Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Cell Death Dis ; 6: e1816, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26181197

ABSTRACT

The Sall2 transcription factor is deregulated in several cancers; however, little is known about its cellular functions, including its target genes. Recently, we demonstrated that p53 directly regulates Sall2 expression under genotoxic stress. Here, we investigated the role of Sall2 in the context of cellular response to genotoxic stress. In addition, we further examined the Sall2-p53 relationship during genotoxic stress in primary mouse embryo fibroblasts (MEFs), which are derived from Sall2 knockout mice separately, or in combination with the p53ERTAM knock-in mice. We found that the levels of Sall2 mRNA and protein are dynamically modulated in response to doxorubicin. At early times of stress, Sall2 is downregulated, but increases under extension of the stress in a p53-independent manner. Based on caspase-3/7 activities, expression of cleaved poly (ADP-ribose) polymerase, expression of cleaved caspase-3 and induction of proapoptotic proteins, Sall2 expression was correlated with cellular apoptosis. Consequently, Sall2-/- MEFs have decreased apoptosis, which relates with increased cell viability in response to doxorubicin. Importantly, Sall2 was required for apoptosis even in the presence of fully activated p53. Searching for putative Sall2 targets that could mediate its role in apoptosis, we identified proapoptotic NOXA/PMAIP1 (phorbol-12-myristate-13-acetate-induced protein 1). We demonstrated that Sall2 positively regulates Noxa promoter activity. Conserved putative Sall2-binding sites at the NOXA promoter were validated in vitro by electrophoretic mobility shift assay and in vivo by ChIP experiments, identifying NOXA as a novel Sall2 target. In agreement, induction of Noxa protein and mRNA in response to doxorubicin was significantly decreased in Sall2-/- MEFs. In addition, studies in leukemia Jurkat T cells support the existence of the Sall2/Noxa axis, and the significance of this axis on the apoptotic response to doxorubicin in cancer cells. Our study highlights the relevance of Sall2 in the apoptotic response to extended genotoxic stress, which is important for understanding its role in normal physiology and disease.


Subject(s)
DNA Damage , Intracellular Signaling Peptides and Proteins/genetics , Leukemia/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis/drug effects , DNA-Binding Proteins , Doxorubicin/administration & dosage , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Leukemia/pathology , Mice , Mice, Knockout , Protein Binding , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription Factors , Tumor Suppressor Protein p53/metabolism
2.
Oncogene ; 33(40): 4877-80, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-24662815

ABSTRACT

Reversibly switchable proteins are powerful tools with which to explore protein function in vitro and in vivo. For example, the activity of many proteins fused to the hormone-binding domain of the modified oestrogen receptor (ER(TAM)) can be regulated by provision or removal of 4-hydroxytamoxifen (4-OHT). Despite the widespread use of ER(TAM) fusions in vivo, inadequate data are available as to the most efficacious routes for systemic tamoxifen delivery. In this study, we have used two well-characterized ER(TAM) fusion proteins, both reversibly activated by 4-OHT, to compare the effectiveness and kinetics of 4-OHT delivery in mice in vivo by either tamoxifen in food or by intraperitoneal injection. Our data indicate that dietary tamoxifen offers an effective, facile and ethically preferable means for long-term activation of ER(TAM) fusion proteins in vivo.


Subject(s)
Antineoplastic Agents/administration & dosage , Receptors, Estrogen/genetics , Tamoxifen/analogs & derivatives , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Genes, Reporter , Injections, Intraperitoneal , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Kinetics , Mice , Rats , Receptors, Estrogen/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Tamoxifen/administration & dosage , Tamoxifen/pharmacology , Transcriptional Activation/drug effects , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
3.
Cell Death Dis ; 4: e468, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23348582

ABSTRACT

Mitotic cell death following prolonged arrest is an important death mechanism that is not completely understood. This study shows that Protein Tyrosine Phosphatase 1B (PTP1B) undergoes phosphorylation during mitotic arrest induced by microtubule-targeting agents (MTAs) in chronic myeloid leukaemia cells. Inhibition of cyclin-dependent kinase 1 (Cdk1) or polo-like kinase 1 (Plk1) during mitosis prevents PTP1B phosphorylation, implicating these kinases in PTP1B phosphorylation. In support of this, Cdk1 and Plk1 co-immunoprecipitate with endogenous PTP1B from mitotic cells. In addition, active recombinant Cdk1-cyclin B1 directly phosphorylates PTP1B at serine 386 in a kinase assay. Recombinant Plk1 phosphorylates PTP1B on serine 286 and 393 in vitro, however, it requires a priming phosphorylation by Cdk1 at serine 386 highlighting a novel co-operation between Cdk1 and Plk1 in the regulation of PTP1B. Furthermore, overexpression of wild-type PTP1B induced mitotic cell death, which is potentiated by MTAs. Moreover, mutation of serine 286 abrogates the cell death induced by PTP1B, whereas mutation of serine 393 does not, highlighting the importance of serine 286 phosphorylation in the execution of mitotic cell death. Finally, phosphorylation on serine 286 enhanced PTP1B phosphatase activity. Collectively, these data reveal that PTP1B activity promotes mitotic cell death and is regulated by the co-operative action of Cdk1 and Plk1 during mitotic arrest.


Subject(s)
Apoptosis/drug effects , CDC2 Protein Kinase/pharmacology , Cell Cycle Proteins/pharmacology , Protein Serine-Threonine Kinases/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Proto-Oncogene Proteins/pharmacology , Antineoplastic Agents/toxicity , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cyclin B1/genetics , Cyclin B1/metabolism , Cyclin B1/pharmacology , Humans , Immunoprecipitation , K562 Cells , Mitosis , Nocodazole/toxicity , Paclitaxel/toxicity , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Serine/chemistry , Polo-Like Kinase 1
4.
Nature ; 443(7108): 214-7, 2006 Sep 14.
Article in English | MEDLINE | ID: mdl-16957739

ABSTRACT

The p53 protein has a highly evolutionarily conserved role in metazoans as 'guardian of the genome', mediating cell-cycle arrest and apoptosis in response to genotoxic injury. In large, long-lived animals with substantial somatic regenerative capacity, such as vertebrates, p53 is an important tumour suppressor--an attribute thought to stem directly from its induction of death or arrest in mutant cells with damaged or unstable genomes. Chemotherapy and radiation exposure both induce widespread p53-dependent DNA damage. This triggers potentially lethal pathologies that are generally deemed an unfortunate but unavoidable consequence of the role p53 has in tumour suppression. Here we show, using a mouse model in which p53 status can be reversibly switched in vivo between functional and inactive states, that the p53-mediated pathological response to whole-body irradiation, a prototypical genotoxic carcinogen, is irrelevant for suppression of radiation-induced lymphoma. In contrast, delaying the restoration of p53 function until the acute radiation response has subsided abrogates all of the radiation-induced pathology yet preserves much of the protection from lymphoma. Such protection is absolutely dependent on p19(ARF)--a tumour suppressor induced not by DNA damage, but by oncogenic disruption of the cell cycle.


Subject(s)
DNA Damage , Lymphoma/metabolism , Lymphoma/pathology , Tumor Suppressor Protein p53/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p16 , DNA Damage/radiation effects , Lymphoma/genetics , Mice , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/metabolism , Neoplasms, Radiation-Induced/pathology , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/genetics
5.
Article in English | MEDLINE | ID: mdl-16869762

ABSTRACT

Cancers arise by an evolutionary process that involves the protracted acquisition by somatic cells of suites of interlocking mutations that uncouple proliferation, survival, migration, and damage responses from the mechanisms (selective pressures) that normally restrain or restrict them in time and space. The relative rareness of cancer cells within the soma, in the face of huge numbers of available cell targets, substantial rates of mutation, and an abundance of proto-oncogenes and tumor suppressor gene targets, indicates that the evolutionary space available to incipient tumor cells is highly restricted. The principal way in which this is achieved is through intrinsic tumor suppression pathways-innate growth arrest and apoptotic programs that fulfill an essentially analogous functional role to checkpoints in the cell cycle machinery by antagonizing the tumorigenic potential of oncogenic mutations. Using switchable transgenic and knockin mouse models, it is possible to identify these various tumor suppressor programs and establish where, when, how, and why they act to forestall neoplasia in each tissue type and, consequently, how and why their failure leads to cancer.


Subject(s)
Neoplasms/genetics , Neoplasms/therapy , Oncogenes , Animals , Cocarcinogenesis , Gene Expression Profiling , Genes, myc , Genes, p53 , Humans , Mice , Mice, Knockout , Models, Biological , Neoplasms/etiology , Neoplasms/pathology , Proto-Oncogene Proteins c-myc/physiology , Tumor Suppressor Protein p53/physiology
6.
Cell Death Differ ; 11(9): 1038-45, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15143346

ABSTRACT

Obligate sensitization to apoptosis provides a safeguard mechanism against the oncogenic potential of Myc. Omomyc is a mutant bHLHZip domain that sequesters Myc in complexes that are unable to bind to the E box recognition element and activate transcription but remain competent for transcriptional repression. Omomyc has the peculiar properties of reverting Myc-induced transformation of tissue culture cells and enhancing Myc proapoptotic function. Thus, Omomyc has the potential to act as a potent suppressor of Myc-induced oncogenesis. To validate the therapeutic potential of Omomyc in vivo, we targeted its expression to the adult suprabasal epidermis of Inv-c-MycER (TAM) transgenic mice which express a switchable form of the Myc protein in suprabasal cells. Activation of Myc induces rapid epidermal hyperplasia and papillomatosis. We show that Omomyc inhibits such Myc-induced papillomatosis, potentiating Myc-dependent apoptosis in a tissue in which it is usually strongly suppressed. Furthermore, Omomyc expression restores the normal keratinocyte differentiation program and skin architecture, both of which are otherwise disrupted by Myc activation. These findings indicate that it is possible to selectively enhance the intrinsic apoptotic pathway mediated by Myc and so quell its oncogenic action.


Subject(s)
Papilloma/metabolism , Papilloma/prevention & control , Proto-Oncogene Proteins c-myc/physiology , Skin Neoplasms/metabolism , Skin Neoplasms/prevention & control , Animals , Apoptosis , Cell Line , Cell Transformation, Neoplastic , Cells, Cultured , Epidermis/metabolism , Flow Cytometry , Genetic Vectors , Humans , Hydroxytestosterones/pharmacology , Immunohistochemistry , Keratinocytes/metabolism , Mice , Mice, Inbred DBA , Mice, Transgenic , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Skin/pathology , Time Factors , Transgenes , Tumor Suppressor Protein p53/metabolism , bcl-X Protein
7.
J Biol Chem ; 276(42): 38417-25, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11479289

ABSTRACT

This study was designed to identify the role of a recently identified Ca(2+)/calmodulin-dependent protein kinase (CaMK)-like kinase (CaMKLK) in neuronal apoptosis. For this purpose, we studied proteolytic cleavage of CaMKLK by caspases in vitro and in neuronal NG108 cells. In addition, we have investigated the effect of overexpression of wild type and mutant CaMKLK proteins on staurosporine- and serum deprivation-induced apoptosis of NG108 cells. We found that CaMKLK is a substrate for caspase-3 and -8, both in vitro and in NG108 cells during staurosporine- and serum withdrawal-induced apoptosis. Substitution of an aspartic acid residue at position 62 in an asparagine residue within a putative caspase cleavage site completely blocked cleavage of CaMKLK, strongly indicating that (59)DEND(62) is the caspase recognition site. Overexpression of an Asp(62) --> Asn CaMKLK mutant protected NG108 cells from staurosporine-induced apoptosis to a similar extent as Bcl-x(L). In contrast, overexpression of wild type CaMKLK did not lead to protection. Moreover, microinjection of Asp(62) --> Asn CaMKLK protected NG108 cells from serum deprivation-induced apoptosis, while overexpression of a caspase-generated noncatalytic N-terminal CaMKLK fragment exacerbated apoptosis. Together, our data suggest that cleavage of CaMKLK and generation of the noncatalytic N-terminal domain of CaMKLK facilitate neuronal apoptosis.


Subject(s)
Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calcium/metabolism , Calmodulin/metabolism , Caspases/metabolism , Neurons/metabolism , Animals , Asparagine/chemistry , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Binding Sites , Calcium-Calmodulin-Dependent Protein Kinase Type 1 , Caspase 3 , Caspase 8 , Caspase 9 , Cell Line , Cells, Cultured , Culture Media, Serum-Free/metabolism , DNA/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Immunohistochemistry , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Protein Synthesis Inhibitors/pharmacology , Rabbits , Reticulocytes/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Staurosporine/pharmacology , Subcellular Fractions , Tumor Cells, Cultured
8.
Nature ; 411(6835): 342-8, 2001 May 17.
Article in English | MEDLINE | ID: mdl-11357141

ABSTRACT

Beneath the complexity and idiopathy of every cancer lies a limited number of 'mission critical' events that have propelled the tumour cell and its progeny into uncontrolled expansion and invasion. One of these is deregulated cell proliferation, which, together with the obligate compensatory suppression of apoptosis needed to support it, provides a minimal 'platform' necessary to support further neoplastic progression. Adroit targeting of these critical events should have potent and specific therapeutic consequences.


Subject(s)
Apoptosis , Cell Cycle , Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Clone Cells/pathology , Disease Progression , Genetic Variation/genetics , Humans , Neoplasms/drug therapy , Neoplasms/genetics
9.
Oncogene ; 20(10): 1164-75, 2001 Mar 08.
Article in English | MEDLINE | ID: mdl-11313860

ABSTRACT

To investigate Mad1 function in vivo, transgenic mice were generated that express a Mad1 transgene in T lineage cells under the control of the proximal lck promoter. Thymus size in lck-Mad1 transgenic mice is drastically reduced although representation of the various thymocyte sub populations appears normal. To investigate more closely any effects of Mad1 expression on thymocytes, we examined thymic selection using MHC class I-restricted H-Y-TCR transgenic mice. Mad1 expression in vivo reduces the efficiency of positive selection. Furthermore, thymocytes and splenic T cells from lck-Mad1 transgenic mice display a profound proliferative defect in response to activation with either PMA/Ionomycin or immobilized anti-CD3/CD28 antibody. This proliferative defect is not reversed by addition of exogenous IL-2 and is p53-independent. The growth inhibition caused by Mad1 is overcome by expression of active c-Myc.


Subject(s)
Lymphocyte Activation/drug effects , Mitogens/pharmacology , Phosphoproteins/biosynthesis , Repressor Proteins/biosynthesis , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , Antigens, CD/physiology , Blotting, Western , Cell Cycle Proteins , DNA Primers/chemistry , Female , Genes, myc/physiology , H-Y Antigen/immunology , Histocompatibility Antigens Class I/immunology , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Nuclear Proteins , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Resting Phase, Cell Cycle/drug effects , Reverse Transcriptase Polymerase Chain Reaction , S Phase/drug effects , Spleen/immunology , Thymus Gland/pathology , Tumor Suppressor Protein p53/metabolism
10.
Oncogene ; 19(15): 1891-900, 2000 Apr 06.
Article in English | MEDLINE | ID: mdl-10773879

ABSTRACT

In order to study the effect of c-Myc activation in T lymphocytes in vivo, we generated transgenic mice that express a 4-hydroxytamoxifen (4-OHT)-dependent switchable c-myc oncoprotein under the control of the proximal lck promoter. Activation of c-MycER causes no obvious alteration in T cell ontogeny. However, using MHC class I restricted H-Y-TCR transgenic mice, we found that c-Myc activation in vivo enhances the efficiency of positive selection. Moreover, splenic T cells derived from lck-c-mycER transgenic mice in which c-Myc had been activated exhibited increased proliferation in vitro in response to activation with anti-CD3/CD28 antibody. Activation of c-MycER also promotes apoptosis in thymocytes in vitro.


Subject(s)
Apoptosis , Gene Expression Regulation, Neoplastic , Genes, myc , T-Lymphocytes/metabolism , Animals , Cell Differentiation , Cell Division , Mice , Mice, Transgenic , T-Lymphocytes/physiology , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Thymus Gland/cytology
11.
Nat Cell Biol ; 2(3): 156-62, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10707086

ABSTRACT

Release of cytochrome c from mitochondria triggers activation of caspase proteases and death of a cell by apoptosis. However, the mechanism and kinetics of cytochrome c release remain unknown. Here we study this event by using green fluorescent protein (GFP)-tagged cytochrome c, and find that the release of cytochrome-c-GFP always precedes exposure of phosphatidylserine and the loss of plasma-membrane integrity - characteristics of apoptotic cells. Once initiated, the release of cytochrome- c-GFP continues until all of the protein is released from all mitochondria in individual cells, within about 5 minutes, regardless of the type or strength of stimulus or the time elapsed since the stimulus was applied. Temperatures ranging from 24 degrees C to 37 degrees C do not change the duration of release, and nor does the addition of caspase inhibitors. Further, we find that the electron-transport chain can maintain the mitochondrial transmembrane potential even after cytochrome c has been released.


Subject(s)
Apoptosis , Cytochrome c Group/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Caspase Inhibitors , Caspases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Cytochrome c Group/genetics , Digitonin/pharmacology , Electron Transport/drug effects , Electron Transport/radiation effects , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins , HeLa Cells , Humans , Image Processing, Computer-Assisted , Intracellular Membranes/drug effects , Intracellular Membranes/radiation effects , Luminescent Proteins/genetics , Membrane Potentials/drug effects , Mitochondria/metabolism , Oligomycins/pharmacology , Phosphatidylserines/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sodium Azide/pharmacology , Temperature , Ultraviolet Rays
12.
EMBO J ; 19(4): 598-611, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10675329

ABSTRACT

We have isolated the recently identified Drosophila caspase DRONC through its interaction with the effector caspase drICE. Ectopic expression of DRONC induces cell death in Schizosaccharomyces pombe, mammalian fibroblasts and the developing Drosophila eye. The caspase inhibitor p35 fails to rescue DRONC-induced cell death in vivo and is not cleaved by DRONC in vitro, making DRONC the first identified p35-resistant caspase. The DRONC pro-domain interacts with Drosphila inhibitor of apoptosis protein 1 (DIAP1), and co-expression of DIAP1 in the developing Drosophila eye completely reverts the eye ablation phenotype induced by pro-DRONC expression. In contrast, DIAP1 fails to rescue eye ablation induced by DRONC lacking the pro-domain, indicating that interaction of DIAP1 with the pro-domain of DRONC is required for suppression of DRONC-mediated cell death. Heterozygosity at the diap1 locus enhances the pro-DRONC eye phenotype, consistent with a role for endogenous DIAP1 in suppression of DRONC activation. Both heterozygosity at the dronc locus and expression of dominant-negative DRONC mutants suppress the eye phenotype caused by reaper (RPR) and head involution defective (HID), consistent with the idea that DRONC functions in the RPR and HID pathway.


Subject(s)
Caspases/genetics , Drosophila Proteins , Drosophila/enzymology , Insect Proteins/genetics , Animals , Apoptosis/genetics , Apoptosis/physiology , Caspase Inhibitors , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Drosophila/genetics , Drosophila/growth & development , Eye/growth & development , Gene Expression Regulation , Genes, Insect , Heterozygote , Inhibitor of Apoptosis Proteins , Phenotype , Promoter Regions, Genetic , Rats , Schizosaccharomyces/genetics , Transfection , Two-Hybrid System Techniques
13.
Proc Natl Acad Sci U S A ; 97(6): 2875-80, 2000 Mar 14.
Article in English | MEDLINE | ID: mdl-10688892

ABSTRACT

Caspase-3 is an effector of apoptosis in experimental models of Parkinson's disease (PD). However, its potential role in the human pathology remains to be demonstrated. Using caspase-3 immunohistochemistry on the postmortem human brain, we observed a positive correlation between the degree of neuronal loss in dopaminergic (DA) cell groups affected in the mesencephalon of PD patients and the percentage of caspase-3-positive neurons in these cell groups in control subjects and a significant decrease of caspase-3-positive pigmented neurons in the substantia nigra pars compacta of PD patients compared with controls that also could be observed in an animal model of PD. This suggests that neurons expressing caspase-3 are more sensitive to the pathological process than those that do not express the protein. In addition, using an antibody raised against activated caspase-3, the percentage of active caspase-3-positive neurons among DA neurons was significantly higher in PD patients than in controls. Finally, electron microscopy analysis in the human brain and in vitro data suggest that caspase-3 activation precedes and is not a consequence of apoptotic cell death in PD.


Subject(s)
Apoptosis , Brain/enzymology , Caspases/biosynthesis , Caspases/physiology , Dopamine/metabolism , Parkinson Disease/enzymology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Aged , Aged, 80 and over , Animals , Blotting, Western , Brain/ultrastructure , Caspase 3 , Cells, Cultured , Dopamine Agents/pharmacology , Enzyme Activation , Humans , Immunohistochemistry , Male , Mesencephalon/enzymology , Mice , Mice, Inbred C57BL , Neurons/enzymology , Neurons/ultrastructure , Parkinson Disease/metabolism , Rats , Substantia Nigra/enzymology , Tissue Distribution , Ventral Tegmental Area/enzymology
14.
IUBMB Life ; 50(2): 85-90, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11185963

ABSTRACT

Caspase-8 is a member of the cysteine proteases, which are implicated in apoptosis and cytokine processing. Like all caspases, caspase-8 is synthesized as an inactive single polypeptide chain zymogen procaspase and is activated by proteolytic cleavage, through either autoactivation after recruitment into a multimeric complex or trans-cleavage by other caspases. Thus, ligand binding-induced trimerization of death receptors results in recruitment of the receptor-specific adapter protein Fas-associated death domain (FADD), which then recruits caspase-8. Activated caspase-8 is known to propagate the apoptotic signal either by directly cleaving and activating downstream caspases or by cleaving the BH3 Bcl2-interacting protein, which leads to the release of cytochrome c from mitochondria, triggering activation of caspase-9 in a complex with dATP and Apaf-1. Activated caspase-9 then activates further "downstream caspases," including caspase-8. Knockout data indicate that caspase-8 is required for killing induced by the death receptors Fas, tumor necrosis factor receptor 1, and death receptor 3. Moreover, caspase-8-/- mice die in utero as a result of defective development of heart muscle and display fewer hematopoietic progenitor cells, suggesting that the FADD/caspase-8 pathway is absolutely required for growth and development of specific cell types.


Subject(s)
Apoptosis , Arabidopsis Proteins , Caspases/metabolism , Caspases/physiology , Fatty Acid Desaturases/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptotic Protease-Activating Factor 1 , Caspase 8 , Caspase 9 , Cytochrome c Group/metabolism , Enzyme Activation , Humans , Ligands , Mice , Mice, Knockout , Proteins/metabolism
15.
Oncogene ; 18(13): 2181-8, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10327064

ABSTRACT

The contact of natural killer (NK) cells with foreign cells and with certain virus-infected or tumor cells triggers the cytolytic machinery of NK cells. This triggering leads to exocytosis of the cytotoxic NK cell granules. The oncoproteins c-Myc and E1A render cells vulnerable to NK cell mediated cytolysis yet the mechanisms of sensitization are not well understood. In a model where foreign cells (rat fibroblasts) were cocultured with human IL-2 activated NK cells, we observed that NK cells were capable of efficiently killing their targets only if the cells overexpressed the oncogene c-Myc or E1A. Both the parental and the oncogene expressing fibroblasts similarly triggered phosphoinositide hydrolysis in the bound NK cells, demonstrating that NK cells were cytolytically activated in contact with both resistant parental and oncogene expressing sensitive target fibroblasts. The cell death was independent of wild-type p53 and was not inhibited by an anti-apoptotic protein EIB19K. These results provided evidence that c-Myc and E1A activated the NK cell induced cytolysis at a post-triggering stage of NK cell-target cell interaction. In consistence, the c-Myc and E1A overexpressing fibroblasts were more sensitive to the cytolytic effects of isolated NK cell-derived granules than parental cells. The data indicate that oncogenes activate the cytotoxicity of NK cell granules. This mechanism can have a role in directing the cytolytic action of NK cells towards the virus-infected and cancer cells.


Subject(s)
Adenovirus E1A Proteins/physiology , Cytoplasmic Granules/metabolism , Cytotoxicity, Immunologic/physiology , Killer Cells, Natural/immunology , Proto-Oncogene Proteins c-myc/physiology , Actins/chemistry , Adenovirus E1A Proteins/genetics , Animals , Apoptosis/genetics , Biopolymers , Cell Adhesion , Cell Membrane/drug effects , Exocytosis , Fas Ligand Protein , Fibroblasts/immunology , Genes, myc , Genes, p53 , Humans , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Membrane Glycoproteins/physiology , Phosphatidylinositols/physiology , Rats , Recombinant Fusion Proteins/physiology , Signal Transduction , Transfection , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology , Tumor Suppressor Protein p53/physiology , fas Receptor/physiology
16.
Curr Biol ; 9(6): 292-301, 1999 Mar 25.
Article in English | MEDLINE | ID: mdl-10209096

ABSTRACT

BACKGROUND: Inhibitor of apoptosis proteins (IAPs) suppress apoptotic cell death in several model systems and are highly conserved between insects and mammals. All IAPs contain at least one copy of the approximately 70 amino-acid baculovirus IAP repeat (BIR), and this domain is essential for the anti-apoptotic activity of the IAPs. Both the marked structural diversity of IAPs and the identification of BIR-containing proteins (BIRPs) in yeast, however, have led to the suggestion that BIRPs might play roles in other, as yet unidentified, cellular processes besides apoptosis. Survivin, a human BIRP, is upregulated 40-fold at G2-M phase and binds to mitotic spindles, although its role at the spindle is still unclear. RESULTS: We have identified and characterised two Caenorhabditis elegans BIRPs,BIR-1 and BIR-2; these proteins are the only BIRPs in C. elegans. The bir-1 gene is highly expressed during embryogenesis with detectable expression throughout other stages of development; bir-2 expression is detectable only in adults and embryos. Overexpression of bir-1 was unable to inhibit developmentally occurring cell death in C. elegans and inhibition of bir-1 expression did not increase cell death. Instead, embryos lacking bir-1 were unable to complete cytokinesis and they became multinucleate. This cytokinesis defect could be partially suppressed by transgenic expression of survivin, the mammalian BIRP most structurally related to BIR-1, suggesting a conserved role for BIRPs in the regulation of cytokinesis. CONCLUSIONS: BIR-1, a C. elegans BIRP, is probably not involved in the general regulation of apoptosis but is required for embryonic cytokinesis. We suggest that BIRPs may regulate cytoskeletal changes in diverse biological processes including cytokinesis and apoptosis.


Subject(s)
Apoptosis/physiology , Caenorhabditis elegans Proteins , Caenorhabditis elegans/physiology , Cell Division/physiology , Genes, Helminth , Helminth Proteins/physiology , Microtubule-Associated Proteins , Amino Acid Sequence , Animals , Animals, Genetically Modified , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caspase Inhibitors , Caspases/physiology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Germ Cells/cytology , Helminth Proteins/antagonists & inhibitors , Helminth Proteins/genetics , Humans , Inhibitor of Apoptosis Proteins , Mammals/genetics , Molecular Sequence Data , Neoplasm Proteins , Proteins/genetics , Proteins/physiology , RNA, Double-Stranded/pharmacology , Recombinant Fusion Proteins/physiology , Saccharomyces cerevisiae/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Survivin , Transcription, Genetic/drug effects
17.
Neuroreport ; 9(15): 3409-13, 1998 Oct 26.
Article in English | MEDLINE | ID: mdl-9855290

ABSTRACT

Retinal ganglion cells of the fish have the spontaneous capacity to regenerate after nerve crush, a phenomenon known to be facilitated by nerve growth factor (NGF). We have studied the high-affinity NGF receptor TrkA, during the regeneration of the tench (Tinca tinca L.) optic nerve, using immunocytochemical techniques. TrkA-like immunoreactivity increased during the regeneration of the retinal ganglion cells. The increase is followed by a change in the subcellular distribution from perinuclear in control cells to cytoplasmic and perinuclear in regenerating ones. This increase was observed when antibodies against the extracellular domain of TrkA were used; no changes in TrkA-like immunoreactivity were observed with antibodies against the intracellular domain of TrkA. We thus conclude that modulation of TrkA is involved in the regeneration of fish retinal ganglion cells.


Subject(s)
Nerve Regeneration/physiology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Nerve Growth Factor/metabolism , Retinal Ganglion Cells/chemistry , Retinal Ganglion Cells/cytology , 3T3 Cells , Animals , Antibodies , Antibody Specificity , Blotting, Western , Cyprinidae , Epitopes/analysis , Immunohistochemistry , Mice , Nerve Crush , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/analysis , Receptor Protein-Tyrosine Kinases/immunology , Receptor, trkA , Receptors, Nerve Growth Factor/analysis , Receptors, Nerve Growth Factor/immunology
18.
Trends Genet ; 14(9): 364-7, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9769732

ABSTRACT

The MYC proto-oncogene has long been implicated in the control of normal cell growth and its deregulation is associated with the development of neoplasia. The MYC protein has a well-established role as a component of signal-transduction pathways promoting both proliferation and apoptosis. Because signalling pathways that drive cell death and cell proliferation are so tightly coupled, a synergy between genetic lesions leading to suppression of cell death and those promoting cell proliferation is observed during carcinogenesis. We discuss such synergy with respect to the cooperating oncogenes MYC, RAS and BCL2.


Subject(s)
Oncogenes , Animals , Apoptosis , Cell Division , Genes, bcl-2 , Genes, myc , Genes, ras , Humans , Neoplasms/etiology , Neoplasms/genetics , Proto-Oncogene Mas , Signal Transduction
19.
J Neurosci ; 18(17): 6767-75, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9712648

ABSTRACT

Expression of the nerve growth factor (NGF) receptors TrkA and p75(NTR) was found to vary at the surface of PC12 cells in a cell cycle phase-specific manner. This was evidenced by using flow cytometric and microscopic analysis of cell populations labeled with antibodies to the extracellular domains of both receptors. Differential expression of these receptors also was evidenced by biotinylation of surface proteins and Western analysis, using antibodies specific for the extracellular domains of TrkA and p75(NTR). TrkA is expressed most strongly at the cell surface in M and early G1 phases, whereas p75(NTR) is expressed mainly in late G1, S, and G2 phases. This expression reflects the molecular and cellular responses to NGF in specific phases of the cell cycle; in the G1 phase NGF elicits both the anti-mitogenic effect, i.e., inhibition of the G1 to S transition, and the differentiation response whereas a survival effect is provoked elsewhere in the cell cycle. A model is proposed relating these responses to the surface expression of the two receptors. These observations open the way for novel approaches to the investigation of the mechanism of NGF signal transduction.


Subject(s)
Cell Cycle/physiology , Proto-Oncogene Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Nerve Growth Factor/biosynthesis , Animals , Biotinylation , PC12 Cells , Rats , Receptor, Nerve Growth Factor , Receptor, trkA
SELECTION OF CITATIONS
SEARCH DETAIL
...