Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Cell Death Dis ; 4: e643, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23703388

ABSTRACT

TRAIL is a promising anticancer agent, capable of inducing apoptosis in a wide range of treatment-resistant tumor cells. In 'type II' cells, the death signal triggered by TRAIL requires amplification via the mitochondrial apoptosis pathway. Consequently, deregulation of the intrinsic apoptosis-signaling pathway, for example, by loss of Bax and Bak, confers TRAIL-resistance and limits its application. Here, we show that despite resistance of Bax/Bak double-deficient cells, TRAIL-treatment resulted in caspase-8 activation and complete processing of the caspase-3 proenzymes. However, active caspase-3 was degraded by the proteasome and not detectable unless the XIAP/proteasome pathway was inhibited. Direct or indirect inhibition of XIAP by RNAi, Mithramycin A or by the SMAC mimetic LBW-242 as well as inhibition of the proteasome by Bortezomib overcomes TRAIL-resistance of Bax/Bak double-deficient tumor cells. Moreover, activation and stabilization of caspase-3 becomes independent of mitochondrial death signaling, demonstrating that inhibition of the XIAP/proteasome pathway overcomes resistance by converting 'type II' to 'type I' cells. Our results further demonstrate that the E3 ubiquitin ligase XIAP is a gatekeeper critical for the 'type II' phenotype. Pharmacological manipulation of XIAP therefore is a promising strategy to sensitize cells for TRAIL and to overcome TRAIL-resistance in case of central defects in the intrinsic apoptosis-signaling pathway.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , X-Linked Inhibitor of Apoptosis Protein/metabolism , Apoptosis/drug effects , Carcinoma/drug therapy , Carcinoma/metabolism , Carcinoma/pathology , Caspase 3/metabolism , Caspase 8/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , HCT116 Cells , Humans , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , X-Linked Inhibitor of Apoptosis Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/antagonists & inhibitors , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
2.
Cell Death Differ ; 19(12): 1928-38, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22705850

ABSTRACT

The proapoptotic B-cell lymphoma (Bcl)-2 protein Bcl-x(S) encloses the Bcl-2 homology (BH) domains BH3 and BH4 and triggers apoptosis via the multidomain protein Bak, however, the mechanism remained elusive. For investigating Bcl-x(S) efficacy and pathways, an adenoviral vector was constructed with its cDNA under tetracycline-off control. Bcl-x(S) overexpression resulted in efficient apoptosis induction and caspase activation in melanoma cells. Indicative of mitochondrial apoptosis pathways, Bcl-x(S) translocated to the mitochondria, disrupted the mitochondrial membrane potential and induced release of cytochrome c, apoptosis-inducing factor and second mitochondria-derived activator of caspases. In melanoma cells, Bcl-x(S) resulted in significant Bak activation, and Bak knockdown as well as Bcl-x(L) overexpression abrogated Bcl-x(S)-induced apoptosis, whereas Mcl-1 (myeloid cell leukemia-1) knockdown resulted in a sensitization. With regard to the particular role of voltage-dependent anion channel 2 (VDAC2) for inhibition of Bak, we identified here a notable interaction between Bcl-x(S) and VDAC2 in melanoma cells, which was proven in reciprocal coimmunoprecipitation analyses. On the other hand, Bcl-x(S) showed no direct interaction with Bak, and its binding to VDAC2 appeared as also independent of Bak expression. Suggesting a new proapoptotic mechanism, Bcl-x(S) overexpression resulted in disruption of the VDAC2-Bak interaction leading to release of Bak. Further supporting this pathway, overexpression of VDAC2 strongly decreased apoptosis by Bcl-x(S). New proapoptotic pathways are of principle interest for overcoming apoptosis deficiency of melanoma cells.


Subject(s)
Apoptosis , Voltage-Dependent Anion Channel 2/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-X Protein/metabolism , Cell Line, Tumor , HCT116 Cells , Humans , Melanoma/metabolism , Melanoma/pathology , Myeloid Cell Leukemia Sequence 1 Protein , Protein Binding , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , bcl-2 Homologous Antagonist-Killer Protein/antagonists & inhibitors , bcl-2 Homologous Antagonist-Killer Protein/genetics
3.
Cell Death Differ ; 18(7): 1130-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21233848

ABSTRACT

During apoptosis Bcl-2 proteins control permeabilization of the mitochondrial outer membrane leading to the release of cytochrome c. Essential gatekeepers for cytochrome c release are the proapoptotic multidomain proteins, Bax, and Bak. The expression of Bax is upregulated upon cellular stress by the tumor suppressor p53. Despite the high functional homology of Bax and Bak, little is known about how the bak gene is regulated. To investigate its transcriptional regulation in further detail, we have analyzed a region spanning 8200 bp upstream of the bak start codon (within exon 2) for transcription factor-binding sites, and identified three p53 consensus sites (BS1-3). Reporter gene assays in combination with site-directed mutagenesis revealed that only one putative p53-binding site (BS3) is necessary and sufficient for induction of reporter gene expression by p53. Consistently, p53 induces expression of endogenous Bak. At the mRNA level, induction of Bak expression is weaker than induction of Puma and p21. Interestingly, Bak expression can also be induced by p73 that binds however to each of the three p53-binding sites within the bak promoter region. Our data suggest that expression of Bak can be induced by both, p53 and p73 utilizing different binding sites within the bak promoter.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , Apoptosis , Base Sequence , Binding Sites , Cell Line, Tumor , DNA-Binding Proteins/genetics , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Protein Binding , Tumor Protein p73 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
4.
Oncogene ; 27(53): 6707-19, 2008 Dec 04.
Article in English | MEDLINE | ID: mdl-18806827

ABSTRACT

P14(ARF) (p19(ARF) in the mouse) plays a central role in the regulation of cellular proliferation. Although the capacity of p14(ARF) to induce a cell cycle arrest in G1 phase depends on a functional p53/p21-signaling axis, the G2 arrest triggered by p14(ARF) is p53/p21-independent. Using isogeneic HCT116 cells either wild-type or homozygously deleted for p21, 14-3-3sigma or both, we further investigated the cooperative effect of p21 and 14-3-3sigma on cell cycle regulation and apoptosis induction by p14(ARF). In contrast to DNA damage, which induces mitotic catastrophe in 14-3-3sigma-deficient cells, we show here that the expression of p14(ARF) triggers apoptotic cell death, as evidenced by nuclear DNA fragmentation and induction of pan-caspase activities, irrespective of the presence or absence of 14-3-3sigma. The activation of the intrinsic mitochondrial apoptosis pathway by p14(ARF) was confirmed by cytochrome c release from mitochondria and induction of caspase-9- (LEHDase) and caspase-3/7-like (DEVDase) activities. Moreover, 14-3-3sigma/p21 double-deficient cells were exceedingly sensitive to apoptosis induction by p14(ARF) as compared to wild-type cells or cells lacking either gene alone. Notably, p14(ARF)-induced apoptosis was preceded by an arrest in the G2 phase of cell cycle, which coincided with downregulation of cdc2 (cdk1) protein expression and lack of its nuclear localization. This indicates that p14(ARF) impairs mitotic entry by targeting the distal DNA damage-signaling pathway and induces apoptotic cell death, rather than mitotic catastrophe, out of a transient G2 arrest. Furthermore, our data delineate that the disruption of G2/M cell cycle checkpoint control critically determines the sensitivity of the cell toward p14(ARF)-induced mitochondrial apoptosis.


Subject(s)
Apoptosis/physiology , Biomarkers, Tumor/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Exonucleases/metabolism , G1 Phase/physiology , G2 Phase/physiology , Neoplasm Proteins/metabolism , Tumor Suppressor Protein p14ARF/metabolism , 14-3-3 Proteins , Animals , Biomarkers, Tumor/genetics , CDC2 Protein Kinase , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Cyclin B/genetics , Cyclin B/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinases , Cytochromes c/genetics , Cytochromes c/metabolism , DNA Damage/physiology , DNA Fragmentation , Exonucleases/genetics , Exoribonucleases , Humans , Mice , Mitochondria/genetics , Mitochondria/metabolism , Neoplasm Proteins/genetics , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
Oncogene ; 27(11): 1618-28, 2008 Mar 06.
Article in English | MEDLINE | ID: mdl-17828297

ABSTRACT

The glutathione-dependent system is one of the key systems regulating cellular redox balance, and thus cell fate. Cysteine, typically present in its oxidized form cystine in the extracellular space, is regarded as the rate-limiting substrate for glutathione (GSH) synthesis. Cystine is transported into cells by the highly specific amino-acid antiporter system xc-. Since Burkitt's Lymphoma (BL) cells display limited uptake capacity for cystine, and are thus prone to oxidative stress-induced cell death, we stably expressed the substrate-specific subunit of system xc-, xCT, in HH514 BL cells. xCT-overexpressing cells became highly resistant to oxidative stress, particularly upon GSH depletion. Contrary to previous predictions, the increase of intracellular cysteine did not affect the cellular GSH pool, but concomitantly boosted extracellular cysteine concentrations. Even though cells were depleted of bulk GSH, xCT overexpression maintained cellular integrity by protecting against lipid peroxidation, a very early event in cell death progression. Our results show that system xc- protects against oxidative stress not by elevating intracellular GSH levels, but rather creates a reducing extracellular environment by driving a highly efficient cystine/cysteine redox cycle. Our findings show that the cystine/cysteine redox cycle by itself must be viewed as a discrete major regulator of cell survival.


Subject(s)
Amino Acid Transport System y+/metabolism , Apoptosis , Cysteine/metabolism , Cystine/metabolism , Glutathione/metabolism , Oxidative Stress , Animals , Antimetabolites, Antineoplastic/pharmacology , Blotting, Northern , Buthionine Sulfoximine/pharmacology , Caspases/metabolism , Cell Survival/drug effects , Fluorescent Antibody Technique , Glutamic Acid/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Immunoblotting , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Oxidants/pharmacology , Oxidation-Reduction , Reactive Oxygen Species/metabolism
6.
Oncogene ; 27(10): 1387-96, 2008 Feb 28.
Article in English | MEDLINE | ID: mdl-17724463

ABSTRACT

Among the members of the Bcl-2 family, the multidomain proteins Bax and Bak are crucial for the activation of mitochondria. However, it is still unclear whether they act in a unique and distinct manner or whether they exhibit redundant functions. To systematically investigate their activation on a single-cell level, we established MCF-7 cell lines stably expressing GFP-fusion variants of these proteins. We found that MCF-7/GFP-Bak cells showed an increased sensitivity to apoptosis induction by staurosporine, actinomycin D, TRAIL and overexpression of Puma compared to GFP-Bax-expressing cells. Independently of the death stimulus used, oligomerization of endogenous and exogenous Bak was mostly detected prior to an activation of Bax, whereas cells displaying oligomerized Bax in the absence of Bak clusters were not observed. In addition, activation of Bax but not Bak was attenuated by a caspase inhibitor. Consistent with this, caspase-3-deficient MCF-7 cells displayed a significantly reduced activation of endogenous Bax than caspase-3-proficient MCF-7 cells. Thus, our data strongly suggest that diverse apoptotic stimuli preferentially engage the Bak pathway, whereas the triggering of Bax occurs, at least partially, downstream of mitochondrial caspase activation, most likely constituting a positive feedback loop for the amplification of the death signal.


Subject(s)
Apoptosis/physiology , Mitochondria/metabolism , Signal Transduction/physiology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Apoptosis/drug effects , Caspase Inhibitors , Caspases/genetics , Cell Line, Tumor , Dactinomycin/pharmacology , Enzyme Inhibitors/pharmacology , Genes, Reporter , Green Fluorescent Proteins/genetics , Humans , Mitochondria/enzymology , Mitochondria/genetics , Signal Transduction/drug effects , Staurosporine/pharmacology , TNF-Related Apoptosis-Inducing Ligand/physiology , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/physiology
7.
Oncogene ; 26(49): 7038-48, 2007 Oct 25.
Article in English | MEDLINE | ID: mdl-17486061

ABSTRACT

Renal cell carcinoma (RCC) is resistant to chemotherapy, and this resistance is mirrored by a high apoptosis resistance of many RCC lines in vitro. Here, we report the loss of the pro-apoptotic BH3-only protein Bim in a large part of clinical RCC cases and provide evidence for a functional relevance of this loss. Immunohistochemistry of clear cell renal cell carcinoma cases and corresponding normal kidney showed strong Bim reactivity in renal tubules of all cases but loss of Bim in 35 of 45 RCC samples. Out of nine RCC cell lines investigated, six showed strongly diminished or undetectable levels of Bim protein by western blotting. Four RCC lines of varying apoptosis sensitivity were analysed further. Bcl-2, Bcl-x(L), Mcl-1, Bax and Bak expression did not correlate with apoptosis sensitivity. All cell lines underwent apoptosis upon forced expression of Bax and Bim, suggesting an upstream difference. In all four lines, adriamycin induced p53 but not its targets Puma or Noxa. However, apoptosis sensitivity correlated with levels of Bim protein. Bim siRNA reduced apoptosis sensitivity in a susceptible cell line. Furthermore, inhibition of histone deacetylation restored Bim expression in cell lines. These data suggest that Bim has a function as a tumor suppressor in RCC.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Adenoviridae/genetics , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Bcl-2-Like Protein 11 , Blotting, Western , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/secondary , Down-Regulation , Doxorubicin/pharmacology , Flow Cytometry , Humans , Immunoglobulin G/immunology , Kidney Neoplasms/pathology , Membrane Proteins/genetics , Membrane Proteins/immunology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/pharmacology , Rabbits , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
8.
Oncogene ; 25(50): 6582-94, 2006 Oct 26.
Article in English | MEDLINE | ID: mdl-16847458

ABSTRACT

In contrast to the initial notion that the biological activity of p14(ARF) strictly depends on a functional mdm-2/p53 signaling axis, we recently demonstrated that p14(ARF) mediates apoptosis in a p53/Bax-independent manner. Here, we show that p14(ARF) induces breakdown of the mitochondrial membrane potential and cytochrome c release before triggering caspase-9- and caspase-3/7-like activities in p53/Bax-deficient DU145 prostate cancer cells expressing wild-type Bak. Re-expression of Bax in these cells failed to further enhance p14(ARF)-induced apoptosis, suggesting that p14(ARF)-induced apoptosis primarily depends on Bak but not Bax in these cells. To further define the role of Bak and Bax in p14(ARF)-induced mitochondrial apoptosis, we employed short interference RNA for the knockdown of bak in isogeneic, p53 wild-type HCT116 colon cancer cells either proficient or deficient for Bax. There, combined loss of Bax and Bak attenuated p14(ARF)-induced apoptosis whereas single loss of Bax or Bak was only marginally effective, as in the case of DU145. Notably, HCT116 cells deficient for Bax and Bak failed to release cytochrome c and showed attenuated activation of caspase-9 (LEHDase) and caspase-3/caspase-7 (DEVDase) upon p14(ARF) expression. These data indicate that p14(ARF) triggers apoptosis via a Bax/Bak-dependent pathway in p53-proficient HCT116, whereas Bax is dispensable in p53-deficient DU145 cells. Nevertheless, a substantial proportion of p14(ARF)-induced cell death proceeds in a Bax/Bak-independent manner. This is also the case for inhibition of clonogenic growth that occurs, at least in part, through an entirely Bax/Bak-independent mechanism.


Subject(s)
Mitochondria/physiology , Tumor Suppressor Protein p14ARF/metabolism , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/genetics , Apoptosis/physiology , Apoptosis Inducing Factor/metabolism , Caspase 3/metabolism , Caspase 7/metabolism , Caspase 9/metabolism , Caspases/metabolism , Cell Survival , Genes, p53/physiology , HCT116 Cells , Humans , Mitochondria/genetics , Mitochondria/metabolism , Models, Biological , Tumor Cells, Cultured
9.
Oncogene ; 25(37): 5145-54, 2006 Aug 24.
Article in English | MEDLINE | ID: mdl-16636678

ABSTRACT

We and others have demonstrated already that TRAIL (TNF-related apoptosis-inducing ligand) is a very promising candidate for molecular targeted anticancer therapy, especially when combined with ionizing radiation or other DNA-damaging agents. Agonist monoclonal antibodies that activate and are specific for the death signaling TRAIL receptors are an alternative method to stimulate the programmed cell death pathway. Phase 1 clinical trials have subsequently been conducted and shown a very good tolerability of these antibodies. In order to assess the efficacy of TRAIL receptor stimulation to induce cell death by this alternate method, we studied the combination of the agonistic-TRAIL receptor antibodies HGS-ETR1 and HGS-ETR2 with radiation in vitro and in vivo. Induction of apoptosis after combined treatment with TRAIL receptor antibodies HGS-ETR1 and/or HGS-ETR2 (0.01, 0.1, 1.0 mg/ml) and irradiation with 2, 5 or 10 Gy was determined by fluorescence microscopy and Western blot analysis of caspase-8 and PARP. The colorectal tumour cell lines Colo 205, HCT 116 and HCT-15 were used for in vitro experiments. Growth delay experiments were performed with combined treatment with fractionated irradiation (days 1-5 and 3 Gy single dose/day) and the receptor antibodies (intraperitonially, three different concentrations, application on days 1, 4 and 8) on Colo 205 xenograft-bearing NMRI (nu/nu) nude mice. HGS-ETR1 and HGS-ETR2 induced apoptotic cell death in a dose-dependent fashion and significantly increased cell death in combination with irradiation in vitro when compared to either irradiation or antibody treatment alone. The efficacy of the combined treatment seems to be at least partially Bax-dependent. Similar to the results from cell culture experiments, in vivo experiments demonstrated a dose-dependent delay in tumour growth after combined treatment. In vivo, in the Colo205 xenograft model, HGS-ETR2 revealed a higher activity than HGS-ETR1. This is the first study to demonstrate significant efficacy of combined treatment with the monoclonal agonistic TRAIL receptor antibodies HGS-ETR1 and HGS-ETR2 and ionising radiation in in vitro and in vivo models. We postulate that HGS-ETR1 and HGS-ETR2 will be very promising new agents in the field of molecular targeted multi-modality anticancer therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apoptosis Regulatory Proteins/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/radiotherapy , Membrane Glycoproteins/immunology , Tumor Necrosis Factor-alpha/immunology , Antibodies, Monoclonal/therapeutic use , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/therapeutic use , Cell Division/drug effects , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Humans , Kinetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/therapeutic use , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/therapeutic use
10.
Oncogene ; 25(2): 165-75, 2006 Jan 12.
Article in English | MEDLINE | ID: mdl-16170360

ABSTRACT

Knowledge of the type of biological reaction to chemotherapy is a prerequisite for its rational enhancement. We previously showed that irinotecan-induced DNA damage triggers in the HCT116p53(wt) colon carcinoma cell line a long-term cell cycle arrest and in HCT116p53(-/-) cells apoptosis (Magrini et al., 2002). To compare the contribution of long-term cell cycle arrest and that of apoptosis to inhibition of cell proliferation after irinotecan-induced DNA damage, we used this isogenic system as well as the cell lines LS174T (p53(wt)) and HT-29 (p53(mut)). Both p53(wt) cell lines responded to damage by undergoing a long-term tetraploid G1 arrest, whereas the p53(mut) cell lines underwent apoptosis. Cell cycle arrest as well as apoptosis caused a similar delay in cell proliferation. Irinotecan treatment also induced in mouse tumours derived from the p53(wt) cell lines a tetraploid G1 arrest and in those derived from the p53-deficient cell lines a transient G2/M arrest and apoptosis. The delay of tumour growth was in the same range in both groups, that is, arrest- and apoptosis-mediated tumour growth inhibition was comparable. In conclusion, cell cycle arrest as well as apoptosis may be equipotent mechanisms mediating the chemotherapeutic effects of irinotecan.


Subject(s)
Apoptosis , Cell Cycle , Colonic Neoplasms/pathology , DNA Damage , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Western , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Proliferation , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Female , Flow Cytometry , Humans , Irinotecan , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation/genetics , Necrosis , Ploidies , Topoisomerase I Inhibitors , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology
11.
Oncogene ; 25(7): 972-80, 2006 Feb 16.
Article in English | MEDLINE | ID: mdl-16331277

ABSTRACT

There is an ongoing controversy regarding the relevance of apoptosis induction by ionizing irradiation as compared with other end points including transient or permanent cell cycle arrest of damaged cells. Here, we show that such permanent cell cycle arrest and apoptosis represent two sides of the same coin. MCF-7 cells fail to express procaspase-3, which results in resistance to apoptosis induced by anticancer drugs. Conversely, restoration of procaspase-3 sensitizes MCF-7 cells to chemotherapeutics including epirubicine, etoposide and taxol. In contrast, irradiation does not trigger apoptotic cell death but results in prolonged arrest in the G2 phase of the cell division cycle regardless of procaspase-3 expression. This suggested that the propensity of MCF-7 cells to arrest at the G2 checkpoint results in resistance to apoptosis upon gamma-irradiation. This G2 arrest was associated with upregulation of p21CIP/WAF-1. Inhibition of DNA-damage-induced stress kinases and p21CIP/WAF-1 expression by caffeine abrogated G2 arrest and induced apoptosis of the irradiated cells in a caspase-3-dependent manner. Inhibition of cell cycle progression by adenoviral expression of the cyclin dependent kinase inhibitor p21CIP/WAF-1 prevented apoptosis upon caffeine treatment indicating that cell cycle progression, that is, G2-release, is required for induction of apoptosis. Likewise, cells homozygously deleted for p21CIP/WAF-1 (HCT116 p21-/-) display enhanced irradiation-induced apoptosis via a caspase-3-dependent mechanism. These data indicate that the disruption of G2 checkpoint control overcomes cell cycle arrest and resistance to gamma-irradiation-induced cell death. Thus, DNA damage may trigger a permanent G2 arrest as an initial inactivation step of tumor cells where the phenomenon of apoptosis is hidden unless cell cycle arrest is overcome. The efficient induction of apoptosis upon G2 release thereby depends on the propensity to activate the key executioner caspase-3. This finding is of crucial importance for the understanding of molecular steps underlying the efficacy of ionizing radiation to delete tumor cells.


Subject(s)
Apoptosis , Breast Neoplasms/radiotherapy , Carcinoma/radiotherapy , Caspases/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Radiation Tolerance , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/metabolism , Caffeine/pharmacology , Carcinoma/metabolism , Caspase 3 , G2 Phase/drug effects , G2 Phase/radiation effects , Gamma Rays , Humans , Radiation Tolerance/drug effects , Tumor Cells, Cultured , Up-Regulation
12.
Oncogene ; 25(15): 2160-9, 2006 Apr 06.
Article in English | MEDLINE | ID: mdl-16288206

ABSTRACT

Pro- and antiapoptotic proteins of the large Bcl-2 family are critical regulators of apoptosis via the mitochondrial pathway. Whereas antiapoptotic proteins of the family share all four Bcl-2 homology domains (BH1-BH4), proapoptotic members may lack some of these domains, but all so far described proapoptotic Bcl-2 proteins enclose BH3. The bcl-x gene gives rise to several alternative splice products resulting in proteins with distinct functions as the antiapoptotic Bcl-xL and proapoptotic Bcl-xS. Here, we describe a novel Bcl-x splice product of 138 amino acids termed Bcl-xAK (Atypical Killer), which encloses the Bcl-2 homology domains BH2 and BH4 as well as the transmembrane domain, but lacks BH1 and BH3. Weak endogenous expression of Bcl-xAK was seen in melanoma and other tumor cells. Interestingly, its overexpression by applying a tetracycline-inducible expression system resulted in significant induction of apoptosis in melanoma cells, which occurred in synergism with drug-induced apoptosis. After exogenous overexpression, Bcl-xAK was localized both in mitochondrial and in cytosolic cell fractions. By these findings, a completely new class of Bcl-2-related proteins is introduced, which promotes apoptosis independently from the BH3 domain and implies additional, new mechanisms for apoptosis regulation in melanoma cells.


Subject(s)
Alternative Splicing , Apoptosis , Melanoma/pathology , Skin Neoplasms/pathology , bcl-X Protein/physiology , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Cloning, Molecular , Cytosol/metabolism , Doxycycline/pharmacology , HeLa Cells , Humans , Mitochondria/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Subcellular Fractions
13.
Oncogene ; 25(15): 2213-22, 2006 Apr 06.
Article in English | MEDLINE | ID: mdl-16288204

ABSTRACT

Apoptin, a chicken anemia virus-derived protein, selectively induces apoptosis in transformed but not in normal cells, thus making it a promising candidate as a novel anticancer therapeutic. The mechanism of apoptin-induced apoptosis is largely unknown. Here, we report that contrary to previous assumptions, Bcl-2 and Bcl-xL inhibit apoptin-induced cell death in several tumor cell lines. In contrast, deficiency of Bax conferred resistance, whereas Bax expression sensitized cells to apoptin-induced death. Cell death induction by apoptin was associated with cytochrome c release from mitochondria as well as with caspase-3 and -7 activation. Benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone, a broad spectrum caspase inhibitor, was highly protective against apoptin-induced cell death. Apoptosis induced by apoptin required Apaf-1, as immortalized Apaf-1-deficient fibroblasts as well as tumor cells devoid of Apaf-1 were strongly protected. Thus, our data indicate that apoptin-induced apoptosis is not only Bcl-2- and caspase dependent, but also engages an Apaf-1 apoptosome-mediated mitochondrial death pathway.


Subject(s)
Apoptosis , Capsid Proteins/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Apoptotic Protease-Activating Factor 1 , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 3 , Caspase 7 , Caspase Inhibitors , Caspases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Cytochromes c/metabolism , Enzyme Activation , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Male , Mitochondria/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteins/antagonists & inhibitors , Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured , bcl-X Protein/genetics
14.
Cell Death Differ ; 13(4): 619-27, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16322756

ABSTRACT

We report for the first time inactivation of a tissue-specific Bcl-2 homology domain 3 (BH3)-only protein as a common aspect in human cancer. In detail, we show that loss of the BH3-only protein natural born killer (Nbk)/Bcl-2-interacting killer (Bik) is a common feature of clear-cell renal cell carcinoma (RCC). While strong Nbk expression is found in the renal tubuli and the epithelial lining of the glomerula, a consistent loss of Nbk expression was observed in primary RCC tissue and RCC cell lines. Mutation of Nbk is, however, rare, whereas deletion of the Nbk gene at 22q13.2 is frequent. In addition to loss of heterozygosity (LOH), DNA methylation mediates transcriptional silencing of the Nbk gene. The conditional restoration of Nbk/Bik expression led to apoptotic death of RCC but not of nonmalignant renal epithelia. A broader expression analysis of RCC cell lines for BH3-only proteins revealed that loss of Nbk coincides with failure to express Bim, whereas Puma, Bid and BNIP3 are readily detectable and, in case of Puma, inducible by p53. These data delineate a role for defects in BH3-only proteins as tumor suppressors in RCC and may explain at the same time the impressive clinical apoptosis resistance of RCC.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Carcinoma, Renal Cell/metabolism , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/metabolism , Membrane Proteins/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Enzyme Inhibitors/pharmacology , Female , Gene Deletion , Humans , Kidney Neoplasms/genetics , Loss of Heterozygosity , Male , Membrane Proteins/genetics , Middle Aged , Mitochondrial Proteins , RNA, Messenger/metabolism , Transfection , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
15.
Internist (Berl) ; 46(8): 835-6, 838-42, 844-6, 2005 Aug.
Article in German | MEDLINE | ID: mdl-16010519

ABSTRACT

Recent insights in disease pathogenesis and mechanisms of resistance to therapy of malignant tumors provide a rational basis for the development of novel therapeutic strategies that target genetic defects and deregulated signaling events in malignant tumors. In contrast to conventional therapeutics, small molecule inhibitors or monoclonal antibodies allow for a far more selective, targeted therapy of tumors that carry a corresponding target structure or gene expression profile. Molecular therapeutics therefore necessitate clinical deployment of genetic diagnostics for the identification of those patients who have a chance to benefit from these novel targeted therapies.


Subject(s)
Drug Delivery Systems/methods , Gene Targeting/methods , Genetic Therapy/methods , Immunotherapy/methods , Molecular Biology/methods , Molecular Probe Techniques , Neoplasms/diagnosis , Neoplasms/therapy , Animals , Humans , Pharmacogenetics/methods
16.
Cell Death Differ ; 10(4): 461-7, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12719723

ABSTRACT

Gastric cancer is highly refractory to DNA-damaging therapies. We therefore studied both gene mutation and protein expression of p53 and Bax in a cohort of 116 patients with gastric cancer who underwent R0-resection with a curative intent. Bax mutation was independent from severe microsatellite instability (MSI), that is, global mismatch repair deficiency as determined by analysis of BAT-25/BAT-26 microsatellite markers. Thus, Bax-frameshift mutation is a feature of tumors with low MSI. In contrast and as expected, no p53 mutations were observed in the microsatellite instable tumors. p53 Mutation or p53 overexpression did not have an impact on disease prognosis. p53-Inactivation was, however, associated with an extremely poor prognosis in the subgroup of patients with Bax-mutated tumors. Thus, we show for the first time that the combined mutation of p53 and Bax, two key regulators of the mitochondrial apoptosis pathway, results in an extremely aggressive tumor biology and poor clinical prognosis.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma/genetics , Microsatellite Repeats/genetics , Mutation/genetics , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/genetics , Stomach Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Apoptosis/genetics , Carcinoma/diagnosis , Carcinoma/mortality , Cell Transformation, Neoplastic/genetics , Cohort Studies , DNA Repair/genetics , Humans , Prognosis , Stomach Neoplasms/diagnosis , Stomach Neoplasms/mortality , Survival Rate , bcl-2-Associated X Protein
17.
Cell Death Differ ; 10(4): 477-84, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12719725

ABSTRACT

Inactivation of p53 has been shown to correlate with poor prognosis and drug resistance in malignant tumors. Nevertheless, few reports have directly shown such effects in primary tumor cells. Here, we investigated the p53 mutational status in 138 B-CLL samples and compared these findings with drug and gamma-irradiation sensitivity profiles. p53 mutations resulted not only in a shorter survival but, notably also in selective resistance to alkylating agents, fludarabine and gamma-irradiation. In contrast, no such effect was observed for vincristine, anthracyclines and glucocorticoids. Thus, these latter compounds induce cell death at least in part by p53-independent pathways. Interestingly, p53 mutations clustered in patients who had received prior chemotherapy. In fact, we show for the first time that treatment with DNA-damaging alkylating agents correlates with occurrence of p53 mutations in a clinical setting. This finding may explain at least to some extent the development of resistance to second-line anticancer chemotherapy.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , DNA Damage/drug effects , Drug Resistance, Neoplasm/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation/drug effects , Tumor Suppressor Protein p53/drug effects , Vidarabine/analogs & derivatives , Apoptosis/drug effects , Apoptosis/genetics , Chlorambucil/adverse effects , Cyclophosphamide/adverse effects , DNA Damage/genetics , DNA Mutational Analysis , Female , Gamma Rays/therapeutic use , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Male , Middle Aged , Risk Factors , Survival Rate , Tumor Suppressor Protein p53/genetics , Vidarabine/adverse effects
18.
Gene Ther ; 9(21): 1438-46, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12378406

ABSTRACT

Adenoviral gene transfer of immunmodulatory molecules has been employed successfully in tumor vaccination studies to induce rejection of transplanted syngeneic tumors. In contrast, the response observed when treating chemically induced murine tumors is rather limited. The same applies for human malignancies. A number of reasons including poor transduction efficiency or insufficient T cell infiltration have been held accountable for this lack of efficacy. However, little attention has been given to effects of the adenoviral transduction itself on the T cell system. Here, we show that T cells are sensitized for activation-induced cell death after co-culture with adenovirally infected tumor cells. The levels of CD95/Fas ligand or TNF-alpha, both known mediators of activation induced cell death, however were not affected by the presence of adenovirus-infected target cells. Furthermore, supernatant transfer from adenovirally transduced or non-infected tumor cell cultures did not result in increased T cell apoptosis. This suggests that cell contact rather than a soluble factor is responsible for the induction of T cell apoptosis upon co-culture with adenovirally transduced tumor cells. Interestingly, and in line with our previous observations, activation-induced cell death was partially inhibited if T cells were co-cultured with tumor cells adenovirally transduced to express IL-7 and CD80, both molecules having the capacity to prevent T cell apoptosis.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Lymphocyte Activation , Osteosarcoma/therapy , T-Lymphocytes/pathology , Apoptosis , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Coculture Techniques , Genetic Engineering , Humans , Interleukin-7/genetics , Interleukin-7/immunology , Osteosarcoma/virology , Transduction, Genetic , Tumor Cells, Cultured
19.
Leukemia ; 16(6): 1035-44, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12040435

ABSTRACT

In B-CLL, non-proliferating B cells accumulate due to defective apoptosis. Cytotoxic therapies trigger apoptosis and deregulation of apoptotic pathways contributes to chemoresistance. Loss of the apoptosis-promoting Bax has been implicated in resistance to cytotoxic therapy. We therefore evaluated ex vivo drug sensitivity of CLL, producing chemoresponse data which are prognostic indicators for B-CLL, in particular in the case of purine nucleoside analogs. To analyze the underlying mechanisms of drug resistance, we compared endogenous Bax and Bcl-2 expression to ex vivo response to eight drugs, and to survival in 39 B-CLL patients. We found that reduced Bax levels correlated well with ex vivo resistance to traditional B-CLL therapies - anthracyclines, alkylating agents and vincristine (all P < 0.04). Surprisingly, no such relationship was observed for the purine nucleoside analogs or corticosteroids (all P > 0.5). Mutational analysis of p53 could not explain the loss of Bax protein expression. Levels of Bcl-2 were not associated with sensitivity to any drug. In contrast to the ex vivo data, neither Bax or Bcl-2 expression nor doxorubicin sensitivity were associated with increased survival whereas sensitivity to fludarabine correlated with better overall survival (P = 0.031). These findings suggest that the resistance to purine nucleoside analogs and corticosteroids in B-CLL is due to inactivation of pathways different from those activated by anthracyclines, vinca alkaloids and alkylating agents and may be the molecular rationale for the efficacy of purine analogs in this disease.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Proto-Oncogene Proteins/metabolism , Aged , Apoptosis , Chlorambucil/pharmacology , Cladribine/pharmacology , Cyclophosphamide/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Glucocorticoids/pharmacology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Male , Middle Aged , Proto-Oncogene Proteins c-bcl-2/metabolism , Vidarabine/analogs & derivatives , Vidarabine/pharmacology , bcl-2-Associated X Protein
20.
Leukemia ; 15(11): 1735-42, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11681415

ABSTRACT

The stilbene phytochemicals resveratrol and piceatannol have been reported to possess substantial antitumorigenic and antileukemic activities, respectively. Although recent experimental data revealed the proapoptotic potency of resveratrol, the molecular mechanisms underlying the antileukemic activity have not yet been studied in detail. In the present study, we show that resveratrol, as well as the hydroxylated analog piceatannol, are potent inducers of apoptotic cell death in BJAB Burkitt-like lymphoma cells with an ED50 concentration of 25 microM. Further experiments revealed that treatment of BJAB cells with both substances led to a concentration-dependent activation of caspase-3 and mitochondrial permeability transition. Using BJAB cells overexpressing a dominant-negative mutant of the Fas-associated death domain (FADD) adaptor protein to block death receptor-mediated apoptosis, we demonstrate that resveratrol- and piceatannol-induced cell death in these cells is independent of the CD95/Fas signaling pathway. To explore the antileukemic properties of both compounds in more detail, we extended our study to primary, leukemic lymphoblasts. Interestingly, piceatannol but not resveratrol is a very efficient inducer of apoptosis in this ex vivo assay with leukemic lymphoblasts of 21 patients suffering from childhood lymphoblastic leukemia (ALL).


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis , Burkitt Lymphoma/pathology , Leukemia, Lymphoid/pathology , Stilbenes/pharmacology , Adolescent , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/ultrastructure , Caspase 3 , Caspases/metabolism , Cell Nucleus/ultrastructure , Child , Child, Preschool , DNA Fragmentation , Dose-Response Relationship, Drug , Female , Humans , Infant , Male , Membrane Potentials , Mitochondria/physiology , Resveratrol , Signal Transduction , Tumor Cells, Cultured , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...