Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
PLoS One ; 7(9): e45327, 2012.
Article in English | MEDLINE | ID: mdl-23028936

ABSTRACT

Differently from the antiapoptotic action most commonly assigned to peroxisome proliferators (PPs), we demonstrated that some of them, clofibrate (CF) in particular, display clearcut apoptogenic properties on rat hepatoma cell lines. We and others could confirm that CF as well as various other PPs can induce apoptosis in a variety of cells, including human liver, breast and lung cancer cell lines. The present study was aimed at investigating the cytotoxic action of CF on a neoplastic line of different origin, the human T leukemia Jurkat cells. We observed that CF rapidly triggers an extensive and morphologically typical apoptotic process on Jurkat cells, though not in primary T cells, which is completely prevented by the polycaspase inhibitor zVADfmk. Gene silencing studies demonstrated that CF-induced apoptosis in Jurkat cells is partially dependent on activation of caspase 2. Looking for a possible trigger of caspase 2 activation, we observed increased levels of phosphorylated eIF2α and JNK in CF-treated cells. Moreover, intracellular Ca(2+) homeostasis was perturbed. Together, these findings are suggestive for the occurrence of ER stress, an event that is known to have the potential to activate caspase 2. The present observations demonstrate that CF induces in Jurkat cells a very fast and extensive apoptosis, that involves induction of ER stress and activation of caspases 2 and 3. Since apoptosis in Jurkat cells occurs at pharmacologically relevant concentrations of CF, the present findings encourage further in depth analysis in order to work out the potential implications of CF cytotoxcity on leukemic cells.


Subject(s)
Apoptosis/drug effects , Caspase 2/metabolism , Clofibrate/pharmacology , Endoplasmic Reticulum Stress/physiology , Jurkat Cells/drug effects , Jurkat Cells/enzymology , Blotting, Western , Caspase 2/genetics , Flow Cytometry , Humans , Jurkat Cells/cytology
2.
Exp Hematol ; 40(5): 379-85, 2012 May.
Article in English | MEDLINE | ID: mdl-22269118

ABSTRACT

The biology of T-cell acute lymphoblastic leukemia (ALL) is characterized by functional pre-T-cell receptor (TCR) signaling. Non-T-cell activation linker (NTAL) is a nonenzymatic transmembrane adaptor molecule that is involved in the proximal signaling of lymphocytes. In our previous work, we found an association between high NTAL expression in T-cell ALL blasts and a favorable response to initial glucocorticoid treatment. In the present study, we confirm our previous observation in an experimental model. In addition, the molecular mechanism of the contribution of NTAL to malignant T-cell ALL blast signaling and to methylprednisolone-induced cell death is analyzed. In the in vitro experiments, we used the T-cell ALL Jurkat cell line (Jurkat/wt) and derived Jurkat cell line with stable NTAL expression (Jurkat/NTAL(+)). Cell signaling and cell death after methylprednisolone treatment and after TCR stimulation were analyzed using flow cytometry, Western blot, and quantitative polymerase chain reaction. Jurkat/NTAL(+) cells are significantly more sensitive to both methylprednisolone treatment and TCR-induced stimulation. In addition, after TCR stimulation, Jurkat/NTAL(+) cells show a higher level of intracellular extracellular signal-regulated kinase 1/2 (ERK) phosphorylation and increased expression of the CD69 activation marker on the cell surface than the Jurkat/wt cells. The ERK inhibitor U0126 almost completely abrogates TCR-induced cell death and, importantly, reverses the sensitizing effect of the NTAL protein on methylprednisolone-induced cell death. In conclusion, NTAL acts as a tumor suppressor that enhances the proximal signaling of leukemic blasts. The key downstream molecule responsible for the biological effect of TCR signaling is ERK. Higher ERK phosphorylation leads to enhanced cell death after TCR stimulation and increases cell sensitivity to methylprednisolone-induced cell death.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Methylprednisolone/pharmacology , Neoplasm Proteins/physiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Butadienes/pharmacology , Drug Resistance, Neoplasm/drug effects , Humans , In Vitro Techniques , Jurkat Cells/drug effects , Jurkat Cells/enzymology , Lectins, C-Type/metabolism , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Proteins/antagonists & inhibitors , Nitriles/pharmacology , Phosphorylation/drug effects , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Receptors, Antigen, T-Cell/drug effects
3.
J Dairy Sci ; 93(9): 3925-30, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20723665

ABSTRACT

Abnormal methylation of the promoter of several genes is common in patients with acute lymphoblastic leukemia. Methylation of DNA is brought about by DNA methyltransferases (DNMT). Bovine lactoferricin (Lfcin B) is a cationic peptide that possesses potent in vitro and in vivo anticancer activity and might affect the expression of DNMT1. In the current study, we determined the mRNA and protein expression of DNMT1 in Jurkat T-leukemia cells, after incubation with Lfcin B, by real-time quantitative reverse transcription PCR and Western blot analysis. The results of real-time quantitative reverse transcription PCR showed that DNMT1 expression in Jurkat T-leukemia cells was reduced after treatment with Lfcin B, and Lfcin B reduced the half-life of DNMT1 mRNA from approximately 8 to 2h. The results of Western blot analysis showed that the expression of DNMT1 protein was down-modulated by Lfcin B in Jurkat T-leukemia cells. Moreover, we found that protein biosynthesis in Jurkat T-leukemia cells was essential for Lfcin B to down-modulate the expression of DNMT1.


Subject(s)
Jurkat Cells/drug effects , Lactoferrin/pharmacology , Repressor Proteins/analysis , Animals , Blotting, Western , Cattle , Dactinomycin/pharmacology , Down-Regulation/drug effects , Half-Life , Humans , Jurkat Cells/enzymology , Protein Biosynthesis/drug effects , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
4.
Article in Chinese | MEDLINE | ID: mdl-21280324

ABSTRACT

OBJECTIVE: To study the effect of Itk down regulation on Jurkat cell proliferation and inflammatory cytokines production, and provide useful data for Itk as an attractive target for potential drugs. METHODS: Three shRNAs against different region of Itk were constructed and cotransfected with pEGFP-C1-hItk. The shRNA, which can knock down Itk, was selected and packed into lentivirus. After Jurkat cells were transfected with shRNA lentivirus, the change of Itk protein expression, cell proliferation and cytokines production was observed. RESULTS: Itk mRNA was reduced about 55% in Jurkat cells transfected with Itk-shRNA1, compared with that in control cells shRNAnon (P < 0.05). Knocking down Itk expression had a profound inhibitory effect on Jurkat cell proliferation. In addition, there was a substantial decrease in level of cytokines, such as IL-2, IL-5, IL-10 and IFN-gamma, produced by cell transfected with Itk-shRNA1. CONCLUSION: Knocking down Itk expression can inhibit Jurkat cell proliferation and inflammatory cytokines production.


Subject(s)
Cytokines/genetics , Down-Regulation , Jurkat Cells/enzymology , Protein-Tyrosine Kinases/immunology , Animals , Cell Proliferation , Cytokines/immunology , Gene Knockdown Techniques , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-5/genetics , Interleukin-5/immunology , Jurkat Cells/cytology , Jurkat Cells/immunology , Mice , Protein-Tyrosine Kinases/genetics
5.
Leukemia ; 23(8): 1507-14, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19282831

ABSTRACT

Interactions between inhibitors of the proteasome and histone deacetylases have been examined in human T-leukemia/lymphoma cells both in vitro and in vivo. Co-exposure of cells to bortezomib and suberoylanilide hydroxamic acid (SAHA) synergistically induces T-leukemia/lymphoma cells to undergo apoptosis, consistent with a significant increase in mitochondrial injury and caspase activation. These events are accompanied by inhibition of cyto-protective signaling pathways, including the nuclear factor (NF)-kappaB, Raf-1/mitogen-induced extracellular kinase (MEK)/extracellular signal-related kinase (ERK) and AKT pathways, and activation of stress-related cascades, including the stress-activated kinases c-jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38MAPK). Moreover, bortezomib in conjunction with SAHA efficiently induces apoptosis of primary T-leukemia/lymphoma cells and inhibits tumor growth in a murine xenograft model established with subcutaneous injection of Jurkat cells. Taken together, these findings confirm the synergistic anti-tumor effect of the proteasome and histone deacetylase inhibitors, and provide an insight into the future clinical applications of bortezomib-SAHA combining regimen in treating T-cell malignancies.


Subject(s)
Boronic Acids/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Leukemia, T-Cell/pathology , Lymphoma, T-Cell/pathology , Neoplasm Proteins/antagonists & inhibitors , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Pyrazines/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Bortezomib , Cell Line, Tumor/drug effects , Cell Line, Tumor/pathology , Drug Synergism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/physiology , Jurkat Cells/drug effects , Jurkat Cells/enzymology , Jurkat Cells/transplantation , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/enzymology , Lymphoma, T-Cell/enzymology , Mice , Mice, Nude , Protein Kinases/physiology , Vorinostat , Xenograft Model Antitumor Assays
6.
Oral Microbiol Immunol ; 24(2): 146-51, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19239642

ABSTRACT

INTRODUCTION: Tumour necrosis factor-alpha converting enzyme (TACE), also known as ADAM17, is a membrane-bound metalloprotease and disintegrin. It is produced by a number of host cells and is known to shed and release cell-bound cytokines, particularly members of the tumour necrosis factor family. The aim of this study was to investigate the effect of Porphyromonas gingivalis on TACE production by a human T-cell line, to identify putative virulence factors involved in this process, and to investigate the effect of doxycycline. METHODS: P. gingivalis 6-day culture supernatants were used to challenge Jurkat T cells for 6 h. Secreted and cell-associated TACE levels were measured by enzyme-linked immunosorbent assay, whereas messenger RNA expression was investigated by quantitative real-time polymerase chain reaction. To investigate the involvement of cysteine proteases or proteinaceous components in general, P. gingivalis culture supernatants were treated with the specific chemical inhibitor TLCK or heat-inactivated, respectively. The effect of doxycycline on the regulation of TACE secretion by P. gingivalis was also investigated. RESULTS: P. gingivalis challenge resulted in a concentration-dependent enhancement of TACE messenger RNA expression and protein release by Jurkat cells. TLCK treatment or heat treatment of P. gingivalis culture supernatants decreased TACE release to control levels. Doxycycline inhibited TACE secretion dose dependently. CONCLUSION: The induction of TACE by T cells in response to P. gingivalis may in turn favour the shedding of host cell-bound cytokines into the local microenvironment, potentially amplifying the inflammatory response. In the present experimental system, P. gingivalis cysteine proteases are involved in TACE release by T cells.


Subject(s)
ADAM Proteins/biosynthesis , Cysteine Endopeptidases/metabolism , Jurkat Cells/enzymology , Porphyromonas gingivalis/physiology , ADAM17 Protein , Adhesins, Bacterial/metabolism , Anti-Bacterial Agents/pharmacology , Culture Media, Conditioned/pharmacology , Doxycycline/pharmacology , Gene Expression , Gingipain Cysteine Endopeptidases , Humans , Jurkat Cells/drug effects , Jurkat Cells/microbiology , Lipopolysaccharides/physiology , Protease Inhibitors/pharmacology , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/biosynthesis , Tosyllysine Chloromethyl Ketone/pharmacology , Virulence Factors
7.
FASEB J ; 22(7): 2243-52, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18263700

ABSTRACT

Live-cell detection of intracellular enzyme activity requires that substrates are cell-permeable and that the generated products are easily detected and retained in cells. Our objective was to create a novel fluorogenic substrate that could be used for real-time detection of apoptosis in living cells. We have synthesized a highly cell-permeable caspase-3 substrate, DEVD-NucView488, by linking a fluorogenic DNA-binding dye to the caspase-3 recognition sequence that renders the dye nonfunctional. On substrate cleavage, the dye is released and becomes highly fluorescent on binding to DNA. DEVD-NucView488 detected caspase-3 activation within a live-cell population much earlier and with higher sensitivity compared with other apoptosis reagents that are currently available. Furthermore, cells incubated with DEVD-NucView488 exhibited no toxicity and normal apoptotic progression. DEVD-NucView488 is an ideal substrate for kinetic studies of caspase-3 activation because it detects caspase-3 activity in real-time and also efficiently labels DNA in nuclei of caspase-3-activated cells for real-time fluorescent visualization of apoptotic morphology. The strategy utilized in the design of this fluorogenic substrate can be applied in future endeavors to develop substrates for detecting real-time intracellular enzyme activity.


Subject(s)
Caspase 3/metabolism , Peptide Fragments/metabolism , Binding Sites , DNA/metabolism , Enzyme Activation , HeLa Cells/enzymology , Humans , Jurkat Cells/enzymology , Kinetics , Substrate Specificity
8.
J Food Prot ; 70(10): 2377-82, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17969621

ABSTRACT

The potential use of ricin as a biological weapon in food highlights the necessity for the development of food-specific detection methods. Current methods for the detection of ricin consist of various immunoassays, which detect only one subunit of the ricin toxin and therefore may not be indicative of a biologically active molecule. An in vivo assay, such as a mouse bioassay, can indicate the biological activity of the toxin; however, this method is not feasible for laboratories that do not have animal testing facilities. The purpose of this study was to develop an in vitro assay for the detection of biologically active ricin in beverages and liquid foods. Acidic and high-protein beverages were spiked with either purified ricin or ground castor beans and added to cultured human Jurkat cells. After an overnight incubation, the supernatant was tested for lactate dehydrogenase (LDH) activity with a colorimetric assay. LDH was released from the cytosol upon cell damage and was positively correlated with cell death. Ricin was detectable in all the matrices tested, with a sensitivity of 10 to 100 pg/ml. Biologically active ricin was detectable in all the matrices incubated with ground castor bean material. This method provides a confirmatory way to detect biologically active ricin that can be utilized by laboratories lacking animal facilities.


Subject(s)
Beverages/analysis , Food Contamination/analysis , Jurkat Cells/drug effects , Ricin/isolation & purification , Ricinus communis/chemistry , Biological Assay , Bioterrorism , Colorimetry , Dose-Response Relationship, Drug , Humans , Jurkat Cells/enzymology , L-Lactate Dehydrogenase/metabolism , Sensitivity and Specificity
9.
Biochem Biophys Res Commun ; 345(2): 608-16, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16690028

ABSTRACT

Apoptosis involves a cascade of biochemical and morphological changes resulting in the systematic disintegration of the cell. Caspases are central mediators of this process. Supporting and primary roles for serine proteases as pro-apoptotic mediators have also been highlighted. Evidence for such roles comes largely from the use of pharmacological inhibitors; as a consequence information regarding their apoptotic function and biochemical properties has been limited. Here, we circumvented limitations associated with traditional serine protease inhibitors through use of a fluorescently labelled inhibitor of serine proteases (FLISP) that allowed for analysis of the specificity, regulation and positioning of apoptotic serine proteases within a classical apoptotic cascade. We demonstrate that staurosporine triggers a caspase-dependant induction of chymotrypsin-like activity in the nucleus of apoptotic Jurkat T cells. We show that serine protease activity is required for the generation of late stage nuclear events including condensation, fragmentation and DNA degradation. Furthermore, we reveal caspase-dependant activation of two chymotrypsin-like protein species that we hypothesize mediate cell death-associated nuclear events.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Cell Nucleus/physiology , Chymotrypsin/metabolism , Jurkat Cells/enzymology , Apoptosis/drug effects , Caspase Inhibitors , Cell Nucleus/drug effects , DNA Fragmentation/drug effects , DNA Fragmentation/physiology , Enzyme Activation , Fluorescent Dyes , Humans , Jurkat Cells/cytology , Serine Proteinase Inhibitors/pharmacology , Staurosporine/pharmacology , Substrate Specificity
10.
Cancer Lett ; 227(2): 169-74, 2005 Sep 28.
Article in English | MEDLINE | ID: mdl-16112419

ABSTRACT

Telomerase activity is repressed in normal human somatic cells, but is activated in most cancers, suggesting that telomerase may be an important target for cancer therapy. Inhibition of telomerase in cancer cells has been shown to limit the growth of human cancer cells in culture. In this study, we report that helenalin, a natural sesquiterpene lactone, is a potent and selective inhibitor for human telomerase. In vitro studies indicate that this drug can inactivate telomerase directly in a manner that is dependent on concentration and time. The inhibitory action of this drug on telomerase is selective since the presence of excessive externally added proteins did not protect the inhibition and all of the other enzymes tested in this study were not inhibited by this drug. Furthermore, we demonstrated that helenalin can inhibit the expression of hTERT and telomerase in hematopoietic cancer cells. Therefore, the anti-tumor activity of helenalin is attributed, at least in part, to the inhibition of telomerase.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Sesquiterpenes/pharmacology , Telomerase/antagonists & inhibitors , Telomerase/metabolism , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/metabolism , DNA-Directed RNA Polymerases/metabolism , HL-60 Cells/enzymology , Humans , In Vitro Techniques , Jurkat Cells/enzymology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sesquiterpenes, Guaiane , Telomerase/genetics , Toxins, Biological
11.
Cell Death Differ ; 12(10): 1344-57, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15905878

ABSTRACT

In Jurkat cells, the decreased cell growth rate associated with a long-lasting deactivation of the mammalian target of rapamycin (mTOR)/p70 ribosomal S6 kinase (S6K)-signaling pathway generates a cell population of progressively reduced cellular mass and size. When promoted by rapamycin as prototype inhibitor, the mTOR deactivation-dependent cell size reduction was associated with slowed, but not suppressed, proliferation. Small-size cells were significantly protected from apoptosis induced by Fas/Apo-1 death-receptor activation (as shown by reduced procaspase cleavage and decreased catalytic activity of relevant caspases) or by stress signals-dependent mitochondrial perturbation (as shown by reduced cleavage of caspase-2, lower dissipation of mitochondrial membrane potential and decreased release of cytochorome c and apoptosis-inducing factor from mitochondria). Protection faded when reactivation of the mTOR/S6K pathway promoted the cell recovery to normal size. These results suggest that cells induced to reduce their mass by the mTOR deactivation-dependent inhibition of cell growth become more resilient to lethal assaults by curbing the cell's suicidal response.


Subject(s)
Apoptosis/physiology , Jurkat Cells/cytology , Protein Kinases/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Cell Growth Processes/physiology , Cell Size , Chromones/pharmacology , Cytochrome c Group/metabolism , Energy Metabolism , Enzyme Inhibitors/pharmacology , Humans , Jurkat Cells/enzymology , Jurkat Cells/metabolism , Leucine/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Morpholines/pharmacology , Phosphorylation , Protein Kinases/physiology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
12.
Chem Res Toxicol ; 18(3): 420-7, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15777082

ABSTRACT

The role of mitochondria and apical caspases in apoptosis induced by the benzene metabolite hydroquinone (HQ) remains to be elucidated. Here, we investigated the involvement of mitochondria and activation of the apical caspases-8 and -9 in HQ induced apoptosis in myeloperoxidase (MPO)-rich HL-60 and MPO-deficient Jurkat T cells. Treatment of HL-60 and Jurkat cells with HQ resulted in apoptosis as assessed by phosphatidyl serine (PS) exposure, loss of mitochondrial transmembrane potential (MTP), release of cytochrome c, and processing of apical caspases-8 and -9 and executioner caspase-3. In HQ-treated HL-60 cells, pretreatment with the pan-caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone (ZVAD), which did not inhibit PS exposure, also failed to abrogate the loss of MTP and release of cytochrome c. However, complete processing of caspase-9 was inhibited in the presence of ZVAD. In marked contrast, in HQ-treated Jurkat cells, ZVAD significantly abrogated PS exposure, loss of MTP, and caspase-9 processing but not release of cytochrome c. Although ZVAD-sensitive caspase-8 processing occurred in both cell types, pretreatment with either fas-receptor blocking ZB4 or fas-ligand NOK1 neutralizing antibodies did not inhibit HQ-induced apoptosis. In conclusion, our results demonstrate that HQ induced apoptosis in Jurkat cells occurs via a ZVAD-inhibitable, caspase-dependent process, while in HL-60 cells, apoptosis occurs predominantly via caspase-independent mechanisms. Our results emphasize that both caspase-dependent and independent mechanisms should be considered in the intrinsic apoptotic pathway induced by HQ.


Subject(s)
Apoptosis/drug effects , Caspases/biosynthesis , HL-60 Cells/drug effects , Hydroquinones/toxicity , Jurkat Cells/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Caspases/adverse effects , Drug Combinations , Enzyme Inhibitors/pharmacology , Flow Cytometry , HL-60 Cells/enzymology , HL-60 Cells/pathology , Humans , Jurkat Cells/enzymology , Jurkat Cells/pathology , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/enzymology , Phosphatidylserines/metabolism
13.
Biochem J ; 382(Pt 2): 651-7, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15209560

ABSTRACT

The first step in caspase activation is transition of the latent zymogen to an active form. For the initiator caspases, this occurs through dimerization of monomeric zymogens at an activating complex. Recent studies have suggested that FLIP(L) [FLICE-like inhibitory protein, long form; FLICE is FADD (Fas-associated death domain protein)-like interleukin-1beta-converting enzyme], previously thought to act solely as an inhibitor of caspase-8 activation, can under certain circumstances function to enhance caspase activation. Using an in vitro induced-proximity assay, we demonstrate that activation of caspases-8 and -10 occurs independently of cleavage of either the caspase or FLIP(L). FLIP(L) activates caspase-8 by forming heterodimeric enzyme molecules with substrate specificity and catalytic activity indistinguishable from those of caspase-8 homodimers. Significantly, the barrier for heterodimer formation is lower than that for homodimer formation, suggesting that FLIP(L) is a more potent activator of caspase-8 than is caspase-8 itself.


Subject(s)
Caspases/metabolism , Intracellular Signaling Peptides and Proteins/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspase 10 , Caspase 8 , Cell Line, Tumor , Dimerization , Enzyme Activation/physiology , Enzyme Precursors/metabolism , Humans , Hydrolysis , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells/enzymology
14.
Biochem J ; 381(Pt 3): 841-6, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15117283

ABSTRACT

Human selenoprotein P (SeP), a selenium-rich plasma glycoprotein, is presumed to contain ten selenocysteine residues; one of which is located at the 40th residue in the N-terminal region and the remaining nine localized in the C-terminal third part. We have shown that SeP not only catalyses the reduction of phosphatidylcholine hydroperoxide by glutathione [Saito, Hayashi, Tanaka, Watanabe, Suzuki, Saito and Takahashi (1999) J. Biol. Chem. 274, 2866-2871], but also supplies its selenium to proliferating cells [Saito and Takahashi (2002) Eur. J. Biochem. 269, 5746-5751]. Treatment of SeP with plasma kallikrein resulted in a sequential limited proteolysis (Arg-235-Gln-236 and Arg-242-Asp-243). The N-terminal (residues 1-235) and C-terminal (residues 243-361) fragments exhibited enzyme activity and selenium-supply activity respectively. These results confirm that SeP is a bi-functional protein and suggest that the first selenocysteine residue is the active site of the enzyme and the remaining nine residues function as a selenium supplier.


Subject(s)
Peptides/chemistry , Proteins/chemistry , Binding Sites , Blood Coagulation/physiology , Cell Line, Tumor , Glutathione Peroxidase/metabolism , Humans , Hydrolysis , Jurkat Cells/enzymology , Jurkat Cells/metabolism , Peptide Fragments/blood , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptides/metabolism , Plasma Kallikrein/metabolism , Plasma Kallikrein/physiology , Protein Structure, Tertiary , Proteins/metabolism , Selenium/metabolism , Selenoprotein P , Selenoproteins , Sequence Homology, Amino Acid , Substrate Specificity
15.
Biochem J ; 380(Pt 2): 425-33, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-14992689

ABSTRACT

Among the three isoenzymes of neuraminidase (Neu) or sialidase, Neu-1 has been suggested to be induced by cell activation and to be involved in IL (interleukin)-4 biosynthesis in murine T-cells. In the present study, we found that antigen-induced airway eosinophilia, a typical response dependent on Th2 (T-helper cell type 2) cytokines, as well as mRNA expression of Th2 cytokines, including IL-4, are suppressed in Neu-1-deficient mice, thereby demonstrating the in vivo role of murine Neu-1 in regulation of Th2 cytokines. To elucidate the roles of various sialidases in human T-cell activation, we investigated their tissue distribution, gene induction and function. Neu-1 is the predominant isoenzyme at the mRNA level in most tissues and cells in both mice and humans, including T-cells. T-cells also have significant levels of Neu-3 mRNAs, albeit much lower than those of Neu-1, whereas the levels of Neu-2 mRNAs are minimal. In human T-cells, both Neu-1 and Neu-3 mRNAs are significantly induced by T-cell-receptor stimulation, as is sialidase activity against 4-methylumbelliferyl- N -acetylneuramic acid (a substrate for both Neu-1 and Neu-3) and the ganglioside G(D1a) [NeuAcalpha2-3Galbeta1-3GalNAcbeta1-4(NeuAcalpha2-3)Galbeta1-4Glcbeta1-cer] (a substrate for Neu-3, but not for Neu-1). The expression of the two sialidase genes may be under differential regulation. Western blot analysis and enzymic comparison with recombinant sialidases have revealed that Neu-3 is induced as a major isoform in activated cells. The induction of Neu-1 and Neu-3 in T-cells is unique. In human monocytes and neutrophils stimulated with various agents, the only observation of sialidase induction has been by IL-1 in neutrophils. Functionally, a major difference has been observed in Jurkat T-cell lines over-expressing Neu-1- and Neu-3. Upon T-cell receptor stimulation, IL-2, interferon-gamma, IL-4 and IL-13 are induced in the Neu-1 line, whereas in the Neu-3 line the same cytokines are induced, with the exception of IL-4. Taken together, these results suggest an important immunoregulatory role for both Neu-1 and Neu-3 in humans.


Subject(s)
Cell Membrane/enzymology , Lysosomes/enzymology , Neuraminidase/biosynthesis , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/enzymology , Animals , CD28 Antigens/pharmacology , CD3 Complex/pharmacology , CD4-Positive T-Lymphocytes/enzymology , Cell Line , Cell Line, Tumor , Cells, Cultured , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Enzyme Induction/drug effects , Enzyme Induction/physiology , Eosinophilia/etiology , Eosinophilia/pathology , Gene Expression Regulation, Enzymologic/physiology , Humans , Insecta/cytology , Isoenzymes/biosynthesis , Isoenzymes/physiology , Jurkat Cells/enzymology , Leukocyte Count , Lung/drug effects , Lung/pathology , Lymphocyte Activation/physiology , Mice , Mice, Inbred DBA , Mice, Inbred Strains , Neuraminidase/deficiency , Neuraminidase/physiology , Organ Specificity/physiology , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/enzymology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Transcriptional Activation
16.
Haematologica ; 89(1): 11-20, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14754601

ABSTRACT

BACKGROUND AND OBJECTIVES: Fanconi anemia (FA) cells are characteristically hypersensitive to bifunctional alkylating agents, notably mitomycin C (MMC), causing increased programmed cell death (PCD). FA cells also have abnormalities in mitochondrial function. We hypothesized that the abnormalities in PCD are mitochondrially mediated. We examined mitochondrial function in FA cells, comparing the intrinsic death pathway induced by MMC with the extrinsic pathway via Fas antibody, which can bypass the mitochondria. DESIGN AND METHODS: Normal and FA lymphoblastoid cell lines were treated with MMC or agonistic anti-Fas antibody. PCD was assessed using flow cytometry, Western blot analysis, and DNA gel electrophoresis. RESULTS: FA cells showed hypersensitivity to MMC, but slight resistance to Fas-mediated PCD. MMC induced chromatin condensation, but not apoptotic body formation. Fas induced classical apoptosis. MMC failed to induce mitochondrial depolarization, while some depolarization occurred with anti-Fas. These results suggested that MMC failed to induce caspase activity in FA cells. No cleavage of caspase 3 was observable and PCD was not inhibited by the caspase inhibitor zVAD-fmk. Fas-induced caspase 3 cleavage, and cell death was inhibited by zVAD-fmk. There were common downstream abnormalities in the execution phase of PCD, as both agonists failed to cleave PARP, or to induce nucleosomal fragmentation. INTERPRETATION AND CONCLUSIONS: Our results suggest that mitochondrial function in FA cells is abnormal, resulting in necrotic or caspase independent PCD, but that further abnormalities may exist downstream of the mitochondria. This may have implications in explaining in vivo aspects of FA.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Apoptosis/drug effects , Fanconi Anemia/drug therapy , Mitomycin/therapeutic use , fas Receptor/immunology , Antibodies, Monoclonal/pharmacology , Apoptosis/physiology , Caspase 3 , Caspases/metabolism , Caspases/physiology , Cell Cycle Proteins/genetics , Cell Line , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Enzyme Activation/drug effects , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group Proteins , Humans , Jurkat Cells/drug effects , Jurkat Cells/enzymology , Jurkat Cells/metabolism , Lymphocytes/drug effects , Lymphocytes/enzymology , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Mitomycin/pharmacology , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Poly(ADP-ribose) Polymerases/metabolism , Tumor Cells, Cultured
17.
Br J Cancer ; 89(7): 1366-74, 2003 Oct 06.
Article in English | MEDLINE | ID: mdl-14520473

ABSTRACT

CD26/dipeptidyl peptidase IV (DPPIV) is a cell surface-bound ectopeptidase with important roles in T-cell activation and tumour biology. We now report that CD26/DPPIV enhances sensitivity to apoptosis induced by the antineoplastic agents doxorubicin and etoposide. In particular, CD26/DPPIV presence is associated with increased susceptibility to the mitochondrial pathway of apoptosis, documented by enhanced cleavage of poly (ADP ribose) polymerase (PARP), caspase-3 and caspase-9, Bcl-xl, and Apaf-1, as well as increased expression of death receptor 5 (DR5). We also show that the caspase-9-specific inhibitor z-LEHD-fmk inhibits drug-mediated apoptosis, leading to decreased PARP and caspase-3 cleavage, and reduced DR5 expression. Importantly, through detailed studies that demonstrate the association between topoisomerase II alpha expression and DPPIV activity, our data provide further evidence of the key role played by CD26 in biological processes.


Subject(s)
Apoptosis/drug effects , DNA Topoisomerases, Type II/metabolism , Dipeptidyl Peptidase 4/physiology , Enzyme Inhibitors/pharmacology , Annexin A5/metabolism , Antigens, Neoplasm , Antineoplastic Agents/pharmacology , Apoptotic Protease-Activating Factor 1 , Blotting, Western , Caspases/metabolism , Cell Nucleus/metabolism , DNA-Binding Proteins , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Etoposide/pharmacology , Flow Cytometry , Humans , Jurkat Cells/drug effects , Jurkat Cells/enzymology , Jurkat Cells/pathology , Membrane Potentials/drug effects , Mitochondria/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Propidium/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/metabolism , Topoisomerase II Inhibitors , Transfection , bcl-X Protein
18.
Exp Cell Res ; 290(2): 420-6, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14567999

ABSTRACT

Myriadenolide is a diterpene that we have recently isolated from the extract of Alomia myriadenia (Asteraceae). Here we show for the first time that myriadenolide has caspase-dependent cytotoxic activity against human leukemia cells from both lymphocytic (Jurkat) and monocytic (THP-1) lineages, because preincubation of Jurkat or THP-1 cells with the broad-spectrum caspase inhibitor z-VAD-fmk completely abrogated cell death. Moreover, the mitochondrial pathway is implicated as mitochondrial depolarization and caspase-9 and caspase-3 activation were observed. Interestingly, caspase-8 and cleavage of the proapoptotic member of the Bcl-2 family BID was also observed during apoptosis induced by myriadenolide, suggesting a role for the caspase-8/BID pathway. However, interference with Fas or TNFR1 signaling did not interfere with apoptosis in our experimental system. Furthermore, pretreatment of cells with the caspase-3 inhibitor DEVD-fmk completely blocked the activation of caspase-8, suggesting that the activation of the caspase-8/BID pathway is part of an amplification loop initiated by caspase-3. Taken together, our results indicate myriadenolide as a novel candidate for the treatment of hematological malignancies.


Subject(s)
Apoptosis/drug effects , Asteraceae/chemistry , Caspases/metabolism , Diterpenes/pharmacology , Mitochondria/drug effects , Antigens, CD/metabolism , Blotting, Western , Caspase 3 , Caspase 8 , Caspase 9 , Cell Cycle , Cytochromes c/metabolism , Diterpenes/isolation & purification , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Jurkat Cells/drug effects , Jurkat Cells/enzymology , Membrane Potentials/drug effects , Mitochondria/enzymology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Signal Transduction/drug effects , fas Receptor/metabolism
19.
Microsc Res Tech ; 62(3): 192-200, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14506684

ABSTRACT

Two distinct types of cell death have been described: apoptosis and necrosis. However, it is becoming increasingly clear that the differences between these two types are far less numerous than initially thought. Morphological analyses might provide important information to distinguish apoptotic from necrotic samples. We recently reported that in necrotic, but not apoptotic, HL-60 human myeloid leukaemia cells, the nuclear protein topoisomerase IIalpha concentrated in nucleoli. In order to ascertain whether or not this phenomenon was restricted to a peculiar cell type or could be detected also in cells of lymphoid lineage, we performed an investigation aimed at defining the localization of topoisomerase IIalpha in apoptotic and necrotic Jurkat human T lymphoblastoid cells. Immunofluorescence staining demonstrated that topoisomerase IIalpha was excluded from the condensed chromatin of apoptotic cells, whereas in necrotic cells it was localized in discrete nuclear dots. Immuno-electron microscopy analysis showed that topoisomerase IIalpha was undetectable in nucleoli of normal and apoptotic cells, whereas it was present in the nucleolus of necrotic cells irrespectively of the type of inducer used (ethanol, H(2)O(2), HgCl(2)). Taken together, our findings identify topoisomerase IIalpha as a potential morphological marker useful to discriminate between apoptotic and necrotic cells.


Subject(s)
Apoptosis , DNA Topoisomerases, Type II/metabolism , Jurkat Cells/enzymology , Necrosis , Antigens, Neoplasm , Cell Differentiation , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , DNA-Binding Proteins , Fluorescent Antibody Technique , Humans , Immunoblotting , Jurkat Cells/pathology , Microscopy, Electron
20.
Anticancer Res ; 23(2B): 1235-43, 2003.
Article in English | MEDLINE | ID: mdl-12820377

ABSTRACT

The current study characterizes the lipogenic enzyme fatty acid synthase (FAS; EC 2.3.1.85) in pediatric tumor cell lines of neural or neural crest origin [medulloblastoma (Daoy), malignant rhabdoid tumor of kidney (SM II), retinoblastoma (Y79), and neuroblastoma (SK-N-SH)]. Constitutive FAS content and activity in these lines were compared to human fibroblast cell line Hs27. Hs27 exhibits low levels of FAS and recapitulates enzyme status in normal human tissues under most physiological conditions. Western analysis detected significantly larger amounts of FAS protein in Y79 and SK-N-SH than Daoy, SM II and Hs27. Incorporation of radiolabeled malonyl-CoA into total cellular lipid revealed that enzyme activity correlated with amount. Increased FAS content and activity in Y79 and SK-N-SH relative to the other cell lines and Hs27, in particular, implied enzyme activation in retinoblastoma and neuroblastoma lineages. The enzyme also showed evidence of hormonal regulation, as dexamethasone induced FAS protein in Daoy and SK-N-SH. However, hormonal induction of FAS protein levels did not correlate with activity levels, which led us to speculate phosphorylation as a means of regulating the enzyme's activity. Finally, the FAS inhibitor cerulenin was investigated for its ability to suppress tumor cell growth. After four days of propagation, short-term treatment of cell lines with drug produced mean IC50s less than 10.5 micrograms/ml (i.e., 5.6 +/- 1.9 for SM II; 9.3 +/- 1.5 for Daoy; 10.2 +/- 0.2 for SK-N-SH; and 10.4 +/- 2.6 for Y79). Annexin V assays revealed that cerulenin initiated apoptosis. The antineoplastic properties of cerulenin documented here are consistent with prior studies showing its cytotoxic effects upon other types of cancer cells and illustrate the potential utility of FAS inhibition as a novel chemotherapeutic approach.


Subject(s)
Brain Neoplasms/enzymology , Cerulenin/pharmacology , Fatty Acid Synthases/metabolism , Kidney Neoplasms/enzymology , Medulloblastoma/enzymology , Neoplasm Proteins/metabolism , Neuroblastoma/enzymology , Rhabdoid Tumor/enzymology , Apoptosis/drug effects , Brain Neoplasms/pathology , Cell Lineage , Child , Culture Media, Serum-Free/pharmacology , Dexamethasone/pharmacology , Enzyme Induction/drug effects , Fatty Acid Synthases/antagonists & inhibitors , Fibroblasts/enzymology , Humans , Inhibitory Concentration 50 , Jurkat Cells/drug effects , Jurkat Cells/enzymology , Kidney Neoplasms/pathology , Lipids/biosynthesis , Malonyl Coenzyme A/metabolism , Medulloblastoma/pathology , Neoplasm Proteins/antagonists & inhibitors , Nerve Growth Factor/pharmacology , Neural Crest/enzymology , Neuroblastoma/pathology , Palmitic Acid/pharmacology , Progesterone Congeners/pharmacology , Promegestone/pharmacology , Rhabdoid Tumor/pathology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL