Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 339
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Exp Med ; 149(1): 100-13, 1979 Jan 01.
Article in English | MEDLINE | ID: mdl-216763

ABSTRACT

When deprived of oxygen, Bacille Calmette-Guérin (BCG)-activated macrophages no longer lysed P388 lymphoma cells. Both H2O2 release and cytotoxicity by BCG-activated macrophages and by granulocytes triggered with phorbol myristate acetate (PMA) were markedly inhibited when the glucose concentration in the medium was reduced to 0.03 mM or less, or if glucose were replaced with galactose. Catalase abolished PMA-triggered cytotoxicity by both types of effector cells, whereas superoxide dismutase had no effect. Ferricytochrome C reduced the cytotoxicity of BCG-activated macrophages, an effect which was largely reversed by superoxide dismutase. 10 drugs, thought to quench singlet oxygen and/or scavenge hydroxyl radical, did not affect cytotoxicity in this system. Neither azide nor cyanide reduced cytolysis, but there was marked inhibition by lactoperoxidase and iodide. This suggested that cytotoxicity was not dependent upon myeloperoxidase, and that lactoperoxidase may have diverted H2O2 from the oxidation of target cells to oxidation of substances in serum. Mouse erythrocytes, although sensitive targets, interfered with the cytolysis of lymphoma cells, probably by competition for H2O2. Starch particles with covalently bound glucose oxidase resembled macrophages in their spatial relation to the target cells and in the flux of H2O2 they generated from their surface, but were not expected to produce any other potentially toxic products. Such particles lysed lymphoma cells, and the lysis was prevented by catalase. Neither arginase nor thymidine appeared to be involved in cytolysis by BCG-activated macrophages under the conditions used. These findings demonstrated that release of H2O2 was both necessary and sufficient for cytolysis by BCG-activated macrophages and by granulocytes when pharmacologically triggered.


Subject(s)
Granulocytes/physiology , Hydrogen Peroxide/metabolism , Macrophages/physiology , Animals , Catalase/metabolism , Cytochrome c Group/pharmacology , Free Radicals , Glucose/metabolism , Glucose Oxidase/metabolism , Mice , Oxygen/physiology , Peroxidases/metabolism , Superoxide Dismutase/metabolism , Tetradecanoylphorbol Acetate/pharmacology
2.
J Cell Biol ; 167(3): 405-10, 2004 Nov 08.
Article in English | MEDLINE | ID: mdl-15533997

ABSTRACT

In Drosophila, activation of the apical caspase DRONC requires the apoptotic protease-activating factor homologue, DARK. However, unlike caspase activation in mammals, DRONC activation is not accompanied by the release of cytochrome c from mitochondria. Drosophila encodes two cytochrome c proteins, Cytc-p (DC4) the predominantly expressed species, and Cytc-d (DC3), which is implicated in caspase activation during spermatogenesis. Here, we report that silencing expression of either or both DC3 and DC4 had no effect on apoptosis or activation of DRONC and DRICE in Drosophila cells. We find that loss of function mutations in dc3 and dc4, do not affect caspase activation during Drosophila development and that ectopic expression of DC3 or DC4 in Drosophila cells does not induce caspase activation. In cell-free studies, recombinant DC3 or DC4 failed to activate caspases in Drosophila cell lysates, but remarkably induced caspase activation in extracts from human cells. Overall, our results argue that DARK-mediated DRONC activation occurs independently of cytochrome c.


Subject(s)
Apoptosis , Caspases/metabolism , Cytochrome c Group/physiology , Drosophila melanogaster/cytology , Animals , Cell Line , Cytochrome c Group/metabolism , Cytochrome c Group/pharmacology , Cytochromes c/physiology , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Enzyme Activation , Humans , Mutation , Protein Binding , Recombinant Proteins/pharmacology
3.
J Cell Biol ; 149(3): 623-34, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10791976

ABSTRACT

How Bcl-2 and its pro-survival relatives prevent activation of the caspases that mediate apoptosis is unknown, but they appear to act through the caspase activator apoptosis protease-activating factor 1 (Apaf-1). According to the apoptosome model, the Bcl-2-like proteins preclude Apaf-1 activity by sequestering the protein. To explore Apaf-1 function and to test this model, we generated monoclonal antibodies to Apaf-1 and used them to determine its localization within diverse cells by subcellular fractionation and confocal laser scanning microscopy. Whereas Bcl-2 and Bcl-x(L) were prominent on organelle membranes, endogenous Apaf-1 was cytosolic and did not colocalize with them, even when these pro-survival proteins were overexpressed or after apoptosis was induced. Immunogold electron microscopy confirmed that Apaf-1 was dispersed in the cytoplasm and not on mitochondria or other organelles. After the death stimuli, Bcl-2 and Bcl-x(L) precluded the release of the Apaf-1 cofactor cytochrome c from mitochondria and the formation of larger Apaf-1 complexes, which are steps that presage apoptosis. However, neither Bcl-2 nor Bcl-x(L) could prevent the in vitro activation of Apaf-1 induced by the addition of exogenous cytochrome c. Hence, rather than sequestering Apaf-1 as proposed by the apoptosome model, Bcl-2-like proteins probably regulate Apaf-1 indirectly by controlling upstream events critical for its activation.


Subject(s)
Cytoplasm/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Antibodies, Monoclonal/immunology , Apoptosis , Apoptotic Protease-Activating Factor 1 , Caspases/metabolism , Cell Line , Cytochrome c Group/pharmacology , Cytoplasm/ultrastructure , Enzyme Activation , Fluorescent Antibody Technique , Humans , Microscopy, Confocal , Microscopy, Immunoelectron , Proteins/immunology , bcl-X Protein
4.
J Cell Biol ; 144(5): 915-26, 1999 Mar 08.
Article in English | MEDLINE | ID: mdl-10085291

ABSTRACT

The mechanism by which membrane-bound Bcl-2 inhibits the activation of cytoplasmic procaspases is unknown. Here we characterize an intracellular, membrane-associated form of procaspase-3 whose activation is controlled by Bcl-2. Heavy membranes isolated from control cells contained a spontaneously activatable caspase-3 zymogen. In contrast, in Bcl-2 overexpressing cells, although the caspase-3 zymogen was still associated with heavy membranes, its spontaneous activation was blocked. However, Bcl-2 expression had little effect on the levels of cytoplasmic caspase activity in unstimulated cells. Furthermore, the membrane-associated caspase-3 differed from cytosolic caspase-3 in its responsiveness to activation by exogenous cytochrome c. Our results demonstrate that intracellular membranes can generate active caspase-3 by a Bcl-2-inhibitable mechanism, and that control of caspase activation in membranes is distinct from that observed in the cytoplasm. These data suggest that Bcl-2 may control cytoplasmic events in part by blocking the activation of membrane-associated procaspases.


Subject(s)
Caspases/metabolism , Enzyme Precursors/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Apoptosis , Caspase 3 , Caspase Inhibitors , Cell Line , Coumarins/metabolism , Cytochrome c Group/pharmacology , Enzyme Activation , Enzyme Precursors/antagonists & inhibitors , Humans , Hydrolysis , Oligopeptides/metabolism , Subcellular Fractions/metabolism
5.
J Cell Biol ; 151(3): 483-94, 2000 Oct 30.
Article in English | MEDLINE | ID: mdl-11062251

ABSTRACT

Phosphoinositide 3 kinase/Akt pathway plays an essential role in neuronal survival. However, the cellular mechanisms by which Akt suppresses cell death and protects neurons from apoptosis remain unclear. We previously showed that transient expression of constitutively active Akt inhibits ceramide-induced death of hybrid motor neuron 1 cells. Here we show that stable expression of either constitutively active Akt or Bcl-2 inhibits apoptosis, but only Bcl-2 prevents the release of cytochrome c from mitochondria, suggesting that Akt regulates apoptosis at a postmitochondrial level. Consistent with this, overexpressing active Akt rescues cells from apoptosis without altering expression levels of endogenous Bcl-2, Bcl-x, or Bax. Akt inhibits apoptosis induced by microinjection of cytochrome c and lysates from cells expressing active Akt inhibit cytochrome c induced caspase activation in a cell-free assay while lysates from Bcl-2-expressing cells have no effect. Addition of cytochrome c and dATP to lysates from cells expressing active Akt do not activate caspase-9 or -3 and immunoprecipitated Akt added to control lysates blocks cytochrome c-induced activation of the caspase cascade. Taken together, these data suggest that Akt inhibits activation of caspase-9 and -3 by posttranslational modification of a cytosolic factor downstream of cytochrome c and before activation of caspase-9.


Subject(s)
Apoptosis , Mitochondria/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Sphingosine/analogs & derivatives , Apoptosis/drug effects , Caspase 3 , Caspase 9 , Caspases/metabolism , Cell Extracts , Cell Line , Cell Survival/drug effects , Cell-Free System , Cytochrome c Group/administration & dosage , Cytochrome c Group/antagonists & inhibitors , Cytochrome c Group/metabolism , Cytochrome c Group/pharmacology , Enzyme Activation/drug effects , Humans , Hybrid Cells , Microinjections , Mitochondria/drug effects , Mitochondria/metabolism , Motor Neurons/cytology , Motor Neurons/drug effects , Motor Neurons/enzymology , Motor Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Precipitin Tests , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Sphingosine/pharmacology , bcl-2-Associated X Protein , bcl-X Protein
6.
Science ; 282(5392): 1318-21, 1998 Nov 13.
Article in English | MEDLINE | ID: mdl-9812896

ABSTRACT

Caspases are intracellular proteases that function as initiators and effectors of apoptosis. The kinase Akt and p21-Ras, an Akt activator, induced phosphorylation of pro-caspase-9 (pro-Casp9) in cells. Cytochrome c-induced proteolytic processing of pro-Casp9 was defective in cytosolic extracts from cells expressing either active Ras or Akt. Akt phosphorylated recombinant Casp9 in vitro on serine-196 and inhibited its protease activity. Mutant pro-Casp9(Ser196Ala) was resistant to Akt-mediated phosphorylation and inhibition in vitro and in cells, resulting in Akt-resistant induction of apoptosis. Thus, caspases can be directly regulated by protein phosphorylation.


Subject(s)
Apoptosis , Caspases/metabolism , Caspase 9 , Caspase Inhibitors , Cell Line , Cytochrome c Group/pharmacology , Enzyme Precursors/metabolism , Humans , Mass Spectrometry , Mutation , Peptide Fragments/metabolism , Phosphorylation , Phosphoserine/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins p21(ras)/metabolism , Recombinant Fusion Proteins/metabolism , Transfection
7.
Science ; 267(5197): 515-8, 1995 Jan 27.
Article in English | MEDLINE | ID: mdl-7824949

ABSTRACT

Small changes in the peptide-major histocompatibility complex (MHC) molecule ligands recognized by antigen-specific T cell receptors (TCRs) can convert fully activating complexes into partially activating or even inhibitory ones. This study examined early TCR-dependent signals induced by such partial agonists or antagonists. In contrast to typical agonist ligands, both an antagonist and several partial agonists stimulated a distinct pattern of zeta chain phosphorylation and failed to activate associated ZAP-70 kinase. These results identify a specific step in the early tyrosine phosphorylation cascade that is altered after TCR engagement with modified peptide-MHC molecule complexes. This finding may explain the different biological responses to TCR occupancy by these variant ligands.


Subject(s)
Histocompatibility Antigens Class II/pharmacology , Membrane Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Amino Acid Sequence , Animals , Clone Cells , Cytochrome c Group/pharmacology , Enzyme Activation , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Interleukin-2/biosynthesis , L Cells , Ligands , Lymphocyte Activation , Mice , Molecular Sequence Data , Mutation , Peptide Fragments/pharmacology , Phosphorylation , Receptors, Antigen, T-Cell/agonists , Receptors, Antigen, T-Cell/antagonists & inhibitors , Signal Transduction , Tyrosine/metabolism , ZAP-70 Protein-Tyrosine Kinase
8.
Neuron ; 21(1): 77-85, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9697853

ABSTRACT

Vertebrate neurogenesis is initiated by the organizer factors that inhibit antineuralizing activities of bone morphogenetic proteins (BMPs) in the ectoderm. Here, we report a candidate mediator of neuralization, SoxD. Expression of SoxD starts at late blastula stages widely in the prospective ectoderm and becomes restricted to the dorsal ectoderm by mid-gastrula stages. SoxD expression is enhanced by the neural inducer Chordin and is suppressed by BMP4 and its downstream genes. Microinjection of SoxD mRNA causes ectopic formation of neural tissues in vivo and induces neural and neuronal markers in the isolated animal cap. Injection of a dominant-negative form of SoxD mRNA can block neuralization of ectoderm caused by attenuation of BMP signals and can strongly suppress formation of anterior neural tissues in vivo. These data show that SoxD functions as an essential mediator of downstream signaling of neural induction.


Subject(s)
Bacterial Proteins , Cytochrome c Group/physiology , Nerve Tissue/embryology , Xenopus/embryology , Amino Acid Sequence , Animals , Bone Morphogenetic Proteins/physiology , Cytochrome c Group/genetics , Cytochrome c Group/pharmacology , Ectoderm/cytology , Ectoderm/drug effects , Embryo, Nonmammalian/physiology , Gene Expression/physiology , Molecular Sequence Data , Signal Transduction/physiology
9.
Neuron ; 21(4): 695-705, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9808457

ABSTRACT

Sympathetic neurons undergoing programmed cell death after nerve growth factor (NGF) deprivation are shown to exhibit a protein synthesis-dependent, BAX-dependent loss of cytochrome c from the mitochondria. However, cytoplasmic microinjection of cytochrome c was insufficient to induce cell death in NGF-maintained sympathetic neurons. In contrast, microinjection of cytochrome c rapidly induced a caspase-dependent death in NGF-deprived, Bax-deficient or NGF-deprived, cycloheximide-treated neurons. Cells needed to be deprived of NGF for 15-20 hr before they acquired competence to die with injection of cytochrome c. These data suggest that NGF deprivation induced the translocation of cytochrome c and another event, which we term as competence-to-die, that was independent of macromolecular synthesis and BAX function. Both these processes were required for neurons to undergo apoptosis.


Subject(s)
Cytochrome c Group/pharmacology , Cytoplasm/physiology , Neurons/drug effects , Neurons/physiology , Proto-Oncogene Proteins c-bcl-2 , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Caspases/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cycloheximide/pharmacology , Cytochrome c Group/metabolism , Dose-Response Relationship, Drug , Mice , Microinjections , Mitochondria/metabolism , Nerve Growth Factors/deficiency , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/metabolism , Superior Cervical Ganglion/cytology , bcl-2-Associated X Protein
10.
Neuron ; 32(2): 289-300, 2001 Oct 25.
Article in English | MEDLINE | ID: mdl-11683998

ABSTRACT

We have examined the roles of Hsc70 and auxilin in the uncoating of clathrin-coated vesicles (CCVs) during neuronal endocytosis. We identified two peptides that inhibit the ability of Hsc70 and auxilin to uncoat CCVs in vitro. When injected into nerve terminals, these peptides inhibited both synaptic transmission and CCV uncoating. Mutation of a conserved HPD motif within the J domain of auxilin prevented binding to Hsc70 in vitro and injecting this mutant protein inhibited CCV uncoating in vivo, demonstrating that the interaction of auxilin with Hsc70 is critical for CCV uncoating. These studies establish that auxilin and Hsc70 participate in synaptic vesicle recycling in neurons and that an interaction between these proteins is required for CCV uncoating.


Subject(s)
Carrier Proteins/physiology , Clathrin-Coated Vesicles/metabolism , HSP70 Heat-Shock Proteins/physiology , Membrane Proteins/physiology , Presynaptic Terminals/metabolism , Adaptor Proteins, Vesicular Transport , Adenosine Triphosphate/pharmacology , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Carrier Proteins/pharmacology , Cattle , Cytochrome c Group/chemistry , Cytochrome c Group/pharmacology , Endocytosis , HSC70 Heat-Shock Proteins , HSP70 Heat-Shock Proteins/pharmacology , Membrane Proteins/chemistry , Membrane Proteins/pharmacology , Molecular Sequence Data , Mutagenesis , Neurons/ultrastructure , Neurotransmitter Agents/metabolism , Peptide Fragments/pharmacology , Synaptic Transmission/drug effects
11.
Nat Neurosci ; 4(1): 29-37, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11135642

ABSTRACT

Although the requirement of neurotrophins for the prevention of cell death in the peripheral nervous system is well established, their physiological involvement in nerve growth is still unclear. To address this question, we generated a mouse that expresses the green fluorescent protein in post-mitotic neurons, allowing the repeated visualization of all motor and sensory axons during development. We imaged the growth of these axons into the limb bud of day 10.5 embryos. Sensory axons, but rarely motor axons, were targeted to ectopically placed beads containing any of the neurotrophins NGF, BDNF, NT-3 or NT-4/5. Conversely, a combination of function-blocking monoclonal antibodies to NGF, BDNF and NT-3 dramatically inhibited elongation of both sensory and motor axons in the limb bud, indicating that the growth of mixed nerves is dependent upon neurotrophins during development.


Subject(s)
Nerve Growth Factors/metabolism , Peripheral Nerves/growth & development , Animals , Antibodies, Monoclonal/pharmacology , Axons/drug effects , Axons/physiology , Brain-Derived Neurotrophic Factor/pharmacology , Cell Differentiation/drug effects , Cell Division/drug effects , Cytochrome c Group/pharmacology , Drug Carriers , Limb Buds/embryology , Limb Buds/growth & development , Limb Buds/innervation , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Motor Neurons/cytology , Motor Neurons/drug effects , Motor Neurons/metabolism , Nerve Growth Factor/pharmacology , Nerve Growth Factors/pharmacology , Neurons, Afferent/cytology , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Peripheral Nerves/drug effects , Peripheral Nerves/embryology , Spinal Nerves
12.
Curr Biol ; 9(3): 147-50, 1999 Feb 11.
Article in English | MEDLINE | ID: mdl-10021389

ABSTRACT

Caspases, a family of specific proteases, have central roles in apoptosis [1]. Caspase activation in response to diverse apoptotic stimuli involves the relocalisation of cytochrome c from mitochondria to the cytoplasm where it stimulates the proteolytic processing of caspase precursors. Cytochrome c release is controlled by members of the Bcl-2 family of apoptosis regulators [2] [3]. The anti-apoptotic members Bcl-2 and Bcl-xL may also control caspase activation independently of cytochrome c relocalisation or may inhibit a positive feedback mechanism [4] [5] [6] [7]. Here, we investigate the role of Bcl-2 family proteins in the regulation of caspase activation using a model cell-free system. We found that Bcl-2 and Bcl-xL set a threshold in the amount of cytochrome c required to activate caspases, even in soluble extracts lacking mitochondria. Addition of dATP (which stimulates the procaspase-processing factor Apaf-1 [8] [9]) overcame inhibition of caspase activation by Bcl-2, but did not prevent the control of cytochrome c release from mitochondria by Bcl-2. Cytochrome c release was accelerated by active caspase-3 and this positive feedback was negatively regulated by Bcl-2. These results provide evidence for a mechanism to amplify caspase activation that is suppressed at several distinct steps by Bcl-2, even after cytochrome c is released from mitochondria.


Subject(s)
Caspases/metabolism , Cytochrome c Group/pharmacology , Proto-Oncogene Proteins c-bcl-2/pharmacology , Animals , Apoptosis , Caspase 3 , Cell-Free System , Cytochrome c Group/physiology , Enzyme Activation/drug effects , Feedback , HeLa Cells , Humans , Mitochondria/enzymology , Oligopeptides/pharmacology , Oocytes , Proto-Oncogene Proteins c-bcl-2/physiology , Recombinant Fusion Proteins/pharmacology , Xenopus Proteins , Xenopus laevis , bcl-X Protein
13.
J Clin Invest ; 60(6): 1266-79, 1977 Dec.
Article in English | MEDLINE | ID: mdl-199619

ABSTRACT

Normal and cytochalasin B-treated human granulocytes have been studied to determine some of the interrelationships between phagocytosis-induced respiration and superoxide and hydrogen peroxide formation and release into the extracellular medium by intact cells. By using the scopoletin fluorescent assay to continuously monitor extracellular hydrogen peroxide concentrations during contact of cells with opsonized staphylococci, it was demonstrated that the superoxide scavengers ferricytochrome c and nitroblue tetrazolium significantly reduced the amount of H(2)O(2) released with time from normal cells but did not abolish it. This inhibitory effect was reversed by the simultaneous addition of superoxide dismutase (SOD), whereas the addition of SOD alone increased the amount of detectable H(2)O(2) in the medium. The addition of sodium azide markedly inhibited myeloperoxidase-H(2)O(2)-dependent protein iodination and more than doubled H(2)O(2) release, including the residual amount remaining after exposure of the cells to ferricytochrome c, suggesting its origin from an intracellular pool shared by several pathways for H(2)O(2) catabolism. When cells were pretreated with cytochalasin B and opsonized bacteria added, reduced oxygen consumption was observed, but this was in parallel to a reduction in specific binding of organisms to the cells when compared to normal. Under the influence of inhibited phagosome formation by cytochalasin B, the cells released an increased amount of superoxide and peroxide into the extracellular medium relative to oxygen consumption, and all detectable peroxide release could be inhibited by the addition of ferricytochrome c. Decreased H(2)O(2) production in the presence of this compound could not be ascribed to diminished bacterial binding, decreased oxidase activity, or increased H(2)O(2) catabolism and was reversed by the simultaneous addition of SOD. Furthermore, SOD and ferricytochrome c had similar effects on another H(2)O(2)-dependent reaction, protein iodination, in both normal and cytochalasin B cells. When oxygen consumption, O(2.) (-), and H(2)O(2) release were compared in the presence of azide under identical incubation conditions, the molar relationships for normal cells were 1.00:0.34:0.51 and for cytochalasin B-treated cells 1.00:0.99:0.40, respectively. Nonopsonized, or opsonized but disrupted, bacteria did not stimulate any of these metabolic functions. The results indicate that with normal cells approximately 50% of H(2)O(2) released during phagocytosis is derived directly from O(2.) (-) by dismutation, the remainder appearing from an (intra)cellular source shared by azide-inhibitable heme enzymes. With cytochalasin B treatment the evidence is consistent with the derivation of all H(2)O(2) from an O(2.) (-) precursor which is released from the cell surface. Furthermore, when activated by phagocytic particle binding, the neutrophil O(2.) (-) generating system appears to make more of this compound than can be accounted for by dismutation to H(2)O(2). This establishes conditions for the direct participation of both compounds in the microbicidal and cytocidal activity of these cells.


Subject(s)
Cytochalasin B/pharmacology , Granulocytes/metabolism , Hydrogen Peroxide/metabolism , Leukocytes/metabolism , Phagocytosis , Azides/pharmacology , Cytochrome c Group/pharmacology , Horseradish Peroxidase , Humans , Nitroblue Tetrazolium/pharmacology , Scopoletin , Superoxide Dismutase/pharmacology , Superoxides/biosynthesis
14.
J Clin Invest ; 73(6): 1629-37, 1984 Jun.
Article in English | MEDLINE | ID: mdl-6327766

ABSTRACT

Chemotaxis and generation of the oxidative burst by phagocytes are among the biological functions thought to require methylation reaction(s) for their expression. The present study investigated the effect of different stimuli of the oxidative burst on lipid methylation by human elutriated monocytes as measured by methyl group incorporation from [methyl-3H]methionine into both phospholipid and neutral lipid extracts. Normal monocytes, incubated at 37 degrees C for 1 h with 2 microM methionine, incorporated 10.2-fmol/10(6) cells and 73.6-fmol/10(6) cells of methyl groups into neutral lipids and phospholipids, respectively. Stimulators of the respiratory burst, such as the chemotactic peptide N-formyl-L-methionyl-L-leucyl-L-phenylalanine, the tumor promoter, 12-O-tetradecanoyl phorbol-13-acetate, and the calcium ionophore, A23187, decreased the incorporation of methyl groups into both neutral lipids and phospholipids in a similar manner. Increasing the concentration of methionine in the medium reversed or attenuated the inhibition achieved at lower levels. An inverse relationship existed between the degree of methylation and the extent of stimulation of the oxidative burst, measured as superoxide anion (O-2) release. Stimulated monocytes oxidized methionine to methionine sulfoxide (which cannot act as a methyl-donor), and this was dependent on activation of the respiratory burst. Elimination of the accumulated methionine sulfoxide by replacement of the medium or by prevention of extracellular methionine oxidation by catalase did not effectively restore the normal level of methylation in stimulated cells, and the reduced methylation was not primarily related to a defective methionine uptake by stimulated monocytes. These data suggest that intracellular events related to activation of the respiratory burst are responsible for the decreased lipid methylation in stimulated cells, possibly by their leading to intracellular formation of methionine sulfoxide and by their limiting the availability of methyl-donor. This mechanism may be of potential relevance for the expression of biological functions where methionine-dependent reactions are involved.


Subject(s)
Lipids/blood , Methionine/blood , Monocytes/metabolism , Oxygen Consumption , Phospholipids/blood , Calcimycin/pharmacology , Catalase/pharmacology , Cell Movement/drug effects , Cytochrome c Group/pharmacology , Humans , Kinetics , Methylation , Monocytes/drug effects , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Superoxide Dismutase/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Tritium
15.
Mol Cell Biol ; 18(12): 7565-74, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9819442

ABSTRACT

Although much is known about the multiple mechanisms which induce apoptosis, comparatively little is understood concerning the execution phase of apoptosis and the mechanism(s) of cell killing. Several reports have demonstrated that cellular translation is shut off during apoptosis; however, details of the mechanism of translation inhibition are lacking. Translation initiation factor 4G (eIF4G) is a crucial protein required for binding cellular mRNA to ribosomes and is known to be cleaved as the central part of the mechanism of host translation shutoff exerted by several animal viruses. Treatment of HeLa cells with the apoptosis inducers cisplatin and etoposide resulted in cleavage of eIF4G, and the extent of its cleavage correlated with the onset and extent of observed inhibition of cellular translation. The eIF4G-specific cleavage activity could be measured in cell lysates in vitro and was inhibited by the caspase inhibitor Ac-DEVD-CHO at nanomolar concentrations. A combination of in vivo and in vitro inhibitor studies suggest the involvement of one or more caspases in the activation and execution of eIF4G cleavage. Furthermore recombinant human caspase 3 was expressed in bacteria, and when incubated with HeLa cell lysates, was shown to produce the same eIF4G cleavage products as those observed in apoptotic cells. In addition, purified caspase 3 caused cleavage of purified eIF4G, demonstrating that eIF4G could serve as a substrate for caspase 3. Taken together, these data suggest that cellular translation is specifically inhibited during apoptosis by a mechanism involving cleavage of eIF4G, an event dependent on caspase activity.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Peptide Initiation Factors/metabolism , Protein Biosynthesis/genetics , Amino Acid Chloromethyl Ketones/pharmacology , Caspase 3 , Cisplatin/pharmacology , Cytochrome c Group/pharmacology , Deoxyadenine Nucleotides/pharmacology , Etoposide/pharmacology , Eukaryotic Initiation Factor-4G , HeLa Cells , Humans , Kinetics , Oligopeptides/pharmacology , Protease Inhibitors/pharmacology , Recombinant Proteins/metabolism
16.
Mol Cell Biol ; 19(8): 5800-10, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10409766

ABSTRACT

Growth factors signaling through the phosphoinositide 3-kinase/Akt pathway promote cell survival. The mechanism by which the serine/threonine kinase Akt prevents cell death remains unclear. We have previously shown that Akt inhibits the activity of DEVD-targeted caspases without changing the steady-state levels of Bcl-2 and Bcl-x(L). Here we show that Akt inhibits apoptosis and the processing of procaspases to their active forms by delaying mitochondrial changes in a caspase-independent manner. Akt activation is sufficient to inhibit the release of cytochrome c from mitochondria and the alterations in the inner mitochondrial membrane potential. However, Akt cannot inhibit apoptosis induced by microinjection of cytochrome c. We also demonstrated that Akt inhibits apoptosis and cytochrome c release induced by several proapoptotic Bcl-2 family members. Taken together, our results show that Akt promotes cell survival by intervening in the apoptosis cascade before cytochrome c release and caspase activation via a mechanism that is distinct from Bad phosphorylation.


Subject(s)
Apoptosis , Cytochrome c Group/metabolism , Mitochondria/metabolism , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology , Animals , Apoptosis/radiation effects , Apoptosis Regulatory Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspases/physiology , Cytochrome c Group/administration & dosage , Cytochrome c Group/pharmacology , Enzyme Activation/radiation effects , Intracellular Membranes/ultrastructure , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Microinjections , Mitochondrial Proteins , Models, Biological , Phosphorylation , Protein Biosynthesis , Protein Processing, Post-Translational , Proteins/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Rats , Ultraviolet Rays , bcl-2 Homologous Antagonist-Killer Protein , bcl-2-Associated X Protein , bcl-Associated Death Protein
17.
Cancer Res ; 36(2 Pt 1): 291-7, 1976 Feb.
Article in English | MEDLINE | ID: mdl-177191

ABSTRACT

Previously, we reported that the rate of metabolism of methyl sterol intermediates of cholesterol biosynthesis by broken-cell preparations of Morriss hepatoma 7777 is very slow, whereas the intact tumors are known to synthesize cholesterol quite efficiently. Active preparations have now been obtained by substitution of pyrophosphate for phosphate buffer. Although substitution of pyrophosphate buffer markedly enhances microsomal methyl sterol demethylation rates 3- to 4-fold in hepatoma 7777, other microsomal enzymes and electron carriers in either liver or a more slowly growing hepatoma appear to be unaffected by pyrophosphate. Several properties of the active microsomal methyl sterol demethylase have now been compared for control rat liver, host liver, tumor 7777, and tumor 5123C. Conditions necessary for the assay of initial velocities of enzymic reactions in the tumor microsomes have been established with respect to the amount of protein, time-course, concentrations of cofactors and substrate, pH, and other variables. The K'm and the responses to the variables studied above are very similar for methyl sterol demethylase of microsomes isolated from control liver, host liver, tumor 5123C, and tumor 7777. The multienzymic demethylase in the various preparations has been found to be inhibited similarly by in vitro additions of cyanide, cytochrome c, and bile salts. Thus, the enzymes of the microsomal-bound 4-methyl sterol demethylase of cholesterol biosynthesis appear to be very similar in liver and these 2 Morris hepatomas. When xenobiotic inducers of microsomal oxidases, such as phenobarbital and methylcholanthrene, are administered to normal and tumor-bearing rats, elevated rates of methyl sterol demethylation are observed with isolated liver microsomes obtained from both normal and tumor-bearing rats. Similar increases are not observed in the tumors. Furthermore, daily administration of an intestinal bile acid sequestrant elevates hepatic methyl sterol demethylase, but statistically significant changes were not observed in tumors 7777 and 5123C. Since the enzymes of methyl sterol demethylase appear to be grossly similar in liver and these hepatomas, regulation of the activity of the multienzymic system contained in the tumors may be altered. On the other hand, these agents in vivo simply may not affect liver and the hepatomas similarly, due to a lack of uptake of the foreign substances by the tumor that has been transplanted to the thighs.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , Steroid Hydroxylases/metabolism , Animals , Buffers , Cholesterol/biosynthesis , Cholestyramine Resin/pharmacology , Cyanides/pharmacology , Cytochrome c Group/pharmacology , Diphosphates , Edetic Acid , Liver/enzymology , Male , Methylcholanthrene/pharmacology , Microsomes, Liver/enzymology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Neoplasms, Experimental/enzymology , Phenobarbital/pharmacology , Phosphates , Rats , Sterols
18.
J Neurosci ; 22(18): 8018-27, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12223555

ABSTRACT

Sympathetic neuronal apoptosis after nerve growth factor (NGF) deprivation requires the activation of two events: a protein synthesis-dependent, Bax-dependent release of mitochondrial cytochrome c and a protein synthesis-independent, Bax-independent development of competence. Unlike in most cells, cytosolic cytochrome c is not sufficient to induce cell death in NGF-maintained sympathetic neurons but can do so in neurons that have developed competence. We report that cytosolic cytochrome c-induced apoptosis in competent sympathetic neurons is completely dependent on caspase-9. In addition, the neuroprotective agents KCl and chlorophenylthio-cAMP are potent inhibitors of the development-of-competence pathway in NGF-deprived sympathetic neurons. We also find that the development of competence is reversible. Readdition of NGF reverses competence, and neurons can regain their resistance to cytosolic cytochrome c. Importantly, we examined the mechanism of development of competence and report that the inability of cytochrome c to activate caspases in NGF-maintained sympathetic neurons can be overcome with exogenous Smac that inhibits the inhibitor of apoptosis (IAP) family of proteins. Microinjection of cytochrome c and Smac, but neither alone, induces rapid cell death in NGF-maintained neurons. These data suggest that development of competence may be the result of the loss of the function of one or more members of the IAP family of caspase inhibitors that is needed before cytochrome c can activate caspases and induce cell death in neurons.


Subject(s)
Apoptosis/physiology , Carrier Proteins/pharmacology , Caspases/metabolism , Mitochondrial Proteins/pharmacology , Neurons/drug effects , Proteins , Proto-Oncogene Proteins c-bcl-2 , Sympathetic Nervous System/drug effects , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Caspase 9 , Caspases/deficiency , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cytochrome c Group/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/metabolism , Inhibitor of Apoptosis Proteins , Insect Proteins/antagonists & inhibitors , Mice , Mice, Inbred ICR , Mice, Knockout , Microinjections , Nerve Growth Factor/pharmacology , Neurons/cytology , Neurons/metabolism , Phosphoinositide-3 Kinase Inhibitors , Potassium Chloride/pharmacology , Protein Kinase C/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Signal Transduction/drug effects , Staurosporine/pharmacology , Sympathetic Nervous System/cytology , Sympathetic Nervous System/metabolism , bcl-2-Associated X Protein
19.
Biochim Biophys Acta ; 1142(1-2): 194-202, 1993 Apr 05.
Article in English | MEDLINE | ID: mdl-8384490

ABSTRACT

We have investigated the motional dynamics of cytochrome c in the intact, functional rat liver mitochondrion. To do this, functional, FITC-cytochrome c (fluorescein isothiocyanate monoderivatized cytochrome c) was incorporated into the intermembrane space (IMS) of intact mitochondria through encapsulation of cytochrome c into asolectin liposomes followed by low pH-induced fusion of the liposomes with the outer membranes of the mitochondria. A cytochrome c controlled enrichment of between 15%-50% (1800-7200 molecules incorporated per mitochondrion) was obtained. All cytochrome c incorporated, regardless of the quantity, participated in the function of electron transport, indicative of a functional, independent random diffusant. Resonance energy transfer was determined from the IMS-entrapped functional FITC-cytochrome c to octadecylrhodamine B incorporated into the mitochondrial membranes. Resonance energy transfer from FITC-cytochrome c to octadecylrhodamine B in isolated inner or outer mitochondrial membranes (IMM and OMM, respectively) was also measured. We found substantial differences in the effects of ionic strength (I) on the proximity of cytochrome c to isolated IMM and OMM. Interactions with isolated IMM were very dynamic, i.e., very I-dependent, and cytochrome c binding to IMM was significant only at very low I. I-dependent interactions of cytochrome c with isolated OMM were less I-dependent than those for the IMM. However, FITC-cytochrome c was essentially released from IMM and OMM at physiological I. The proximity of FITC-cytochrome c to each mitochondrial membrane after its incorporation into the IMS of intact mitochondria in the condensed configuration was estimated at different external, bulk I using: (a) resonance energy transfer from IMS-entrapped FITC-cytochrome c to octadecylrhodamine B-label evenly distributed in both mitochondrial membranes; and (b) resonance energy transfer from IMS-entrapped FITC-cytochrome c to octadecylrhodamine B-label concentrated in the OMM. Resonance energy transfer showed that the average distance between cytochrome c and the two IMS-membrane surfaces increased with increasing IMS-I, approaching a maximal measurable distance of 85 A at 150 mM I. This result is consistent with a dissociation of FITC-cytochrome c and both membranes of intact mitochondria at physiological I, i.e., when the activity of cytochrome c in electron transport is highest. Our findings reveal a primarily three-dimensional diffusion mode for IMS-cytochrome c during its function in electron transport in intact mitochondria at physiological I, and offer further evidence that mitochondrial electron transport is a process driven by random collisions between its independently diffusing electron transferring, redox components.


Subject(s)
Cytochrome c Group/metabolism , Mitochondria, Liver/enzymology , Animals , Cytochrome c Group/pharmacology , Electron Transport , Fluorescein-5-isothiocyanate , Hydrogen-Ion Concentration , Intracellular Membranes/metabolism , Liposomes , Osmolar Concentration , Rats , Rats, Sprague-Dawley
20.
Biochim Biophys Acta ; 610(2): 229-34, 1980 Dec 11.
Article in English | MEDLINE | ID: mdl-6260148

ABSTRACT

Superoxide anion was photogenerated upon illumination of nucleic acids with fluorescent light in a solution containing phosphate buffer, pH 7.8 and riboflavin. DNA was a better reducing substrate for this reaction than was RNA. A similar riboflavin-sensitized photoreaction caused single- and double-strand scissions of supercoiled PM2 DNA as detected by electrophoresis in agarose gels. None of specific scavengers or quenchers for superoxide anion and other active oxygen species prevented the DNA strand breaks. However, among the flavin triplet-state quenchers, potassium iodide, butylated hydroxyanisole, and ferricytochrome c protected the supercoiled DNA from photodegradation; butylated hydroxytoluene, alpha-tocopherol, tyrosine and hemoglobin did not have any protective effect. These results indicate that triplet-state riboflavin or a derivative formed from it participate directly in the observed riboflavin-sensitized DNA photodegradation and that active oxygen species are not directly involved.


Subject(s)
DNA/radiation effects , Light , Potassium Iodide/pharmacology , Riboflavin/pharmacology , Butylated Hydroxyanisole/pharmacology , Cytochrome c Group/pharmacology , DNA, Superhelical/radiation effects , DNA, Viral/radiation effects , Fluorescence , Photochemistry , Superoxides
SELECTION OF CITATIONS
SEARCH DETAIL