Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
Add more filters










Publication year range
1.
Cell Biol Toxicol ; 19(3): 135-43, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12945742

ABSTRACT

5-Lipoxygenase activating protein (FLAP) functions as a facilitator of 5-lipoxygenase (5-LOX) activity. However, on the basis of the induction of apoptosis by the FLAP inhibitor MK886 in cells lacking 5-LOX, it is possible that this fatty acid-binding protein has other activities. This study was designed to examine potential roles of FLAP in apoptosis and cell proliferation. Overexpression of FLAP protein (2.2-fold) was achieved by stable transfection of IL-3-dependent murine prolymphoid progenitor cells (FL5.12) with a construct expressing the cDNA under a CMV promoter. The overexpressed protein was localized to nuclear membranes as with endogenous FLAP. The initial growth rate of FLAP-transfected cells was greater than that of control cells. After 48 h, when cell density had increased, the growth rate of FLAP-transfected cells declined substantially and there and there was a decrease in viability relative to control transfected cells. The FLAP-transfected cells were also more susceptible to withdrawal of IL-3 than were control cells. There was, however, no difference between FLAP and control cells in their susceptibility to MK886, NDGA, or etoposide during the log growth phase. Overexpression of FLAP did not alter Bcl-xL protein expression, but did decrease Bax protein and somewhat increased COX-1 and COX-2 mRNA levels. The failure of increased FLAP to alter susceptibility to MK886 provides further support to the concept that this agent induces apoptosis by mechanisms unrelated to FLAP. The data also suggest that FLAP can affect cell proliferation.


Subject(s)
Apoptosis/physiology , Arachidonate 5-Lipoxygenase/metabolism , B-Lymphocytes/metabolism , Carrier Proteins/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2 , 5-Lipoxygenase-Activating Proteins , Animals , Apoptosis/drug effects , B-Lymphocytes/drug effects , Carrier Proteins/genetics , Cell Count , Cell Division/physiology , Cell Survival/physiology , Cells, Cultured , Cyclooxygenase 1 , Cyclooxygenase 2 , Dose-Response Relationship, Drug , Etoposide/pharmacology , Indoles/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , Lipoxygenase Inhibitors/pharmacology , Masoprocol/pharmacology , Membrane Proteins/genetics , Mice , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Transfection , bcl-2-Associated X Protein
2.
Toxicol Lett ; 143(3): 241-5, 2003 Aug 28.
Article in English | MEDLINE | ID: mdl-12849684
3.
Toxicol Appl Pharmacol ; 174(3): 273-81, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11485388

ABSTRACT

Apoptosis induced in the IL3-dependent murine pro-B lymphocytic (FL5.12) cell line by the 5-lipoxygenase activating protein inhibitor MK886 is accompanied by the rapid loss of the anti-apoptotic bcl-x(L) and bcl-2, but not the proapoptotic bax proteins (Datta et al., J. Biol. Chem. 273, 28163-28169, 1998). Since several reports indicate important roles for noncaspase proteases in apoptosis, the participation of lysosomes, as well as serine, cysteine, or aspartic acid proteases, in the effects of MK886 were investigated. Consistent with the involvement of various proteases, lysosomal degranulation was evident, as observed by a decrease in acridine orange fluorescence at 2 h and an increase in cytosolic beta-hexosaminidase activity at 4 h after treating FL5.12 cells with 10 microM MK886. The disappearance of bcl-x(L) from FL5.12 cells upon MK886 treatment was prevented in a dose-dependent manner by pretreatment with leupeptin, pepstatin, phenylmethylsulfonyl fluoride, or the broad-spectrum caspase inhibitor Boc-D-FMK. Each of the noncaspase protease inhibitors partially inhibited MK886-induced apoptosis as measured by phosphatidylserine externalization and DNA fragmentation. The noncaspase inhibitors also blocked about half of the increase in caspase-3-like activity. Boc-D-FMK completely inhibited this enzyme and prevented apoptosis. None of the inhibitors were able to directly inhibit activated caspase-3 in cell lysates, suggesting their effects were upstream of caspase activation. These observations suggest the involvement of various proteases, possibly originating from lysosomes, upstream of active caspase-3, in the loss of bcl-x(L) protein and in the signaling pathway of MK886-induced apoptosis in FL5.12 cells. This pathway may be unique to MK886 since these same protease inhibitors had only minimal effects on etoposide-induced apoptosis and the accompanying moderate loss of bcl-x(L) in FL5.12 cells.


Subject(s)
Apoptosis , Indoles/pharmacology , Lipoxygenase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/analysis , Acridine Orange , Animals , Cell Line , Cell Survival/drug effects , Lysosomes/drug effects , Mice , Microscopy, Confocal , Peptide Hydrolases/metabolism , Protease Inhibitors/pharmacology , bcl-X Protein
4.
Toxicol Appl Pharmacol ; 175(1): 10-8, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11509022

ABSTRACT

The current studies were designed to examine the mechanisms of acute effects of ethanol on cerebellar granule neurons (CGNs) during neurodevelopment, with specific reference to activator protein-1 (AP-1). CGNs, isolated from 3-day-old Sprague-Dawley rats and cultured for 3 days, were exposed to 0, 22.5, and 100 mM ethanol for 1 h. Gel shift assays performed on the nuclear protein extracts showed increased AP-1 and heat shock factor-1 (HSF-1) transcriptional activation in response to ethanol. Western blots and RT-PCR showed increased c-JUN and phosphorylated c-JUN (serine 73) protein, as well as c-jun mRNA. Ethanol paradoxically decreased the activity of stress-activated protein kinase-1 (SAPK-1) while increasing p44 and p42 mitogen-activated protein kinase (MAPK) activity. The protein synthesis-inhibiting and SAPK-1 activity-inducing antibiotic, anisomycin (30 and 500 microM) decreased AP-1 transcriptional activation to 47 and 23% of control values, respectively. The anisomycin effect was enhanced in the presence of 100 mM ethanol. Similarly, cycloheximide decreased ethanol-induced AP-1 transcriptional activation. Pretreatment with the MAPK kinase (MEK) pathway inhibitor PD98059 resulted in decreases in both ethanol-induced and control AP-1 DNA binding. Thus this acute ethanol-induced increased AP-1 transcriptional activation requires protein synthesis and involves MEK-independent increased MAPK phosphorylation, on the one hand, and decreased SAPK-1 activity on the other. The ethanol effect is thus ascribed to the activities of alternate kinase pathways and/or the inhibition of (a) protein phosphatase(s). Exposure of CGNs to ethanol for 24 h resulted in decreased AP-1 DNA binding, an observation that could have consequences for overall neuronal function under chronic exposure conditions.


Subject(s)
Central Nervous System Depressants/pharmacology , Cerebellum/drug effects , Ethanol/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Transcription Factor AP-1/biosynthesis , Animals , Cerebellum/enzymology , DNA-Binding Proteins/biosynthesis , Heat Shock Transcription Factors , Mitogen-Activated Protein Kinase 8 , Neurons/enzymology , Rats , Rats, Sprague-Dawley , Transcription Factors
5.
Biochem J ; 356(Pt 3): 899-906, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11389700

ABSTRACT

Although MK886 was originally identified as an inhibitor of 5-lipoxygenase activating protein (FLAP), recent data demonstrate that this activity does not underlie its ability to induce apoptosis [Datta, Biswal and Kehrer (1999) Biochem. J. 340, 371--375]. Since FLAP is a fatty-acid binding protein, it is conceivable that MK886 may affect other such proteins. A family of nuclear receptors that are activated by fatty acids and their metabolites, the peroxisome-proliferator-activated receptors (PPARs), have been implicated in apoptosis and may represent a target for MK886. The ability of MK886 to inhibit PPAR-alpha, -beta and -gamma activity was assessed using reporter assay systems (peroxisome-proliferator response element--luciferase). Using a transient transfection system in monkey kidney fibroblast CV-1 cells, mouse keratinocyte 308 cells and human lung adenocarcinoma A549 cells, 10--20 microM MK886 inhibited Wy14,643 activation of PPAR alpha by approximately 80%. Similar inhibition of PPAR alpha by MK886 was observed with a stable transfection reporter system in CV-1 cells. Only minimal inhibitory effects were seen on PPAR beta and PPAR gamma. MK886 inhibited PPAR alpha by a non-competitive mechanism as shown by its effects on the binding of arachidonic acid to PPAR alpha protein, and a dose-response study using a transient transfection reporter assay in COS-1 cells. An assay assessing PPAR ligand-receptor interactions showed that MK886 prevents the conformational change necessary for active-complex formation. The expression of keratin-1, a protein encoded by a PPAR alpha-responsive gene, was reduced by MK886 in a culture of mouse primary keratinocytes, suggesting that PPAR inhibition has functional consequences in normal cells. Although Jurkat cells express all PPAR isoforms, various PPAR alpha and PPAR gamma agonists were unable to prevent MK886-induced apoptosis. This is consistent with MK886 functioning as a non-competitive inhibitor of PPAR alpha, but may also indicate that PPAR alpha is not directly involved in MK886-induced apoptosis. Although numerous PPAR activators have been identified, the results show that MK886 can inhibit PPAR alpha, making it the first compound identified to have such an effect.


Subject(s)
Indoles/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Apoptosis/drug effects , COS Cells , Gene Expression Regulation/physiology , Humans , Peroxisome Proliferators/pharmacology , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Tumor Cells, Cultured
6.
Toxicology ; 160(1-3): 97-103, 2001 Mar 07.
Article in English | MEDLINE | ID: mdl-11246129

ABSTRACT

The expression of 5-lipoxygenase activating protein (FLAP) in murine hematopoietic FL5.12 cells that are transfected to overexpress bcl-x(L) is less than in control cells. In addition, the withdrawal of IL-3 from the bcl-x(L) overexpressing cells, but not control cells, leads to the rapid loss of FLAP even though these cells, in contrast to control cells, do not undergo apoptosis (Datta et al., J. Biol. Chem. 273, 28163-28169 [1998]). The mechanism(s) underlying these observations is not known. Basal FLAP mRNA levels were actually 2.8-fold higher in bcl-x(L) than control cells indicating that this difference does not have a transcription basis. In addition, an examination of FLAP mRNA levels in response to withdrawal of IL-3 revealed a 2-3-fold increase after 4 and 8 h relative to time-matched samples in both control and bcl-x(L) overexpressing cells. This further indicates that the decrease in FLAP levels in bcl-x(L) overexpressing cells is not related to transcription and suggests an attempt at compensation perhaps in response to increased FLAP degradation/turnover. A proteolytic mechanism was explored by examining the effect of the general caspase inhibitor Boc-D-FMK, and the non-caspase protease inhibitors phenylmethylsulfonyl fluoride (PMSF), pepstatin and leupeptin, on the loss of FLAP in bcl-x(L) overexpressing cells subsequent to IL-3 withdrawal. All inhibitors provided some protection from the loss of FLAP, with PMSF being the most effective, actually increasing FLAP levels above those seen in untreated cells. Given the absence of apoptosis in bcl-x(L) cells, it appears that protease activation is an effect that can accompany a variety of cellular perturbations. The functional consequences of a loss of FLAP in growth-factor deprived cells overexpressing bcl-x(L) is not known. However, these data continue to suggest some link between bcl-x(L) and FLAP.


Subject(s)
Carrier Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Interleukin-3/pharmacology , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , 5-Lipoxygenase-Activating Proteins , Blotting, Northern , Blotting, Western , Caspase Inhibitors , Cells, Cultured , Cloning, Molecular , Enzyme Inhibitors/pharmacology , Leupeptins/pharmacology , Pepstatins/pharmacology , Phenylmethylsulfonyl Fluoride/pharmacology , RNA, Messenger/metabolism , Transfection , bcl-X Protein
7.
Toxicology ; 157(1-2): 67-76, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11164975

ABSTRACT

The number of digital tools available for use by researchers continues to rapidly increase as does the quantity of information available on the web. However, judging the reliability, quality and timeliness of this information is not always easy. One source of information about the profession of toxicology comes from the web sites of the various professional organizations that exist in this field. Given the broad nature of toxicology, there are a large number of such organizations. Some serve broader segments of the profession, while others have a more narrow focus. The web sites they have created are similarly variable, although there are some consistencies in the type of information that is provided. These include mission statements, information on joining the organization, and details on their annual meetings. There is also a consistent decision by these sites not to provide toxicologic information per se, although links to sites that do contain such information are occasionally provided. Overall, the information and tools provided by professional toxicology societies around the world appears to be similar in both quantity and quality to other scientific organizations. It is also apparent that this information is rapidly evolving and will likely be much different within the next 5 years.


Subject(s)
Internet , Societies , Toxicology , Certification , Humans
8.
Toxicology ; 154(1-3): 45-53, 2000 Nov 23.
Article in English | MEDLINE | ID: mdl-11118669

ABSTRACT

Fludarabine is used to treat chronic lymphocytic leukemia. Both in vitro and in vivo studies have indicated that apoptosis is an important mode of fludarabine-induced cell death. However, the apoptotic pathways activated are not known. The effects of apoptotic doses of fludarabine on sphingomyelin, ceramide and the production of reactive oxygen species were investigated in the chronic B-cell leukemia lines WSU and JVM-2. Apoptosis, as assessed by an increase in phosphatidylserine externalization, internucleosomal DNA fragmentation and caspase-3-like activity, was evident by 18 h after fludarabine in both cell lines. The general caspase inhibitor t-butoxycarbonyl-Asp(OMe)-fluoromethyl ketone (OMe, methyl ester) significantly inhibited apoptosis supporting a role for caspases in fludarabine-induced cell death. A 2.5- to threefold elevation in ceramide levels was observed 6 h after fludarabine treatment. Concomitantly, a decrease in sphingomyelin levels was observed. Fumonisin B1 (an inhibitor of ceramide synthase) pretreatment significantly prevented fludarabine-induced ceramide generation and apoptosis. Conversely, C6-ceramide induced apoptosis in both cell lines. No effect of fludarabine on indices of oxidative stress (dichlorofluorescin oxidation and glutathione disulfide formation) were detected, although partial protection from apoptosis, and prevention of ceramide generation and caspase-3 activation, were achieved with N-acetylcysteine. These findings are consistent with the involvement of caspases and ceramide in fludarabine-induced apoptosis in WSU and JVM-2 cells. Oxidative stress does not appear to be induced by fludarabine, although the protective effects of N-acetylcysteine suggest that thiol redox balance may play a role in the apoptotic pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Ceramides/biosynthesis , Fumonisins , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Sphingomyelins/biosynthesis , Vidarabine/analogs & derivatives , Acetylcysteine/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carboxylic Acids/chemistry , Caspase 3 , Caspase Inhibitors , Caspases/analysis , Ceramides/analysis , Chromatography, Thin Layer , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glutathione/analysis , Humans , Image Processing, Computer-Assisted , Phosphatidylserines/physiology , Sphingomyelins/analysis , Tumor Cells, Cultured/drug effects , Vidarabine/pharmacology , Vidarabine/therapeutic use
10.
Toxicology ; 149(1): 43-50, 2000 Aug 14.
Article in English | MEDLINE | ID: mdl-10963860

ABSTRACT

The concept that the highly reactive hydroxyl radical (HO) could be generated from an interaction between superoxide (O(2)(-)) and hydrogen peroxide (H(2)O(2)) was proposed (with Joseph Weiss) in Professor Haber's final paper published in 1934. Until it was recognized that free radicals are produced in biological systems, this finding seemed to have no relevance to biology. However, following the discovery that O(2)(-) was a normal cellular metabolite, it was quickly recognized that the Haber-Weiss reaction (O(2)(-)+H(2)O(2) -->HO+O(2)+HO(-)) might provide a means to generate more toxic radicals. Although the basic reaction has a second order rate constant of zero in aqueous solution and thus cannot occur in biological systems, the ability of iron salts to serve as catalysts was discussed by these authors. Because transition metal ions, particularly iron, are present at low levels in biological systems, this pathway (commonly referred to as the iron-catalyzed Haber-Weiss reaction) has been widely postulated to account for the in vivo generation of the highly reactive HO. Recent data documenting the importance of redox regulation of various cellular signaling pathways makes it clear that free radicals are essential for normal cellular function. However, this also makes it obvious that disruptions of free radical production or defenses at many different levels can lead to adverse effects on cells. While the generation of HO, which is by far the most reactive oxygen species, is generally indicative of an overtly toxic event, it is through studies at this level that we have reached a better understanding of free radicals as both signaling molecules and toxic species.


Subject(s)
Hydrogen Peroxide/toxicity , Hydroxyl Radical/toxicity , Superoxides/toxicity , Animals , Humans , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Hydroxyl Radical/chemistry , Hydroxyl Radical/metabolism , Oxidative Stress/physiology , Superoxides/chemistry , Superoxides/metabolism
11.
Toxicol Sci ; 57(1): 6-15, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10966506

ABSTRACT

Acrolein is a highly electrophilic alpha,beta-unsaturated aldehyde to which humans are exposed in a variety of environmental situations, particularly as a component of smoke. In addition, as a metabolite of cyclophosphamide, acrolein is a major factor in the toxicity and perhaps the therapeutic activity of this important anticancer agent. The exposures to acrolein that are attained in vivo in most situations are quite low and the effects may differ from those seen at acutely toxic doses. At low doses, acrolein inhibits cell proliferation without causing cell death and may enhance apoptosis from secondary toxins, while at higher doses oncosis ensues. Although the acute toxicology of acrolein has been extensively investigated, both in animals and cultured cells, little information exists on the molecular effects of this reactive aldehyde. It is possible that the acrolein-mediated decrease in cell proliferation is caused by effecting changes in the expression of one or more growth- or stress-related genes or transcription factors secondary to a reduction in glutathione (GSH), which is rapidly depleted following acrolein treatment. It is apparent that the activation of the transcription factors nuclear factor kappa B (NF-kappa B) and activator protein 1 (AP-1) can be inhibited by acrolein. The purpose of this review is to assess the literature currently available on the molecular effects of acrolein, to discuss the relationship between effects on glutathione with those on various genes, and to present some new data showing that acrolein actively stimulates genes associated with the electrophile response element.


Subject(s)
Acrolein/toxicity , Carcinogens/toxicity , Environmental Pollutants/toxicity , Animals , Cell Division/drug effects , Humans , Transcription Factors/drug effects
12.
Toxicol Sci ; 53(1): 77-83, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10653524

ABSTRACT

Nordihydroguaiaretic acid (NDGA) induces apoptosis in a variety of cell lines. The mechanism(s) of this effect is not known, although the focus has been on the ability of NDGA to inhibit lipoxygenase (LOX) activities. In the present study, NDGA-induced apoptosis was studied in a murine hematopoietic cell line, FL5.12. Although this cell line lacks detectable LOX protein or activities, NDGA (10 microM) was able to induce apoptosis. There was a massive loss of mitochondrial membrane potential by 4 h after the addition of NDGA, suggesting that this organelle might be targeted by NDGA. A pro-oxidant NDGA effect has been suggested as playing a role in apoptosis. This was supported by the findings that glutathione disulfide levels were increased by 4 h following treatment with 10 microM NDGA, that pretreatment with N-acetylcysteine completely blocked the NDGA-induced loss of membrane potential and apoptosis, and that lipid peroxidation was enhanced in cells treated with NDGA. However, no evidence of increased levels of reactive oxygen could be seen in NDGA-treated cells loaded with dichlorofluorescin diacetate or dihydrorhodamine and analyzed by flow cytometry. Bcl-X(L) protein levels were unaffected by NDGA treatment. Caspase-3 was rapidly activated with a peak at 8 h after FL5.12 cells were treated with NDGA. Ac-DEVD-CHO (25 microM) and boc-asp-FMK (20 microM) both inhibited caspase-3 enzyme activity by 97% 8 h after NDGA treatment. Boc-asp-FMK, a more general caspase inhibitor, delayed NDGA-induced apoptosis while Ac-DEVD-CHO, a more specific inhibitor of caspase-3, had no effect. These results suggest that NDGA-induced apoptosis happens through reactions that depolarize mitochondria, oxidize glutathione and lipids, but do not generate significant amounts of free reactive oxygen species.


Subject(s)
Apoptosis/drug effects , B-Lymphocytes/drug effects , Glutathione/metabolism , Lipoxygenase Inhibitors/pharmacology , Lipoxygenase/deficiency , Masoprocol/pharmacology , Mitochondria/metabolism , Acetylcysteine/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , B-Lymphocytes/enzymology , Blotting, Western , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cell Line , Enzyme-Linked Immunosorbent Assay , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Oligopeptides/pharmacology , Oxidation-Reduction , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein
13.
Biochem J ; 344 Pt 2: 477-85, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10567231

ABSTRACT

Recent evidence supports a role for heat-shock protein 70 (hsp70) and the 26 S proteasome in regulating apoptosis, although the precise nature of their involvement is not known. In the present study, control and Bcl-x(L)-overexpressing, interleukin-3-dependent FL5.12 cell lines were treated with the proteasome inhibitor N-benzoyloxycarbonyl (Z)-Leu-Leu-leucinal (MG132). Basal proteasome activity appeared to be approximately 30% lower in bcl-x(L) cells compared with control cells using a substrate for the chymotrypsin-like activity. However, no difference in proteasome activity was detected using substrates for the trypsin-like or peptidylglutamyl peptide-hydrolysing activities. In addition, protein levels of the 20 S proteasome beta-subunit, as determined by Western blot analyses, were similar in control and bcl-x(L) cells, leading to the conclusion that proteasome activities were the same in these two cell lines. At 24 h after treatment with 500 nM MG132, apoptosis in bcl-x(L) cells (22%) was less than that observed in control cells (34%). Concomitantly, caspase activity in control cells, as assessed by N-acetyl-l-aspartyl-l-glutamyl-l-valyl-l-aspartyl-7-amino-4-methylcou marin (Ac-DEVD-AMC), was twice that observed in bcl-x(L) cells. By 48 h after MG132 treatment, apoptosis and caspase activity in bcl-x(L) cells were similar to those observed in control cells at 24 h. Proteasome inhibition stimulated increases in hsp70 protein levels in control and bcl-x(L) cells by 12 h, although the maximal increases found in bcl-x(L) cells were less. Blocking this induction with hsp70 antisense oligonucleotides potentiated apoptosis after treatment with MG132. Inhibiting caspase activity with a broad-spectrum caspase inhibitor, t-butoxycarbonyl-Asp(OMe)-fluoromethyl ketone, prevented MG132-induced apoptosis. The more specific caspase-3 inhibitor, Ac-DEVD-aldehyde, afforded less protection, although both inhibitors completely inhibited Ac-DEVD-AMC cleavage. These data indicate that both hsp70 and Bcl-x(L) provide some protection against proteasome inhibitor-induced apoptosis.


Subject(s)
Apoptosis , Cysteine Endopeptidases/drug effects , Cysteine Proteinase Inhibitors/pharmacology , HSP70 Heat-Shock Proteins/genetics , Multienzyme Complexes/drug effects , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , B-Lymphocytes , Canavanine/pharmacology , Caspase Inhibitors , Caspases/metabolism , Drug Synergism , Enzyme Activation , Hematopoietic Stem Cells , Interleukin-3/metabolism , Leupeptins/pharmacology , Mice , Proteasome Endopeptidase Complex , bcl-X Protein
14.
Biochem J ; 340 ( Pt 2): 371-5, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10333477

ABSTRACT

The ability of various inhibitors of lipoxygenase (LOX) enzymes and 5-lipoxygenase-activating protein (FLAP) to induce apoptosis has implicated these pathways in the mechanism(s) of this form of cell death. Although FLAP plays an important role in 5-LOX activity, this protein is found at high levels in some cells lacking LOX, suggesting it might mediate other effects. Furthermore, the concentration of MK886, a FLAP inhibitor, required to induce apoptosis is approximately 100-fold more than that required to inhibit LOX, and this compound remains effective in cells lacking LOX. The present study examines the role of FLAP in MK886-induced apoptosis. MK886 induced apoptosis in WSU cells, a human chronic lymphocytic leukaemia cell line that lacks FLAP protein and mRNA, suggesting that this agent is acting independently of FLAP. This conclusion was further supported by the fact that a more specific FLAP inhibitor, MK591, induced only minimal apoptosis in FL5.12 cells, a murine prolymphoid cell line containing FLAP. The role of FLAP was examined more directly by decreasing its expression by more than 50% in FL5.12 cells treated with 10 microM of an antisense oligonucleotide for 48h. This change in FLAP was not accompanied by any increase in apoptosis. Furthermore, FLAP-depleted cells exhibited the same level of apoptosis 8 h after treatment with 10 microM MK886, as did control cells. The increased fluorescence seen in MK886-treated cells loaded with carboxydichlorofluorescein indicates that oxidative reactions are stimulated by this compound, possibly via the release of fatty acids from fatty acid-binding proteins and their subsequent oxidation.


Subject(s)
Apoptosis/drug effects , Carrier Proteins/antagonists & inhibitors , Indoles/pharmacology , Lipoxygenase Inhibitors/pharmacology , Membrane Proteins/antagonists & inhibitors , 5-Lipoxygenase-Activating Proteins , Acetylcysteine/pharmacology , Base Sequence , Carrier Proteins/metabolism , Humans , Indoles/antagonists & inhibitors , Membrane Proteins/metabolism , Oligonucleotides, Antisense/pharmacology , Quinolines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
15.
Cell Death Differ ; 6(1): 61-70, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10200549

ABSTRACT

The mechanism of cell death caused by cytokine deprivation remains largely unknown. FL5.12 cells (a murine prolymphocytic cell line), following interleukin-3 (IL-3) withdrawal, undergo a decrease in intracellular glutathione (GSH) that precedes the onset of apoptosis. In the present study, the induction of apoptosis following IL-3 withdrawal or GSH depletion with DL-buthionine-[S,R,]-sulfoximine (BSO) was examined. Both conditions caused time-dependent increases in phosphatidylserine externalization, acridine orange and ethidium bromide staining, decreases in mitochondrial membrane potential, processing and activation of caspase-3 and proteolysis of the endogenous caspase substrate poly(adenosine diphosphate ribose)polymerase (PARP). Apoptosis induced by IL-3 deprivation but not BSO also caused lamin B1 cleavage, suggesting activation of caspase-6. Despite a more profound depletion of GSH after BSO than withdrawal of IL-3, the extent of apoptosis was somewhat lower. Benzyloxycarbonyl-Val-Ala-Asp(OMe)fluoromethyl ketone (z-VAD.fmk) blocked this caspase activity and prevented cell death after BSO exposure but not after IL-3 deprivation. Following IL-3 withdrawal, the caspase inhibitors z-VAD.fmk and boc-asp(OMe)fluoromethylketone (boc-asp.fmk) prevented the cleavage and activation of caspase-3, the breakdown of lamin B1 and partially mitigated PARP degradation. However, the externalization of phosphatidylserine, the fall in mitochondrial membrane potential and subsequent apoptotic cell death still occurred. These results suggest that IL-3 withdrawal may mediate cell death by a mechanism independent of both caspase activation and the accompanying loss of GSH.


Subject(s)
Apoptosis , Caspases/metabolism , Glutathione/metabolism , Interleukin-3/metabolism , Lamin Type B , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Buthionine Sulfoximine/pharmacology , Caspase 3 , Cell Line , Coumarins/metabolism , Glutathione Disulfide/metabolism , Hematopoietic Stem Cells , Lamins , Membrane Potentials/drug effects , Mice , Mitochondria/drug effects , Nuclear Proteins/metabolism , Oligopeptides/metabolism , Phosphatidylserines/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , Protease Inhibitors/pharmacology , Proteins/metabolism
16.
J Biol Chem ; 274(14): 9200-6, 1999 Apr 02.
Article in English | MEDLINE | ID: mdl-10092592

ABSTRACT

Acrolein is a highly electrophilic alpha,beta-unsaturated aldehyde to which humans are exposed in various situations. In the present study, the effects of sublethal doses of acrolein on nuclear factor kappaB (NF-kappaB) activation in A549 human lung adenocarcinoma cells were investigated. Immediately following a 30-min exposure to 45 fmol of acrolein/cell, glutathione (GSH) and DNA synthesis and NF-kappaB binding were reduced by more than 80%. All parameters returned to normal or supranormal levels by 8 h post-treatment. Pretreatment with acrolein completely blocked 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced activation of NF-kappaB. Cells treated for 1 h with 1 mM diethyl maleate (DEM) showed a 34 and 53% decrease in GSH and DNA synthesis, respectively. DEM also reduced NF-kappaB activation by 64% at 2 h post-treatment, with recovery to within 22% of control at 8 h. Both acrolein and DEM decreased NF-kappaB function approximately 50% at 2 h after treatment with TPA, as shown by a secreted alkaline phosphatase reporter assay. GSH returned to control levels by 8 h after DEM treatment, but proliferation remained significantly depressed for 24 h. Interestingly, DEM caused a profound decrease in NF-kappaB binding, even at doses as low as 0.125 mM that had little effect on GSH. Neither acrolein nor DEM had any effect on the levels of phosphorylated or nonphosphorylated inhibitor kappaB-alpha (IkappaB-alpha). Furthermore, acrolein decreased NF-kappaB activation in cells depleted of IkappaB-alpha by TPA stimulation in the presence of cycloheximide, demonstrating that the decrease in NF-kappaB activation was not the result of increased binding by the inhibitory protein. This conclusion was further supported by the finding that acrolein modified NF-kappaB in the cytosol prior to chemical dissociation from IkappaB with detergent. Together, these data support the conclusion that the inhibition of NF-kappaB activation by acrolein and DEM is IkappaB-independent. The mechanism appears to be related to direct modification of thiol groups in the NF-kappaB subunits.


Subject(s)
Acrolein/pharmacology , Adenocarcinoma/metabolism , DNA-Binding Proteins/metabolism , I-kappa B Proteins , Lung Neoplasms/metabolism , NF-kappa B/metabolism , Cell Division/drug effects , Electrophoresis, Polyacrylamide Gel , Humans , Maleates/pharmacology , NF-KappaB Inhibitor alpha , Phosphorylation , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
17.
J Biol Chem ; 273(43): 28163-9, 1998 Oct 23.
Article in English | MEDLINE | ID: mdl-9774436

ABSTRACT

Inhibitors of 5-lipoxygenase-activating protein (FLAP) have been found to induce apoptosis. The current study examined the expression of FLAP and bcl family proteins and the induction of apoptosis in interleukin-3-dependent control and bcl-xL-overexpressing FL5.12 cell lines after treatment with MK886, a specific FLAP inhibitor. FL5.12 cells contained a substantial amount of FLAP protein and mRNA but surprisingly had no measurable 5-lipoxygenase protein or 5-, 12-, or 15-lipoxygenase activity. The basal level of FLAP protein in cells overexpressing bcl-xL was 70% less than in controls. FLAP disappeared 4 h after withdrawal of interleukin-3 in bcl-xL cells but not in control cells, which underwent apoptosis. A dose- and time-response study revealed that 5 nmol of MK886/10(6) cells was sufficient to induce apoptosis both in control and bcl-xL cells, respectively, but to different degrees. bcl-xL and bcl-2 proteins, but not bax or FLAP, were decreased by 4 h after 5 nmol of MK886/10(6) cells in both cell lines, although the higher levels of bcl-xL in overexpressors took longer to disappear. This early loss of bcl-xL and bcl-2 was not attributable to generalized proteolysis, as shown by Coomassie Blue staining and by the maintenance of bax. Caspase-3 was activated 2 h after MK886 treatment in control cells but not in bcl-xL cells. Inhibition of caspase-3 decreased MK886-induced apoptosis by 50% in control cells. Inhibition of this caspase after MK886 treatment was unable to prevent the loss of bcl-xL in control cells but did provide partial protection for the loss of the transfected form, but not the endogenous form, in overexpressing cells. These data indicate that MK886 induces extensive apoptosis that is partially caspase-3 dependent and may be related to a rapid loss of bcl-xL. Although caspase-3 inhibitors had no effect on the loss of bcl-xL, other caspases or protease systems may still be involved. The absence of 5-lipoxygenase in cells containing FLAP, the lower level of FLAP in bcl-xL cells, the apoptosis-inducing activity of MK886, and the rapid loss of bcl-xL and bcl-2 proteins after treatment with MK886 strongly indicate that FLAP has activities unrelated to lipoxygenase and suggest a possible functional or regulatory link between these proteins, which share similar subcellular localizations.


Subject(s)
Apoptosis , B-Lymphocytes/metabolism , Carrier Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , 5-Lipoxygenase-Activating Proteins , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Arachidonate 5-Lipoxygenase/metabolism , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Enzyme Activation , Indoles/pharmacology , Interleukin-3/pharmacology , Lipoxygenase Inhibitors/pharmacology , Masoprocol/pharmacology , Mice , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction , bcl-X Protein
18.
Int J Sport Nutr ; 8(2): 105-12, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9637190

ABSTRACT

The purpose of this study was to determine whether submaximal exercise significantly changes the concentration of vitamin E (alphaToc) in rat liver and skeletal muscle and to establish a time course for the return to basal levels. Male Sprague-Dawley rats, age 8 to 10 weeks, were randomly divided into sedentary control (Con) (n = 7) and exercise (n = 17) groups. Exercised animals ran 100 min on a motorized treadmill at approximately 70% VO2max for 3 consecutive days. They were then sacrificed immediately postexercise (0Post), 24 hr post (24Post), or 72 hr post (72Post). The gastrocnemius, red vastus lateralis (RV), white vastus lateralis (WV), and liver were excised and analyzed for alphaToc concentration by high-performance liquid chromotography utilizing electrochemical detection. We found that after 3 consecutive days of exercise, alphaToc was reduced in RV and WV at 0Post and 24Post but returned to control values by 72Post. Liver alphaToc content was not changed at 0Post but was significantly reduced at 24Post and 72Post. No significant changes in alphaToc were observed in the gastrocnemius in response to exercise. The data indicate that following an exercise-related decrease, skeletal muscle vitamin E concentration requires more than 24 hr to return to the preexercise concentration, and that the replenishment process may involve redistribution of vitamin E from liver to muscle.


Subject(s)
Liver/metabolism , Muscle, Skeletal/metabolism , Physical Exertion/physiology , Vitamin E/metabolism , Animals , Kinetics , Male , Rats , Rats, Sprague-Dawley
19.
Toxicology ; 125(1): 1-11, 1998 Jan 16.
Article in English | MEDLINE | ID: mdl-9585095

ABSTRACT

2,6-Dithiopurine (DTP) has been proposed as a possible chemopreventive agent because of its ability to react with electrophiles. Acrolein, an electrophilic metabolite of cyclophosphamide (CP) involved in the toxicities of this anticancer drug, can be scavenged by DTP. The present study examined the effect of DTP treatment on CP-mediated bladder and lung toxicity in male ICR mice. Mice fed a diet containing 4% DTP that were treated intraperitoneally (i.p.) with 350 mg/kg CP showed no significant bladder damage (measured as bladder blood content at 48 h) with respect to the group fed a control diet. DTP (50 and 100 mg/kg), given i.p. 0.5 and 7 h after the initial injection of CP, also prevented the bladder damage when compared with the group receiving CP alone. Surprisingly, although neither parenteral CP nor DTP alone caused any mortality at these doses, the combined treatment resulted in 67% mortality within 3 days. At 24 h after CP + DTP, blood urea nitrogen was elevated 6-fold and urine volumes decreased by 70%. Histopathological analyses revealed a diffuse myocardial degeneration and necrosis, severe granular degeneration in the liver, abundant cellularity and infiltrates in interalveolar spaces in the lung and swollen nephron epithelial cells with some necrosis. All mice survived treatment when the dose of CP was lowered to 250 and 25-75 mg/kg DTP was given i.p. 0.5 and 7 h after CP. These DTP regimens reduced the degree of CP-induced lung toxicity, measured by [3H]thymidine incorporation into lung DNA 7 days after CP, in a dose-dependent manner. DTP (75 mg/kg) also reduced CP-induced lung fibrosis estimated by lung hydroxyproline content 28 days after CP. Analyses of urine from mice given CP + DTP revealed large amounts of the metabolic product dithiouric acid, smaller amounts of the parent DTP and several smaller peaks. The major unique metabolite peak was collected and analyzed by mass spectrometry, but did not correspond to either acrolein-DTP or acrolein-dithiouric acid. Thus, either very small amounts of an acrolein adduct are generated, the adduct is broken down to an unidentified product, or the ability of DTP to prevent CP-induced lung and bladder damage is related to some other mechanism. The possibility that mercapturic acid metabolites of acrolein released the parent electrophile in the urine was not supported by the finding that probenecid did not prevent CP-induced bladder toxicity.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , Cyclophosphamide/toxicity , Cystitis/drug therapy , Lung Diseases/drug therapy , Purines/therapeutic use , Animals , Cystitis/chemically induced , Cystitis/pathology , Liver/drug effects , Liver/pathology , Lung/drug effects , Lung/pathology , Lung Diseases/chemically induced , Lung Diseases/pathology , Male , Mice , Mice, Inbred ICR , Probenecid/therapeutic use , Purines/urine , Urinary Bladder/drug effects , Urinary Bladder/pathology
20.
Toxicol Sci ; 42(2): 109-16, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9579023

ABSTRACT

Overexpression of the bcl-2 and the related bcl-xL protooncogene proteins enhance cell survival by inhibiting apoptosis induced by many agents including oxidants. Whether these proteins contribute to survival in oxidant-resistant cells is not known. The current study assessed the expression of bcl-2 and bcl-xL proteins in human glioblastoma U87MG cells and human lung adenocarcinoma A549 cells selected for resistance to 0, 50, 100, 200, and 400 microM H2O2 by exposure to this oxidant one time each passage for 9 months. When examined 7 to 32 days after cessation of peroxide exposure (times when peroxide resistance was maintained), bcl-2 protein levels were significantly increased in most peroxide-resistant U87MG cells. However, the increase was not dose dependent and was not accompanied by an increase in mRNA levels. A549 cells did not express significant levels of bcl-2 protein, although bcl-2 mRNA was detectable. A549 cells expressed large amounts of bcl-xL and immunohistochemistry demonstrated extensive localization of this protein around the nucleus. However, expression of this protein was not altered in peroxide-resistant lines nor was bcl-2 protein increased to a measurable level. U87MG cells also expressed bcl-xL but it was not altered in peroxide-resistant cells. Although the increased bcl-2 protein in peroxide-resistant U87MG cells may contribute to their oxidant tolerance, the lack of a dose-response relationship, the failure to induce bcl-xL protein, and the absence of any bcl-2 or bcl-xL protein induction in peroxide-resistant A549 cells suggest these genes are not primary factors in oxidant resistance.


Subject(s)
Peroxides/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Drug Resistance, Neoplasm , Humans , Immunohistochemistry , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...