured , Cisplatin/pharmacology , Drosophila melanogaster , Eukaryotic Cells/drug effects , Eukaryotic Cells/radiation effects , Fas Ligand Protein , Humans , Immediate-Early Proteins/genetics , Membrane Glycoproteins/pharmacology , Molecular Sequence Data , Mutation/genetics , Saccharomyces cerevisiae , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , TNF-Related Apoptosis-Inducing Ligand , Trans-Activators/genetics , Tumor Necrosis Factor-alpha/pharmacology , Ultraviolet Rays , Viral Proteins/genetics
13.
Neuro Oncol ; 3(4): 229-40, 2001 10.
Article in English | MEDLINE | ID: mdl-11584892

ABSTRACT

Fas (APO-1/CD95/TNFRSF6) is a member of the tumor necrosis/nerve growth factor receptor family that signals apoptotic cell death in sensitive cells. Expression of Fas and its agonistic ligand (FasL/TNFSF6) was investigated in ex vivo pediatric brain tumor specimens of various histologic types. Fas expression was identified in all of the 18 tumors analyzed by flow cytometry and immunohistochemistry. FasL expression was identified in most of the 13 tumors analyzed by both Western analysis and immunohistochemistry. Nine of these tumor specimens were treated with either the agonistic anti-Fas antibody (APO-1) in combination with protein A or FasL in short-term cytotoxicity assays. Sensitivity to apoptosis induced by the topoisomerase II inhibitor, etoposide, was also assessed. Despite the presence of Fas, all the specimens analyzed demonstrated a high degree of resistance to Fas-mediated apoptosis. These 9 specimens also showed a high degree of resistance to etoposide. Only 2 of the 9 specimens were susceptible to etoposide-induced cell death, whereas only 3 were sensitive to Fas-mediated apoptosis. One brain tumor was sensitive to both Fas ligation and etoposide treatment. This contrasted with the high degree of susceptibility to both etoposide- and Fas-induced apoptosis observed in the reference Jurkat cell line. The results suggest that Fas expression may be a general feature of tumors of the CNS and that a significant degree of resistance to Fas-mediated apoptosis may exist in ex vivo pediatric brain tumor specimens.


Subject(s)
Apoptosis/drug effects , Brain Neoplasms/chemistry , Drug Resistance, Neoplasm , Membrane Glycoproteins/analysis , Neoplasm Proteins/analysis , fas Receptor/analysis , Adolescent , Animals , Antineoplastic Agents/pharmacology , Astrocytoma/chemistry , Cell Survival , Child , Child, Preschool , Colorimetry , Culture Media, Conditioned , DNA, Complementary/genetics , Etoposide/pharmacology , Fas Ligand Protein , Female , Ganglioglioma/chemistry , Germinoma/chemistry , Glioblastoma/chemistry , Humans , Infant , Jurkat Cells/drug effects , Male , Medulloblastoma/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Neoplasm Proteins/physiology , Neuroblastoma/pathology , Neuroectodermal Tumors, Primitive/chemistry , Pineal Gland/chemistry , Pinealoma/chemistry , Recombinant Fusion Proteins/physiology , Transfection , Tumor Cells, Cultured , fas Receptor/physiology
14.
Oncogene ; 20(41): 5789-98, 2001 Sep 13.
Article in English | MEDLINE | ID: mdl-11593384

ABSTRACT

FasL and TNF-related apoptosis-inducing ligand (TRAIL) belong to a subgroup of the TNF superfamily which induce apoptosis by binding to their death domain containing receptors. In the present study we have utilized a panel of seven cell lines derived from human malignant gliomas to characterize molecular pathways through which FasL and TRAIL induce apoptosis in sensitive glioma cells and the mechanisms of resistance in cell lines which survive the death stimuli. Our findings indicate that FADD and Caspase-8 are essential for FasL and TRAIL mediated apoptosis in glioma cells. One sensitive cell line (D270) can be protected from FasL and TRAIL induced death by anti-apoptotic Bcl-2 family members while another (D645) cannot, implying that these lines may represent glioma examples of type II and type I cells respectively. For the first time we demonstrate resistance to FasL but not to TRAIL within the one glioma cell line. Furthermore, we report distinct mechanisms of resistance within different glioma lines, including downregulation of Caspase-8 in U373MG. Cycloheximide sensitized four of the resistant cell lines suggesting the presence of labile inhibitors. None of the known apoptosis inhibitors examined accounted for the observed resistance, suggesting novel inhibitors may exist in glioma cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/physiology , Glioma/physiopathology , Membrane Glycoproteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Carrier Proteins/metabolism , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolism , Cisplatin/pharmacology , Cycloheximide/pharmacology , Drug Resistance, Neoplasm , Enzyme Activation , Fas Ligand Protein , Fas-Associated Death Domain Protein , Glioma/metabolism , Humans , Ligands , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured/drug effects , bcl-X Protein
15.
Apoptosis ; 6(5): 331-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11483856

ABSTRACT

We have reconstituted the Apaf-1-activated apoptosis mechanism in Sacchromyces cerevisiae such that the presence of a constitutively active form of Apaf-1 together with both Caspase-9 and Caspase-3 results in yeast death. This system is a good model of the Apaf-1-activated pathway in mammalian cells: MIHA (XIAP/hILP), and to a lesser degree MIHB (c-IAP1/HIAP2) and MIHC (c-IAP-2/HIAP1) can inhibit caspases in this system, and protection by IAPs (inhibitor of apoptosis) can be abrogated by coexpression of the Drosophila pro-apoptotic proteins HID and GRIM or the mammalian protein DIABLO/Smac. Using this system we demonstrate that unlike DIABLO/Smac, other proteins which interact with mammalian IAPs (TAB-1, Zap-1, Traf-1 and Traf-2) do not act to antagonise IAP- mediated caspase inhibition.


Subject(s)
Apoptosis , Caspase Inhibitors , Drosophila Proteins , Proteins/antagonists & inhibitors , Proteins/physiology , Saccharomyces cerevisiae/genetics , Animals , Apoptotic Protease-Activating Factor 1 , Carrier Proteins/physiology , Caspase 3 , Caspase 9 , Caspases/physiology , Inhibitor of Apoptosis Proteins , Mitochondrial Proteins/physiology , Neuropeptides/physiology , Proteins/genetics , Saccharomyces cerevisiae/cytology , Transfection , Viral Proteins/physiology , X-Linked Inhibitor of Apoptosis Protein
16.
EMBO J ; 20(12): 3114-23, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11406588

ABSTRACT

XIAP is a mammalian inhibitor of apoptosis protein (IAP). To determine residues within the second baculoviral IAP repeat (BIR2) required for inhibition of caspase 3, we screened a library of BIR2 mutants for loss of the ability to inhibit caspase 3 toxicity in the yeast Schizosaccharomyces pombe. Four of the mutations, not predicted to affect the structure of the BIR fold, clustered together on the N-terminal region that flanks BIR2, suggesting that this is a site of interaction with caspase 3. Introduction of these mutations into full-length XIAP reduced caspase 3 inhibitory activity up to 500-fold, but did not affect its ability to inhibit caspase 9 or interact with the IAP antagonist DIABLO. Furthermore, these mutants retained full ability to inhibit apoptosis in transfected cells, demonstrating that although XIAP is able to inhibit caspase 3, this activity is dispensable for inhibition of apoptosis by XIAP in vivo.


Subject(s)
Apoptosis , Caspase Inhibitors , Mitochondrial Proteins , Proteins/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/metabolism , Caspase 3 , Caspase 9 , Caspases/genetics , Caspases/metabolism , Enzyme Activation , Gene Expression , Inhibitor of Apoptosis Proteins , Molecular Sequence Data , Mutagenesis , Proteins/genetics , Proteins/physiology , Schizosaccharomyces , Ultraviolet Rays , Viral Proteins/genetics , X-Linked Inhibitor of Apoptosis Protein
17.
J Cell Biol ; 152(3): 483-90, 2001 Feb 05.
Article in English | MEDLINE | ID: mdl-11157976

ABSTRACT

MIHA is an inhibitor of apoptosis protein (IAP) that can inhibit cell death by direct interaction with caspases, the effector proteases of apoptosis. DIABLO is a mammalian protein that can bind to IAPs and antagonize their antiapoptotic effect, a function analogous to that of the proapoptotic Drosophila molecules, Grim, Reaper, and HID. Here, we show that after UV radiation, MIHA prevented apoptosis by inhibiting caspase 9 and caspase 3 activation. Unlike Bcl-2, MIHA functioned after release of cytochrome c and DIABLO from the mitochondria and was able to bind to both processed caspase 9 and processed caspase 3 to prevent feedback activation of their zymogen forms. Once released into the cytosol, DIABLO bound to MIHA and disrupted its association with processed caspase 9, thereby allowing caspase 9 to activate caspase 3, resulting in apoptosis.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Caspases/metabolism , Enzyme Inhibitors/pharmacology , Mitochondrial Proteins , Proteins/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Apoptosis Regulatory Proteins , Apoptotic Protease-Activating Factor 1 , Blotting, Western , Caspase 3 , Caspase 9 , Cell Line , Culture Media, Serum-Free , Enzyme Activation , Flow Cytometry , Humans , Intracellular Signaling Peptides and Proteins , Precipitin Tests , Protein Precursors/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transfection , Ultraviolet Rays , X-Linked Inhibitor of Apoptosis Protein
19.
J Biol Chem ; 275(35): 27084-93, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10825159

ABSTRACT

The caspase family of cysteine proteases plays important roles in bringing about apoptotic cell death. All caspases studied to date cleave substrates COOH-terminal to an aspartate. Here we show that the Drosophila caspase DRONC cleaves COOH-terminal to glutamate as well as aspartate. DRONC autoprocesses itself following a glutamate residue, but processes a second caspase, drICE, following an aspartate. DRONC prefers tetrapeptide substrates in which aliphatic amino acids are present at the P2 position, and the P1 residue can be either aspartate or glutamate. Expression of a dominant negative form of DRONC blocks cell death induced by the Drosophila cell death activators reaper, hid, and grim, and DRONC overexpression in flies promotes cell death. Furthermore, the Drosophila cell death inhibitor DIAP1 inhibits DRONC activity in yeast, and DIAP1's ability to inhibit DRONC-dependent yeast cell death is suppressed by HID and GRIM. These observations suggest that DRONC acts to promote cell death. However, DRONC activity is not suppressed by the caspase inhibitor and cell death suppressor baculovirus p35. We discuss possible models for DRONC function as a cell death inhibitor.


Subject(s)
Aspartic Acid/metabolism , Caspases/metabolism , Drosophila Proteins , Drosophila/enzymology , Glutamic Acid/metabolism , Insect Proteins/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Apoptosis , Caspases/chemistry , Hydrolysis , Inhibitor of Apoptosis Proteins , Molecular Sequence Data , Neuropeptides/metabolism , Sequence Homology, Amino Acid
20.
Cell ; 98(4): 453-63, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10481910

ABSTRACT

Drosophila Reaper (RPR), Head Involution Defective (HID), and GRIM induce caspase-dependent cell death and physically interact with the cell death inhibitor DIAP1. Here we show that HID blocks DIAP1's ability to inhibit caspase activity and provide evidence suggesting that RPR and GRIM can act similarly. Based on these results, we propose that RPR, HID, and GRIM promote apoptosis by disrupting productive IAP-caspase interactions and that DIAP1 is required to block apoptosis-inducing caspase activity. Supporting this hypothesis, we show that elimination of DIAP1 function results in global early embryonic cell death and a large increase in DIAP1-inhibitable caspase activity and that DIAP1 is still required for cell survival when expression of rpr, hid, and grim is eliminated.


Subject(s)
Apoptosis/physiology , Caspase Inhibitors , Drosophila Proteins , Drosophila melanogaster/physiology , Insect Proteins/physiology , Acridine Orange , Animals , Apoptosis/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Epistasis, Genetic , Fluorescent Dyes , Genes, Lethal , In Situ Nick-End Labeling , Inhibitor of Apoptosis Proteins , Insect Proteins/genetics , Morphogenesis/genetics , Neuropeptides/genetics , Neuropeptides/physiology , Peptides/genetics , Peptides/physiology , Recombinant Fusion Proteins/physiology , Saccharomyces cerevisiae/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...