Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
Brain Res ; 1648(Pt B): 603-616, 2016 10 01.
Article in English | MEDLINE | ID: mdl-26923166

ABSTRACT

In neurological disorders, both acute and chronic neural stress can disrupt cellular proteostasis, resulting in the generation of pathological protein. However in most cases, neurons adapt to these proteostatic perturbations by activating a range of cellular protective and repair responses, thus maintaining cell function. These interconnected adaptive mechanisms comprise a 'proteostasis network' and include the unfolded protein response, the ubiquitin proteasome system and autophagy. Interestingly, several recent studies have shown that these adaptive responses can be stimulated by preconditioning treatments, which confer resistance to a subsequent toxic challenge - the phenomenon known as hormesis. In this review we discuss the impact of adaptive stress responses stimulated in diverse human neuropathologies including Parkinson׳s disease, Wolfram syndrome, brain ischemia, and brain cancer. Further, we examine how these responses and the molecular pathways they recruit might be exploited for therapeutic gain. This article is part of a Special Issue entitled SI:ER stress.


Subject(s)
Autophagy , Nervous System Diseases , Proteostasis Deficiencies/complications , Unfolded Protein Response/physiology , Animals , Endoplasmic Reticulum Stress/physiology , Humans , Nervous System Diseases/complications , Nervous System Diseases/metabolism , Nervous System Diseases/therapy , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Ubiquitin/metabolism
2.
Cell Death Differ ; 20(5): 669-75, 2013 May.
Article in English | MEDLINE | ID: mdl-23449394

ABSTRACT

Programmed cell death is an important process during development that serves to remove superfluous cells and tissues, such as larval organs during metamorphosis, supernumerary cells during nervous system development, muscle patterning and cardiac morphogenesis. Different kinds of cell death have been observed and were originally classified based on distinct morphological features: (1) type I programmed cell death (PCD) or apoptosis is recognized by cell rounding, DNA fragmentation, externalization of phosphatidyl serine, caspase activation and the absence of inflammatory reaction, (2) type II PCD or autophagy is characterized by the presence of large vacuoles and the fact that cells can recover until very late in the process and (3) necrosis is associated with an uncontrolled release of the intracellular content after cell swelling and rupture of the membrane, which commonly induces an inflammatory response. In this review, we will focus exclusively on developmental cell death by apoptosis and its role in tissue remodeling.


Subject(s)
Apoptosis/physiology , Morphogenesis/physiology , Animals , Autophagy/physiology , Cell Movement , Drosophila/embryology , Humans , Mice/embryology , Necrosis , Signal Transduction
4.
Cell Death Differ ; 17(6): 912-21, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19960025

ABSTRACT

The mammalian p53 family consists of p53, p63 and p73. Whereas p53 accounts for tumor suppression through cell-cycle arrest and apoptosis, the functions of p63 and p73 are more diverse and also include control of cell differentiation. The Drosophila genome contains only one p53 homolog, Dp53. Previous work has established that Drosophila p53 (Dp53) induces apoptosis, but not cell-cycle arrest. In this study, using the developing eye as a model, we show that Dp53-induced apoptosis is primarily dependent on the pro-apoptotic gene, head involution defective (hid), but not reaper (rpr), and occurs through the canonical apoptosis pathway. Importantly, similar to p63 and p73, expression of Dp53 also inhibits cellular differentiation of photoreceptor neurons and cone cells in the eye independently of its apoptotic function. Intriguingly, expression of the human cell-cycle inhibitor p21 or its Drosophila homolog dacapo (dap) can suppress both Dp53-induced cell death and differentiation defects in Drosophila eyes. These findings provide new insights into the pathways activated by Dp53 and reveal that Dp53 incorporates functions of multiple p53 family members.


Subject(s)
Apoptosis , Cell Differentiation , Drosophila Proteins/physiology , Tumor Suppressor Protein p53/physiology , Animals , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Drosophila/cytology , Drosophila/metabolism , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , Eye/cytology , Eye/metabolism , Humans , Neuropeptides/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
5.
Cell Death Differ ; 15(7): 1132-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18437164

ABSTRACT

Insects have made major contributions to understanding the regulation of cell death, dating back to the pioneering work of Lockshin and Williams on death of muscle cells during postembryonic development of Manduca. A physically smaller cousin of moths, the fruit fly Drosophila melanogaster, offers unique advantages for studying the regulation of cell death in response to different apoptotic stimuli in situ. Different signaling pathways converge in Drosophila to activate a common death program through transcriptional activation of reaper, hid and grim. Reaper-family proteins induce apoptosis by binding to and antagonizing inhibitor of apoptosis proteins (IAPs), which in turn inhibit caspases. This switch from life to death relies extensively on targeted degradation of cell death proteins by the ubiquitin-proteasome pathway. Drosophila IAP-1 (Diap1) functions as an E3-ubiquitin ligase to protect cells from unwanted death by promoting the degradation of the initiator caspase Dronc. However, in response to apoptotic signals, Reaper-family proteins are produced, which promote the auto-ubiquitination and degradation of Diap1, thereby removing the 'brakes on death' in cells that are doomed to die. More recently, several other ubiquitin pathway proteins were found to play important roles for caspase regulation, indicating that the control of cell survival and death relies extensively on targeted degradation by the ubiquitin-proteasome pathway.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Signal Transduction , Animals , Caspases/metabolism , Drosophila melanogaster/enzymology , Inhibitor of Apoptosis Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Regeneration , Ubiquitin/metabolism
6.
Cell Death Differ ; 14(8): 1508-17, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17464325

ABSTRACT

Programmed cell death (PCD) in the Drosophila ovary occurs either during mid-oogenesis, resulting in degeneration of the entire egg chamber or during late oogenesis, to facilitate the development of the oocyte. PCD during oogenesis is regulated by mechanisms different from those that control cell death in other Drosophila tissues. We have analyzed the role of caspases in PCD of the female germline by examining caspase mutants and overexpressing caspase inhibitors. Imprecise P-element excision was used to generate mutants of the initiator caspase strica. While null mutants of strica or another initiator caspase, dronc, display no ovary phenotype, we find that strica exhibits redundancy with dronc, during both mid- and late oogenesis. Ovaries of double mutants contain defective mid-stage egg chambers similar to those reported previously in dcp-1 mutants, and mature egg chambers with persisting nurse cell nuclei. In addition, the effector caspases drice and dcp-1 also display redundant functions during late oogenesis, resulting in persisting nurse cell nuclei. These findings indicate that caspases are required for nurse cell death during mid-oogenesis, and participate in developmental nurse cell death during late oogenesis. This reveals a novel pathway of cell death in the ovary that utilizes strica, dronc, dcp-1 and drice, and importantly illustrates strong redundancy among the caspases.


Subject(s)
Apoptosis/physiology , Caspases/physiology , Drosophila Proteins/physiology , Drosophila/cytology , Drosophila/enzymology , Oogenesis/physiology , Animals , Animals, Genetically Modified , Apoptosis/genetics , Base Sequence , Caspases/genetics , DNA, Complementary/genetics , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/genetics , Female , Gene Expression Regulation, Developmental , Genes, Insect , Mutation , Oogenesis/genetics
7.
Mol Cell ; 5(3): 445-55, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10882130

ABSTRACT

The steroid hormone ecdysone signals the stage-specific programmed cell death of the larval salivary glands during Drosophila metamorphosis. This response is preceded by an ecdysone-triggered switch in gene expression in which the diap2 death inhibitor is repressed and the reaper (rpr) and head involution defective (hid) death activators are induced. Here we show that rpr is induced directly by the ecdysone-receptor complex through an essential response element in the rpr promoter. The Broad-Complex (BR-C) is required for both rpr and hid transcription, while E74A is required for maximal levels of hid induction. diap2 induction is dependent on betaFTZ-F1, while E75A and E75B are each sufficient to repress diap2. This study identifies transcriptional regulators of programmed cell death in Drosophila and provides a direct link between a steroid signal and a programmed cell death response.


Subject(s)
Apoptosis/genetics , Drosophila Proteins , Drosophila/embryology , Drosophila/genetics , Ecdysone/pharmacology , Salivary Glands/cytology , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/metabolism , Dimerization , Fushi Tarazu Transcription Factors , Gene Expression Regulation, Developmental , Homeodomain Proteins , Inhibitor of Apoptosis Proteins , Insect Proteins/genetics , Insect Proteins/metabolism , Metamorphosis, Biological , Neuropeptides/genetics , Peptides/genetics , Receptors, Cytoplasmic and Nuclear , Receptors, Steroid/metabolism , Response Elements , Signal Transduction , Steroidogenic Factor 1 , Transcription, Genetic
8.
Nat Cell Biol ; 2(6): E100-2, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10854336

ABSTRACT

The recent discovery of a Drosophila orthologue of the p53 tumour suppressor promises new insights into the complex function, regulation and evolution of one of the most intensely studied human disease proteins.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Insect Proteins/metabolism , Neuropeptides/metabolism , Peptides/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/radiation effects , Cell Cycle/radiation effects , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/radiation effects , Humans , Insect Proteins/genetics , Neuropeptides/genetics , Peptides/genetics , Tumor Suppressor Protein p53/genetics
9.
J Biol Chem ; 275(29): 22157-65, 2000 Jul 21.
Article in English | MEDLINE | ID: mdl-10764741

ABSTRACT

Deterin, a new apoptosis inhibitor from Drosophila melanogaster, possesses an unusual structure of only a single baculovirus inhibitor of apoptosis (IAP)-type repeat and no RING finger motif. The biochemical actions of deterin are demonstrated in SF9 and S2 cell transfection assays, in which the expressed protein acts in the cytoplasm to inhibit or deter cells from apoptosis otherwise induced by the caspase-dependent apoptosis activator reaper or by cytotoxicants. A loss of function phenotype for deterin of cell death was indicated by transfections with either a dominant negative deterin mutant or with inhibitory RNA (RNAi) for deterin. The dominant negative C-terminal fragment that antagonized antiapoptotic activity of deterin did not affect antiapoptotic activity of DIAP1 or p35. Both the baculovirus IAP-type repeat (BIR) domain and the alpha-helical C-terminal domain are necessary in both SF9 and S2 cells for deterin to manifest its activity to prevent cell death. The approximately 650-base deterin transcript is present in embryos, third instar larvae, and late stage nurse cells of adult females. The deterin transcript is distributed throughout early stage embryos, whereas in later stage embryos it becomes progressively restricted to the central nervous system and gonads. Whereas the nematode survivin-type IAP has thus far been implicated only as a mitotic regulator, Drosophila deterin constitutes the first invertebrate member of the survivin-type IAP group to exhibit apoptosis-inhibitory activity.


Subject(s)
Apoptosis/physiology , Drosophila Proteins , Drosophila melanogaster , Insect Proteins/physiology , Amino Acid Sequence , Animals , DNA, Complementary/analysis , DNA, Complementary/genetics , Gene Expression Regulation , Molecular Sequence Data , Sequence Alignment , Survivin
10.
Mol Cell Biol ; 20(8): 2907-14, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10733594

ABSTRACT

In Drosophila melanogaster, the induction of apoptosis requires three closely linked genes, reaper (rpr), head involution defective (hid), and grim. The products of these genes induce apoptosis by activating a caspase pathway. Two very similar Drosophila caspases, DCP-1 and drICE, have been previously identified. We now show that DCP-1 has a substrate specificity that is remarkably similar to those of human caspase 3 and Caenorhabditis elegans CED-3, suggesting that DCP-1 is a death effector caspase. drICE and DCP-1 have similar yet different enzymatic specificities. Although expression of either in cultured cells induces apoptosis, neither protein was able to induce DNA fragmentation in Drosophila SL2 cells. Ectopic expression of a truncated form of dcp-1 (DeltaN-dcp-1) in the developing Drosophila retina under an eye-specific promoter resulted in a small and rough eye phenotype, whereas expression of the full-length dcp-1 (fl-dcp-1) had little effect. On the other hand, expression of either full-length drICE (fl-drICE) or truncated drICE (DeltaN-drICE) in the retina showed no obvious eye phenotype. Although active DCP-1 protein cleaves full-length DCP-1 and full-length drICE in vitro, GMR-DeltaN-dcp-1 did not enhance the eye phenotype of GMR-fl-dcp-1 or GMR-fl-drICE flies. Significantly, GMR-rpr and GMR-grim, but not GMR-hid, dramatically enhanced the eye phenotype of GMR-fl-dcp-1 flies. These results indicate that Reaper and Grim, but not HID, can activate DCP-1 in vivo.


Subject(s)
Apoptosis/genetics , Caspases/genetics , Drosophila Proteins , Drosophila melanogaster/genetics , Neuropeptides/genetics , Peptides/genetics , Animals , Gene Expression Regulation , Genes, Insect , Humans
11.
EMBO J ; 19(4): 589-97, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10675328

ABSTRACT

Induction of apoptosis in Drosophila requires the activity of three closely linked genes, reaper, hid and grim. Here we show that the proteins encoded by reaper, hid and grim activate cell death by inhibiting the anti-apoptotic activity of the Drosophila IAP1 (diap1) protein. In a genetic modifier screen, both loss-of-function and gain-of-function alleles in the endogenous diap1 gene were obtained, and the mutant proteins were functionally and biochemically characterized. Gain-of-function mutations in diap1 strongly suppressed reaper-, hid- and grim-induced apoptosis. Sequence analysis of these alleles revealed that they were caused by single amino acid changes in the baculovirus IAP repeat domains of diap1, a domain implicated in binding REAPER, HID and GRIM. Significantly, the corresponding mutant DIAP1 proteins displayed greatly reduced binding of REAPER, HID and GRIM, indicating that REAPER, HID and GRIM kill by forming a complex with DIAP1. These data provide strong in vivo evidence for a previously published model of cell death regulation in Drosophila.


Subject(s)
Apoptosis/genetics , Apoptosis/physiology , Drosophila Proteins , Drosophila/cytology , Drosophila/genetics , Genes, Insect , Insect Proteins/antagonists & inhibitors , Insect Proteins/physiology , Alleles , Animals , Drosophila/metabolism , Eye/anatomy & histology , Inhibitor of Apoptosis Proteins , Insect Proteins/genetics , Microscopy, Electron, Scanning , Models, Biological , Mutation , Neuropeptides/genetics , Peptides/genetics , Phenotype
12.
Nat Cell Biol ; 2(12): 915-21, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11146656

ABSTRACT

Here we describe a protein product of the human septin H5/PNUTL2/CDCrel2b gene, which we call ARTS (for apoptosis-related protein in the TGF-beta signalling pathway). ARTS is expressed in many cells and acts to enhance cell death induced by TGF-beta or, to a lesser extent, by other apoptotic agents. Unlike related septin gene products, ARTS is localized to mitochondria and translocates to the nucleus when apoptosis occurs. Mutation of the P-loop of ARTS abrogates its competence to activate caspase 3 and to induce apoptosis. Taken together, these observations expand the functional attributes of septins previously described as having roles in cytokinesis and cellular morphogenesis.


Subject(s)
Apoptosis/physiology , Cytoskeletal Proteins/physiology , GTP Phosphohydrolases/physiology , Amino Acid Motifs , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Caspases/metabolism , Cloning, Molecular , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , DNA Primers/genetics , Enzyme Activation , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/genetics , Gene Expression , Humans , Mitochondria/metabolism , Molecular Sequence Data , Septins , Sequence Homology, Amino Acid , Signal Transduction , Transfection , Transforming Growth Factor beta/physiology
13.
Genome Biol ; 1(3): REVIEWS0003, 2000.
Article in English | MEDLINE | ID: mdl-11178240

ABSTRACT

Key components of the programmed cell death pathway are conserved between Caenorhabditis elegans, Drosophila melanogaster and humans. The search for additional homologs has been facilitated by the availability of the entire genomic sequence for each of these organisms.


Subject(s)
Apoptosis/physiology , Caenorhabditis elegans/physiology , Drosophila melanogaster/physiology , Signal Transduction , Animals , Apoptosis/genetics , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Conserved Sequence , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Genomics , Humans , Species Specificity
14.
Trends Cell Biol ; 9(12): M49-52, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10611682

ABSTRACT

Active cellular suicide by apoptosis plays important roles in animal development, tissue homeostasis and a wide variety of diseases, including cancer, AIDS, stroke and many neurodegenerative disorders. A central step in the execution of apoptosis is the activation of an unusual class of cysteine proteases, termed caspases, that are widely expressed as inactive zymogens. Originally, the mechanisms for regulating the caspase-based cell death programme seemed to be different in Caenorhabditis elegans, mammals and insects. However, recent results suggest that these apparent differences in the control of cell death reflect our incomplete knowledge, rather than genuine mechanistic differences between different organisms.


Subject(s)
Apoptosis , Drosophila Proteins , Animals , Caspases/metabolism , Drosophila , Enzyme Activation , Humans , Inhibitor of Apoptosis Proteins , Neuropeptides/metabolism , Neuropeptides/physiology , Peptides/metabolism , Peptides/physiology , Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Cell Surface/metabolism
15.
J Cell Biol ; 147(3): 507-18, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10545496

ABSTRACT

KLP64D and KLP68D are members of the kinesin-II family of proteins in Drosophila. Immunostaining for KLP68D and ribonucleic acid in situ hybridization for KLP64D demonstrated their preferential expression in cholinergic neurons. KLP68D was also found to accumulate in cholinergic neurons in axonal obstructions caused by the loss of kinesin light chain. Mutations in the KLP64D gene cause uncoordinated sluggish movement and death, and reduce transport of choline acetyltransferase from cell bodies to the synapse. The inviability of KLP64D mutations can be rescued by expression of mammalian KIF3A. Together, these data suggest that kinesin-II is required for the axonal transport of a soluble enzyme, choline acetyltransferase, in a specific subset of neurons in Drosophila. Furthermore, the data lead to the conclusion that the cargo transport requirements of different classes of neurons may lead to upregulation of specific pathways of axonal transport.


Subject(s)
Axonal Transport , Calcium-Binding Proteins/metabolism , Choline O-Acetyltransferase/metabolism , Drosophila Proteins , Drosophila melanogaster/metabolism , Muscle Proteins/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Cloning, Molecular , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Drosophila melanogaster/enzymology , Genes, Lethal/genetics , Genetic Complementation Test , Kinesins/chemistry , Kinesins/genetics , Kinesins/metabolism , Larva/cytology , Larva/enzymology , Larva/metabolism , Mice , Molecular Sequence Data , Movement , Muscle Proteins/chemistry , Muscle Proteins/genetics , Mutation/genetics , Nervous System/cytology , Nervous System/embryology , Neurons/cytology , Neurons/enzymology , Neurons/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Sequence Homology, Amino Acid
16.
Dev Biol ; 214(2): 385-98, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10525342

ABSTRACT

The Drosophila disconnected (disco) gene is required for the formation of appropriate connections between the larval optic nerve and its target cells in the brain. The disco gene encodes a nuclear protein with two zinc fingers, which suggests that the gene product is a transcription factor. Here, we present data supporting this notion. We find that disco expression in the optic lobe primordium, a group of cells contacted by the developing optic nerve, depends on an autoregulatory feedback loop. We show that wild-type disco function is required for maintenance of disco mRNA and protein expression in the developing optic lobe. In addition, we demonstrate that ubiquitous Disco activity supplied by a heat-inducible gene construct activates expression from the endogenous disco gene specifically in the optic lobe primordium. Consistent with a role of Disco as a transcriptional regulatory protein, we show that portions of the Disco protein are capable of activating the transcription of reporter constructs in a heterologous system. Moreover, we find that the zinc finger portion of Disco binds in vitro to sequences located near the disco transcription unit, suggesting that Disco autoregulates its transcription in the optic lobe primordium by direct binding to a regulatory element in its own promoter.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins , Drosophila/genetics , Gene Expression Regulation, Developmental , Homeostasis , Photoreceptor Cells, Invertebrate/embryology , Transcription Factors/genetics , Animals , Bacterial Proteins/metabolism , Genes, Insect , Genes, Reporter , In Situ Hybridization , Larva/growth & development , Models, Genetic , Optic Lobe, Nonmammalian/embryology , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Serine Endopeptidases/metabolism , Temperature , Transcription, Genetic
17.
Proc Natl Acad Sci U S A ; 96(9): 4936-41, 1999 Apr 27.
Article in English | MEDLINE | ID: mdl-10220397

ABSTRACT

Three genes-reaper, grim, and hid-are crucial to the regulation of programmed cell death in Drosophila melanogaster. Mutations involving all three genes virtually abolish apoptosis during development, and homozygous hid mutants die as embryos with extensive defects in apoptosis. Although Hid is central to apoptosis in Drosophila, it has no mammalian homologue identified to date. We present evidence that expression of Drosophila Hid in mammalian cells induces apoptosis. This activity is subject to regulation by inhibitors of mammalian cell death. We show that the N terminus of Hid, which is a region of homology with Reaper and Grim, is essential for Hid's function in mammalian cells. We demonstrate that Hid is localized to the mitochondria via a hydrophobic region at its C terminus and functionally interacts with BclXL. This study shows that the function of Hid as a death inducer in Drosophila is conserved in mammalian cells and argues for the existence of a mammalian homologue of this critical regulator of apoptosis.


Subject(s)
Apoptosis/genetics , Conserved Sequence , Drosophila Proteins , Drosophila melanogaster/genetics , Neuropeptides/genetics , Peptides/genetics , Animals , Gene Expression Regulation , Genes, Insect , Humans , Transfection
18.
Biotechnol Bioeng ; 62(6): 632-40, 1999 Mar 20.
Article in English | MEDLINE | ID: mdl-9951521

ABSTRACT

One of the main problems in the culture of Chinese Hamster Ovary (CHO) cells continues to be the inability to maintain the viability of the cultures over an extended period of time. The rapid decline in viability at the end of the culture is exacerbated by the absence of serum. In trying to reduce the extent of death in these cultures, we first tried to determine the mode of death. We found that more than 80% of the cells in a standard serum-free batch culture of CHO cells in suspension died via apoptosis--as evidenced by condensed chromatin and the appearance of a characteristic DNA ladder. Furthermore, when protein synthesis was inhibited using cycloheximide, the cells underwent rapid apoptosis indicating that death proteins were present in greater abundance than survival proteins in our CHO cells. Cell lysate from CHO cells showed evidence of cysteine protease (caspase) activity. Caspases of the Interleukin-1-beta-Converting Enzyme (ICE) family, e.g., CPP32, Mch-1, etc., have been implicated in the apoptotic process. Surprisingly, a caspase peptide inhibitor, N-benzyloxycarbonyl-Val-Ala-Asp-fluoro-methyl-ketone (z-VAD.fmk), was unable to substantially extend the life of a serum-free batch culture of CHO cells. In addition, z-VAD.fmk was only marginally able to extend viability in response to withdrawal of growth and survival factors, insulin and transferrin. In both these instances, z-VAD.fmk was able to prevent cleavage of caspase substrates, but not protect cells from death. However, we found that bcl-2 expression was able to significantly extend viabilities in CHO batch culture. Bcl-2 expression also substantially extended the viability of cultures in response to insulin and transferrin withdrawal. These results provide interesting insights into the pathways of death in a CHO cell.


Subject(s)
Apoptosis/drug effects , Apoptosis/genetics , CHO Cells/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Animals , CHO Cells/cytology , CHO Cells/pathology , Caspase 3 , Caspases/analysis , Caspases/drug effects , Caspases/metabolism , Cell Division/drug effects , Cell Division/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Cricetinae , Culture Media, Serum-Free , Cycloheximide/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Humans , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Transfection
19.
Mol Cell ; 4(5): 745-55, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10619022

ABSTRACT

We have identified a Drosophila homolog of Apaf-1 and ced-4, termed hac-1. Like mammalian APAF-1, HAC-1 can activate caspases in a dATP-dependent manner in vitro. During embryonic development, hac-1 is prominently expressed in regions where cells undergo natural death. Significantly, hac-1 transcription is also rapidly induced upon ionizing irradiation, similar to the proapoptotic gene reaper. Loss of hac-1 function causes reduced cell death, and reducing the dosage of hac-1 suppresses ectopic cell killing upon expression of the dcp-1 procaspase in the retina but has little effect on reaper, hid, and grim-mediated killing. Our data indicate that caspase activation and apoptosis in Drosophila are independently controlled by at least two distinct regulatory pathways that converge at the level of caspase activation.


Subject(s)
Apoptosis , Caenorhabditis elegans Proteins , Calcium-Binding Proteins/chemistry , Drosophila Proteins , Drosophila melanogaster/embryology , Helminth Proteins/chemistry , Insect Proteins/chemistry , Insect Proteins/metabolism , Proteins/chemistry , Amino Acid Sequence , Animals , Apoptosis/radiation effects , Apoptotic Protease-Activating Factor 1 , Base Sequence , Caspases/metabolism , Cell Line , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Drosophila melanogaster/radiation effects , Enzyme Activation , Enzyme Precursors/metabolism , Eye/embryology , Eye/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Insect Proteins/genetics , Molecular Sequence Data , Neurons/cytology , Neurons/metabolism , Neuropeptides/genetics , Neuropeptides/physiology , Peptides/genetics , Peptides/physiology , Proteins/genetics , Proteins/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Sequence Homology, Amino Acid , Transcriptional Activation/radiation effects
20.
Cell ; 95(3): 331-41, 1998 Oct 30.
Article in English | MEDLINE | ID: mdl-9814704

ABSTRACT

Extracellular growth factors are required for the survival of most animal cells. They often signal through the activation of the Ras pathway. However, the molecular mechanisms by which Ras signaling inhibits the intrinsic cell death machinery are not well understood. Here, we present evidence that in Drosophila, activation of the Ras pathway specifically inhibits the proapoptotic activity of the gene head involution defective (hid). By using transgenic animals and cultured cells, we show that MAPK phosphorylation sites in Hid are critical for this response. These findings define a novel mechanism by which growth factor signaling directly inactivates a critical component of the intrinsic cell death machinery. These studies provide further insights into the function of ras as an oncogene.


Subject(s)
Apoptosis , Cell Survival , Drosophila Proteins , Extracellular Signal-Regulated MAP Kinases , Neuropeptides/genetics , Signal Transduction , ras Proteins/metabolism , Animals , Animals, Genetically Modified , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Eye/cytology , Genes, Lethal , Genes, Suppressor , Insect Proteins/genetics , Insect Proteins/metabolism , Mutation , Neuropeptides/antagonists & inhibitors , Neuropeptides/metabolism , Peptides/genetics , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Transfection , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...