Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Biochim Biophys Acta ; 1863(4): 499-509, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26721623

ABSTRACT

The effects of autophagy on cell death are highly contextual and either beneficial or deleterious. One prime example for this dual function of autophagy is evidenced by the cell responses to the BH3 mimetic AT-101 that is known to induce either apoptotic or autophagy-dependent cell death in different settings. Based on previous reports, we hypothesized that the expression levels of pro-survival Bcl-2 family members may be key determinants for the respective death mode induced by AT-101. Here we investigated the role of autophagy in the response of MCF7 breast cancer cells to AT-101. AT-101 treatment induced a prominent conversion of LC3-I to LC3-II and apoptotic cell death characterized by the appearance of Annexin-positive/PI-negative early apoptotic cells and PARP cleavage. Inhibition of the autophagy pathway, either through application of 3-MA or by lentiviral knockdown of ATG5, strongly potentiated cell death, indicating a pro-survival function of autophagy. Overexpression of wild type Bcl-xL significantly diminished the net amount of AT-101-induced cell death, but failed to alter the death-enhancing effects of the ATG5 knockdown. This was also observed with the organelle-specific variants Bcl-xL-ActA and Bcl-2-ActA (mitochondrial) as well as Bcl-xL-cb5 and Bcl-2-cb5 (ER) which all reduced AT-101-induced cell death, but did not affect the death-enhancing effects of 3-MA. Collectively, our data indicate that in apoptosis-proficient MCF7 cells, AT-101 triggers Bcl-2- and Bcl-xL-dependent apoptosis and a cytoprotective autophagy response that is independent of the expression and subcellular localization of Bcl-xL and Bcl-2.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Breast Neoplasms/pathology , Gossypol/analogs & derivatives , Proto-Oncogene Proteins c-bcl-2/genetics , bcl-X Protein/genetics , Apoptosis/genetics , Autophagy/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cytoprotection/drug effects , Cytoprotection/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gossypol/pharmacology , Humans , MCF-7 Cells , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Cells, Cultured , bcl-X Protein/metabolism
2.
Br J Cancer ; 114(2): 188-98, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26657652

ABSTRACT

BACKGROUND: Resistance to temozolomide (TMZ) greatly limits chemotherapeutic effectiveness in glioblastoma (GBM). Here we analysed the ability of the Inhibitor-of-apoptosis-protein (IAP) antagonist birinapant to enhance treatment responses to TMZ in both commercially available and patient-derived GBM cells. METHODS: Responses to TMZ and birinapant were analysed in a panel of commercial and patient-derived GBM cell lines using colorimetric viability assays, flow cytometry, morphological analysis and protein expression profiling of pro- and antiapoptotic proteins. Responses in vivo were analysed in an orthotopic xenograft GBM model. RESULTS: Single-agent treatment experiments categorised GBM cells into TMZ-sensitive cells, birinapant-sensitive cells, and cells that were insensitive to either treatment. Combination treatment allowed sensitisation to therapy in only a subset of resistant GBM cells. Cell death analysis identified three principal response patterns: Type A cells that readily activated caspase-8 and cell death in response to TMZ while addition of birinapant further sensitised the cells to TMZ-induced cell death; Type B cells that readily activated caspase-8 and cell death in response to birinapant but did not show further sensitisation with TMZ; and Type C cells that showed no significant cell death or moderately enhanced cell death in the combined treatment paradigm. Furthermore, in vivo, a Type C patient-derived cell line that was TMZ-insensitive in vitro and showed a strong sensitivity to TMZ and TMZ plus birinapant treatments. CONCLUSIONS: Our results demonstrate remarkable differences in responses of patient-derived GBM cells to birinapant single and combination treatments, and suggest that therapeutic responses in vivo may be greatly affected by the tumour microenvironment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Brain Neoplasms/pathology , Dacarbazine/analogs & derivatives , Dipeptides/pharmacology , Glioblastoma/pathology , Indoles/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Animals , Blotting, Western , Caspase 8/drug effects , Caspase 8/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dacarbazine/pharmacology , Drug Resistance, Neoplasm/drug effects , Flow Cytometry , Humans , In Vitro Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Microscopy, Phase-Contrast , Neoplasm Transplantation , Temozolomide , Xenograft Model Antitumor Assays
3.
Apoptosis ; 19(4): 629-42, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24213561

ABSTRACT

Glioblastoma (GBM) is the most aggressive form of primary brain tumour, with dismal patient outcome. Treatment failure is associated with intrinsic or acquired apoptosis resistance and the presence of a highly tumourigenic subpopulation of cancer cells called GBM stem cells. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) has emerged as a promising novel therapy for some treatment-resistant tumours but unfortunately GBM can be completely resistant to TRAIL monotherapy. In this study, we identified Mcl-1, an anti-apoptotic Bcl-2 family member, as a critical player involved in determining the sensitivity of GBM to TRAIL-induced apoptosis. Effective targeting of Mcl-1 in TRAIL resistant GBM cells, either by gene silencing technology or by treatment with R-roscovitine, a cyclin-dependent kinase inhibitor that targets Mcl-1, was demonstrated to augment sensitivity to TRAIL, both within GBM cells grown as monolayers and in a 3D tumour model. Finally, we highlight that two separate pathways are activated during the apoptotic death of GBM cells treated with a combination of TRAIL and R-roscovitine, one which leads to caspase-8 and caspase-3 activation and a second pathway, involving a Mcl-1:Noxa axis. In conclusion, our study demonstrates that R-roscovitine in combination with TRAIL presents a promising novel strategy to trigger cell death pathways in glioblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Caspase 3/metabolism , Caspase 8/metabolism , Cell Line, Tumor/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Enzyme Activation , Gene Silencing , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Purines/pharmacology , Roscovitine
4.
Neuropathol Appl Neurobiol ; 40(2): 205-16, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24117486

ABSTRACT

AIMS: The Far Upstream Element [FUSE] Binding Protein 1 (FUBP1) regulates target genes, such as the cell cycle regulators MYC and p21. FUBP1 is up-regulated in many tumours and acts as an oncoprotein by stimulating proliferation and inhibiting apoptosis. Recently, FUBP1 mutations were identified in approximately 15% of oligodendrogliomas. To date, all reported FUBP1 mutations have been predicted to inactivate FUBP1, which suggests that in contrast to most other tumours FUBP1 may act as a tumour suppressor in oligodendrogliomas. METHODS: As no data are currently available concerning FUBP1 protein levels in gliomas, we examined the FUBP1 expression profiles of human glial tumours by immunohistochemistry and immunofluorescence. We analysed FUBP1 expression related to morphological differentiation, IDH1 and FUBP1 mutation status, 1p/19q loss of heterozygosity (LOH) as well as proliferation rate. RESULTS: Our findings demonstrate that FUBP1 expression levels are increased in all glioma subtypes as compared with normal central nervous system (CNS) control tissue and are associated with increased proliferation. In contrast, FUBP1 immunonegativity predicted FUBP1 mutation with a sensitivity of 100% and a specificity of 90% in our cohort and was associated with oligodendroglial differentiation, IDH1 mutation and 1p/19q loss of heterozygosity (LOH). Using this approach, we detected a to-date undescribed FUBP1 mutation in an oligodendroglioma. CONCLUSION: In summary, our data indicate an association between of FUBP1 expression and proliferation in gliomas. Furthermore, our findings present FUBP1 immunohistochemical analysis as a helpful additional tool for neuropathological glioma diagnostics predicting FUBP1 mutation.


Subject(s)
DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Oligodendroglioma/genetics , Oligodendroglioma/metabolism , Cell Differentiation , Cell Proliferation , Chromosome Deletion , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 19 , Codon, Nonsense , Glioma/genetics , Glioma/metabolism , Humans , Isocitrate Dehydrogenase/genetics , Loss of Heterozygosity , Neurons/metabolism , RNA-Binding Proteins
5.
Biochim Biophys Acta Mol Cell Res ; 1866(2): 175-189, 2019 02.
Article in English | MEDLINE | ID: mdl-30389373

ABSTRACT

Overcoming apoptosis resistance is one major issue in glioblastoma (GB) therapies. Accumulating evidence indicates that resistance to apoptosis in GB is mediated via upregulation of pro-survival BCL2-family members. The synthetic BH3-mimetic ABT-737 effectively targets BCL2, BCL2 like 1 and BCL2 like 2 but still barely affects cell survival which is presumably due to its inability to inhibit myeloid cell leukemia 1 (MCL1). The constitutively active serine/threonine kinase proviral integration site for moloney murine leukemia virus 1 (PIM1) was recently found to be overexpressed in GB patient samples and to maintain cell survival in these tumors. For different GB cell lines, Western Blot, mitochondrial fractionation, fluorescence microscopy, effector caspase assays, flow cytometry, and an adult organotypic brain slice transplantation model were used to investigate the putative PIM1/MCL1 signaling axis regarding potential synergistic effects with ABT-737. We demonstrate that combination of the PIM1 inhibitor SGI-1776 or the pan-PIM kinase inhibitor AZD1208 with ABT-737 strongly sensitizes GB cells to apoptosis. Unexpectedly, this effect was found to be MCL1-independent, but could be partially blocked by caspase 8 (CASP8) inhibition. Remarkably, the analysis of autophagy markers in combination with the observation of massive accumulation and hampered degradation of autophagosomes suggests a completely novel function of PIM1 as a late stage autophagy regulator, maintaining the autophagic flux at the level of autophagosome/lysosome fusion. Our data indicate that PIM1 inhibition and ABT-737 synergistically induce apoptosis in an MCL1-independent but CASP8-dependent manner in GB. They also identify PIM1 as a suitable target for overcoming apoptosis resistance in GB.


Subject(s)
Apoptosis/drug effects , Glioblastoma/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Autophagy/physiology , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cell Survival , Glioma/pathology , Humans , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Nitrophenols/pharmacology , Peptide Fragments/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/physiology , Sulfonamides/pharmacology , Thiazolidines
6.
Oncogene ; 26(12): 1681-92, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-16983338

ABSTRACT

The proteasome has emerged as a novel target for antineoplastic treatment of hematological malignancies and solid tumors, including those of the central nervous system. To identify cell death pathways activated in response to inhibition of the proteasome system in cancer cells, we treated human SH-SY5Y neuroblastoma cells with the selective proteasome inhibitor (PI) epoxomicin (Epoxo). Prolonged exposure to Epoxo was associated with increased levels of poly-ubiquitinylated proteins and p53, release of cytochrome c from the mitochondria, and activation of caspases. Analysis of global gene expression using high-density oligonucleotide microarrays revealed that Epoxo triggered transcriptional activation of the two Bcl-2-homology domain-3-only (BH3-only) genes p53 upregulated modulator of apoptosis (PUMA) and Bim. Subsequent studies in PUMA- and Bim-deficient cells indicated that Epoxo-induced caspase activation and apoptosis was predominantly PUMA-dependent. Further characterization of the transcriptional response to Epoxo in HCT116 human colon cancer cells demonstrated that PUMA induction was p53-dependent; with deficiency in either p53 or PUMA significantly protected HCT116 cells against Epoxo-induced apoptosis. Our data suggest that p53 activation and the transcriptional induction of its target gene PUMA play an important role in the sensitivity of cancer cells to apoptosis induced by proteasome inhibition, and imply that antineoplastic therapies with PIs might be especially useful in cancers with functional p53.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Proteasome Inhibitors , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Base Sequence , Cell Line, Tumor , DNA Primers , Humans , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Protein Folding
7.
Cell Death Differ ; 12(1): 1-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15592359

ABSTRACT

The amyloid precursor protein (APP) has been suggested to regulate gene expression. GeneChip analysis and in vitro kinase assays revealed potent APP-dependent repression of c-Jun, its target gene SPARC and reduced basal c-Jun N-terminal kinase (JNK) activity in PC12 cells overexpressing APP. UV-induced activation of the JNK signalling pathway and subsequent apoptosis were likewise reduced by APP and this effect could be mimicked by the indirect JNK inhibitor CEP-11004. Treatment with a gamma-secretase inhibitor did not affect APP-mediated downmodulation of the JNK signalling pathway, suggesting that the effects might be mediated via alpha-secretase processing of APP. In support of these data, overexpression of the Swedish mutant of APP did not inhibit SPARC expression, UV-induced JNK activation and cell death. Our data suggest an important physiological role of APP and alpha-secretase activity in the control of JNK/c-Jun signalling, target gene expression and cell death activation in response to cytotoxic stress.


Subject(s)
Amyloid beta-Protein Precursor/physiology , Gene Expression Regulation, Neoplastic , JNK Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-jun/genetics , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/genetics , Animals , Apoptosis/physiology , Apoptosis/radiation effects , Aspartic Acid Endopeptidases/metabolism , Carbazoles/pharmacology , Endopeptidases , Humans , Indoles/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , Oligonucleotide Array Sequence Analysis , Osteonectin/genetics , PC12 Cells , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-jun/metabolism , Rats , Signal Transduction/physiology , Transcription Factor AP-1/genetics , Transcription, Genetic/genetics , Ultraviolet Rays
8.
Neuroscience ; 141(4): 1697-701, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16782283

ABSTRACT

In patients suffering from cerebrovascular diseases and traumatic brain damage, increases in serum levels of protein S100B are positively correlated with the severity of the insult. Since high concentrations of S100B have been shown to exert neurotoxic effects, the objective of this study was to characterize the regulatory mechanisms underlying control of S100B release from astrocytes. To that end, we analyzed the kinetics and amount of S100B release in correlation with regulation of S100B gene expression in an in vitro ischemia model. Astrocyte cultures were treated with combined oxygen, serum and glucose deprivation, serum and glucose deprivation or hypoxia alone for 6, 12 and 24 h, respectively. While oxygen, serum and glucose deprivation triggered the most rapid release of S100B, serum and glucose deprivation provoked comparable levels of released S100B at the later time points. In contrast to oxygen, serum and glucose deprivation and serum and glucose deprivation, hypoxia alone elicited only marginal increases in secreted S100B. Parallel analysis of extracellular lactate dehydrogenase and the number of viable cells revealed only moderate cell death in the cultures, indicating that S100B was actively secreted during in vitro ischemia. Interestingly, S100B mRNA expression was potently downregulated after 12 and 24 h of oxygen, serum and glucose deprivation, and prolonged oxygen, serum and glucose deprivation for 48 h was associated with a significant reduction of S100B release at later time intervals, whereas lactate dehydrogenase levels remained constant. Our data suggest that secretion of S100B during the glial response to metabolic injury is an early and active process.


Subject(s)
Astrocytes/metabolism , Brain/cytology , Nerve Growth Factors/metabolism , S100 Proteins/metabolism , Stress, Physiological/metabolism , Animals , Animals, Newborn , Blotting, Northern/methods , Cell Survival , Cells, Cultured , Culture Media, Serum-Free , Gene Expression/physiology , Gene Expression Regulation/physiology , Glucose/deficiency , Hypoxia/metabolism , L-Lactate Dehydrogenase/metabolism , Nerve Growth Factors/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction/methods , S100 Calcium Binding Protein beta Subunit , S100 Proteins/genetics , Time Factors
9.
Oncogene ; 18(51): 7212-8, 1999 Dec 02.
Article in English | MEDLINE | ID: mdl-10602474

ABSTRACT

Dlk (also termed ZIP kinase) is a novel serine/threonine kinase with a unique C-terminal domain that is rich in arginine and contains three putative NLS motifs and a functional lecuine zipper. Dlk is indeed localized in the nucleus where it shows a speckled distribution. To elucidate the biological functions of Dlk, we wanted to identify the signals relevant for nuclear transport and further the nuclear structures which Dlk binds to. Expression of various deletion and point mutations of Dlk as GFP fusion proteins revealed that the leucine zipper is required for association with speckles and the most C-terminal NLS is necessary and sufficient for nuclear transport. Interestingly, a C-terminal deletion mutant defective for nuclear transport exhibited a pronounced colocalization with actin filaments and, even more strikingly, was a very potent inducer of apoptosis. This apoptotic activity was abrogated, however, when this mutant was retargeted to the nucleus via a heterologous NLS from large T, indicating that Dlk only exerts an apoptotic activity in the cytoplasm. To identify the speckle like structures to which Dlk binds we performed immunofluorescence analyses with antibodies directed against representative marker proteins of replication, transcription, or splicing centers. None of these marker proteins revealed a colocalization with Dlk. Instead, we found a partial colocalization with PML bodies which seem to play a key role in regulation of apoptosis. Taken together, these data strongly suggest a functional role for Dlk in control of cell survival which is dependent on its subcellular localization.


Subject(s)
Apoptosis/genetics , Cell Nucleus/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Apoptosis Regulatory Proteins , Biological Transport/genetics , Calcium-Calmodulin-Dependent Protein Kinases , Cell Line , Death-Associated Protein Kinases , Leucine Zippers , Mutation
10.
Oncogene ; 18(51): 7265-73, 1999 Dec 02.
Article in English | MEDLINE | ID: mdl-10602480

ABSTRACT

Dlk/ZIP kinase is a newly discovered serine/threonine kinase which, due to its homology to DAP kinase, was named DAP like kinase, Dlk. This kinase is tightly associated with nuclear structures, it undergoes extensive autophosphorylation and phosphorylates myosin light chain and core histones H3, H2A and H4 in vitro. Moreover, it possesses a leucine zipper which mediates interaction with transcription factor ATF-4, therefore it was called ZIP kinase. We employed the yeast two-hybrid system to identify interaction partners of Dlk that might serve as regulators or targets. Besides ATF-4 and others we found Par-4, a modulator of transcription factor WT1 and mediator of apoptosis. Complex formation between Dlk and Par-4 was confirmed by GST pull-down experiments and kinase reactions in vitro and coexpression experiments in vivo. The interaction domain within Dlk was mapped to an arginine-rich region between residues 338 - 417, rather than to the leucine zipper. Strikingly, coexpression of Dlk and Par-4 lead to relocation of Dlk from the nucleus to the cytoplasm, particularly to actin filaments. These interactions provoked a dramatic reorganization of the cytoskeleton and morphological symptoms of apoptosis, thus suggesting a functional relationship between Dlk and Par-4 in the control of apoptosis.


Subject(s)
Apoptosis/physiology , Carrier Proteins/metabolism , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase Kinases/metabolism , Animals , Apoptosis Regulatory Proteins , Calcium-Calmodulin-Dependent Protein Kinases , Carrier Proteins/genetics , Cell Line, Transformed , Cytoplasm/physiology , Death-Associated Protein Kinases , Escherichia coli , Leucine Zippers , MAP Kinase Kinase Kinases/genetics , Phosphorylation , Rats , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction
11.
Oncogene ; 17(20): 2645-54, 1998 Nov 19.
Article in English | MEDLINE | ID: mdl-9840928

ABSTRACT

We cloned a cDNA coding for a novel serine/threonine kinase, Dlk, a protein of 448 amino acids with a predicted molecular weight of 51.3 kDa. The kinase domain shows 81% amino acid sequence identity to the recently identified DAP kinase (death associated protein kinase) (Deiss et al., Genes & Dev., 9, 15-30, 1995), therefore, the new kinase was called Dlk, for DAP like kinase. Northern analyses revealed a single mRNA species of 1.7 kb which was ubiquitously expressed. However, expression levels varied considerably in different cell lines and tissues. Moreover, expression was downregulated upon UV irradiation. Dlk exhibited autophosphorylation activity, predominantly towards threonine residues and phosphorylated the regulatory subunit of myosin light chain, but in this case exclusively at serine residues. Dlk seems to be tightly associated with insoluble nuclear structures, presumably chromatin, since it was resistant to various rigorous extraction procedures but it was partially released upon DNase I digestion of nuclei. Consistent with this, purified Dlk phosphorylated core histones H3, H2A and H4 as exogenous substrates and endogenous histone H3 in kinase assays with nuclear extracts. Expression as GFP-fusion protein revealed a diffuse as well as a speckled nuclear staining suggesting an association with replication or transcription centers.


Subject(s)
Chromatin/metabolism , Histones/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/isolation & purification , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Cell Line, Transformed , Cells, Cultured , DNA, Complementary/genetics , Death-Associated Protein Kinases , Enzyme Induction/radiation effects , Fibroblasts/enzymology , Leucine Zippers , MAP Kinase Kinase Kinases , Molecular Sequence Data , Myosin Light Chains/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Substrate Specificity , Ultraviolet Rays
12.
J Neurosci ; 21(13): 4551-63, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11426445

ABSTRACT

Exposure of rat hippocampal neurons or human D283 medulloblastoma cells to the apoptosis-inducing kinase inhibitor staurosporine induced rapid cytochrome c release from mitochondria and activation of the executioner caspase-3. Measurements of cellular tetramethylrhodamine ethyl ester fluorescence and subsequent simulation of fluorescence changes based on Nernst calculations of fluorescence in the extracellular, cytoplasmic, and mitochondrial compartments revealed that the release of cytochrome c was preceded by mitochondrial hyperpolarization. Overexpression of the anti-apoptotic protein Bcl-xL, but not pharmacological blockade of outward potassium currents, inhibited staurosporine-induced hyperpolarization and apoptosis. Dissipation of mitochondrial potassium and proton gradients by valinomycin or carbonyl cyanide p-trifluoromethoxy-phenylhydrazone also potently inhibited staurosporine-induced hyperpolarization, cytochrome c release, and caspase activation. This effect was not attributable to changes in cellular ATP levels. Prolonged exposure to valinomycin induced significant matrix swelling, and per se also caused release of cytochrome c from mitochondria. In contrast to staurosporine, however, valinomycin-induced cytochrome c release and cell death were not associated with caspase-3 activation and insensitive to Bcl-xL overexpression. Our data suggest two distinct mechanisms for mitochondrial cytochrome c release: (1) active cytochrome c release associated with early mitochondrial hyperpolarization, leading to neuronal apoptosis, and (2) passive cytochrome c release secondary to mitochondrial depolarization and matrix swelling.


Subject(s)
Apoptosis , Cytochrome c Group/metabolism , Mitochondria/metabolism , Neurons/metabolism , Potassium/metabolism , Animals , Caspase 3 , Caspases/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/pharmacokinetics , Hippocampus/cytology , Hippocampus/metabolism , Immunohistochemistry , Ionophores/pharmacology , Medulloblastoma/metabolism , Neurons/cytology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Protons , Rats , Rats, Inbred F344 , Staurosporine/pharmacology , Transfection , Valinomycin/pharmacology , bcl-X Protein
13.
Neuroscience ; 134(1): 145-53, 2005.
Article in English | MEDLINE | ID: mdl-15964699

ABSTRACT

We investigated the effect of type 1 human immunodeficiency virus (HIV-1) regulatory protein Tat on N-methyl-d-aspartate (NMDA) receptors expressed in Xenopus oocytes by voltage-clamp recording and its role in NMDA-mediated neurotoxicity using cultured rat hippocampal neurons. Tat (0.01-1muM) potentiated NMDA-induced currents of recombinant NMDA receptors. However, in the presence of Zn(2+), the potentiating effect of Tat was much more pronounced, indicating an additional Zn(2+)-related effect on NMDA receptors. Consistently, Tat potentiated currents of the particularly Zn(2+)-sensitive NR1/NR2A NMDA receptor with a higher efficacy, whereas currents from a Zn(2+)-insensitive mutant were only marginally augmented. In addition, chemical-modified Tat, deficient for metal binding, did not reverse Zn(2+)-mediated inhibition of NMDA responses, demonstrating that Tat disinhibits NMDA receptors from Zn(2+)-mediated antagonism by complexing the cation. We therefore investigated the interplay of Tat and Zn(2+) in NMDA-mediated neurotoxicity using cultures of rat hippocampal neurons. Zn(2+) exhibited a prominent rescuing effect when added together with the excitotoxicant NMDA, which could be reverted by the Zn(2+)-chelator tricine. Similar to tricine, Tat enhanced NMDA-mediated neurotoxicity in the presence of neuroprotective Zn(2+) concentrations. Double-staining with antibodies against Tat and the NR1 subunit of the NMDA receptor revealed partial colocalization of the immunoreactivities in membrane patches of hippocampal neurons, supporting the idea of a direct interplay between Tat and glutamatergic transmission. We therefore propose that release of Zn(2+)-mediated inhibition of NMDA receptors by HIV-1 Tat contributes to the neurotoxic effect of glutamate and may participate in the pathogenesis of AIDS-associated dementia.


Subject(s)
Gene Products, tat/metabolism , Gene Products, tat/pharmacology , Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Animals, Newborn , Chromatin , Drug Interactions , Glycine/analogs & derivatives , Glycine/pharmacology , Hippocampus/cytology , Humans , Immunohistochemistry/methods , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Microinjections/methods , Microscopy, Confocal/methods , Mutagenesis/physiology , N-Methylaspartate/pharmacology , Neurons/radiation effects , Oocytes , Patch-Clamp Techniques/methods , Protein Subunits/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/biosynthesis , Toxoids/pharmacology , Xenopus , Zinc/metabolism , Zinc/pharmacology
14.
Oncogene ; 34(40): 5105-13, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25619832

ABSTRACT

Autophagy is a tightly-regulated catabolic process of cellular self-digestion by which cellular components are targeted to lysosomes for their degradation. Key functions of autophagy are to provide energy and metabolic precursors under conditions of starvation and to alleviate stress by removal of damaged proteins and organelles, which are deleterious for cell survival. Therefore, autophagy appears to serve as a pro-survival stress response in most settings. However, the role of autophagy in modulating cell death is highly dependent on the cellular context and its extent. There is an increasing evidence for cell death by autophagy, in particular in developmental cell death in lower organisms and in autophagic cancer cell death induced by novel cancer drugs. The death-promoting and -executing mechanisms involved in the different paradigms of autophagic cell death (ACD) are very diverse and complex, but a draft scenario of the key molecular targets involved in ACD is beginning to emerge. This review provides an up-to-date and comprehensive report on the molecular mechanisms of drug-induced autophagy-dependent cell death and highlights recent key findings in this exciting field of research.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Autophagy/physiology , Neoplasms/drug therapy , Animals , Humans
15.
Cell Death Dis ; 6: e1812, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26158518

ABSTRACT

Breast cancer is a heterogeneous disease and approximately 70% of newly diagnosed breast cancers are estrogen receptor (ER) positive. Out of the two ER types, α and ß, ERα is the only ER that is detectable by immunohistochemistry in breast cancer biopsies and is the predominant subtype expressed in breast tumor tissue. ER-positive tumors are currently treated with anti-hormone therapy to inhibit ER signaling. It is well known that breast cancer cells can develop endocrine resistance and resistance to anti-hormone therapy and this can be facilitated via the autophagy pathway, but so far the description of a detailed autophagy expression profile of ER-positive cancer cells is missing. In the present study, we characterized tumor cell lines ectopically expressing ERα or ERß as well as the breast cancer-derived MCF-7 cell line endogenously expressing ERα but being ERß negative. We could show that ERα-expressing cells have a higher autophagic activity than cells expressing ERß and cells lacking ER expression. Additionally, for autophagy-related gene expression we describe an ERα-specific 'autophagy-footprint' that is fundamentally different to tumor cells expressing ERß or lacking ER expression. This newly described ERα-mediated and estrogen response element (ERE)-independent non-canonical autophagy pathway, which involves the function of the co-chaperone Bcl2-associated athanogene 3 (BAG3), is independent of classical mammalian target of rapamycin (mTOR) and phosphatidylinositol 3 kinase (PI3K) signaling networks and provides stress resistance in our model systems. Altogether, our study uncovers a novel non-canonical autophagy pathway that might be an interesting target for personalized medicine and treatment of ERα-positive breast cancer cells that do not respond to anti-hormone therapy and classical autophagy inhibitors.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Apoptosis Regulatory Proteins/biosynthesis , Breast Neoplasms/genetics , Estrogen Receptor alpha/biosynthesis , Neuroblastoma/genetics , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Autophagy/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/biosynthesis , Estrogen Receptor beta/genetics , Estrogen Replacement Therapy , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Precision Medicine , Signal Transduction
16.
FEBS Lett ; 462(1-2): 187-91, 1999 Nov 26.
Article in English | MEDLINE | ID: mdl-10580117

ABSTRACT

Dlk, also known as ZIP kinase, is a serine/threonine kinase that is tightly associated with nuclear structures. Under certain conditions, which require cytoplasmic localization, Dlk can induce apoptosis. In search for interaction partners that might serve as regulators or targets of this kinase we identified apoptosis antagonizing transcription factor (AATF), a nuclear phosphoprotein of 523 amino acids. The 1.8 kb mRNA seems to be ubiquitously expressed. AATF contains an extremely acidic domain and a putative leucine zipper characteristic of transcription factors. Indeed, a Gal4-BD-AATF fusion protein exhibited strong transactivation activity. Interestingly, AATF interfered with Dlk-induced apoptosis.


Subject(s)
Apoptosis , MAP Kinase Kinase Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins , Transcription Factors/genetics , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinases , Cells, Cultured , DNA, Complementary/analysis , Death-Associated Protein Kinases , Molecular Sequence Data , Nuclear Proteins , Rats , Transcription Factors/metabolism
17.
Eur J Cancer ; 39(2): 249-56, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12509958

ABSTRACT

Dlk/ZIP kinase is one of five members of the death associated protein (DAP) kinase family. DAP kinase is able to induce apoptosis in a p19ARF/p53-dependent manner. We elucidated the potential role of the p19ARF/p53 pathway in Dlk/ZIP kinase-triggered cell death. Overexpression of a constitutively pro-apoptotic form of Dlk/ZIP kinase induced apoptosis in rat fibroblast cells which express wild-type p19ARF and p53. Cell death was characterised by apoptotic membrane blebbing, mitochondrial depolarisation, cytochrome c release and activation of caspase-3. However, Dlk/ZIP kinase-triggered cell death was also observed in p19ARF-deficient and p53-deficient mouse fibroblast cells. Quantitative analysis revealed that the status of p53 had no major influence on cellular susceptibility to Dlk/ZIP kinase-triggered cell death. Loss of p53 did not prevent Dlk/ZIP kinase-induced mitochondrial membrane depolarisation and release of cytochrome c. Furthermore, overexpression of Dlk/ZIP kinase did not lead to an increased expression of pro-apoptotic p53 target genes in either cell line. These data suggest that Dlk/ZIP kinase is able to trigger the mitochondrial apoptosis pathway independent of the p19ARF/p53 signalling pathway.


Subject(s)
Apoptosis/genetics , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Genes, p53/physiology , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p14ARF/physiology , Animals , Apoptosis Regulatory Proteins , Blotting, Western , Cells, Cultured , Cytochrome c Group/metabolism , Death-Associated Protein Kinases , Fluorescent Antibody Technique , Mice , Mice, Inbred BALB C , Mitochondria/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
18.
Neuroscience ; 127(4): 913-20, 2004.
Article in English | MEDLINE | ID: mdl-15312903

ABSTRACT

Increased serum levels of S100B are positively correlated with multiple forms of CNS damage, such as stroke, CNS trauma and neurodegenerative diseases, but also in psychiatric disorders. However, it is currently not known whether increased serum levels of S100B reflect a neuroregenerative or neurodegenerative response. Since glutamate receptor overactivation (excitotoxicity) may contribute to neuronal pathology in psychiatric disorders, we investigated the effect of S100B on N-methyl-d-aspartate (NMDA)-induced neuronal cell death. Here we demonstrate that very low concentrations of S100B significantly protect primary rat hippocampal neurons against NMDA toxicity by activation of transcription factors of the Rel/nuclear factor kappaB (NF-kappaB) family. Further experiments suggest that i) S100B activated expression of the receptor of advanced glycation products (RAGE) gene in neurons and ii) S100B induced a unique composition of the active NF-kappaB complex consisting of the p65 and c-Rel subunits suggesting a novel mechanism for NF-kappaB activation involved in S100B-mediated neuroprotection. Our data suggest that S100B secreted during the glial response to brain injury potently activates p65/c-Rel in a RAGE-dependent manner and may exert neuroprotective and neuroregenerative effects in psychiatric disorders.


Subject(s)
Carrier Proteins/biosynthesis , Excitatory Amino Acid Agonists/toxicity , Hippocampus/drug effects , Neoplasm Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins , Proto-Oncogene Proteins c-rel/biosynthesis , S100 Proteins/pharmacology , Animals , Animals, Newborn , Brain Injuries/genetics , Brain Injuries/metabolism , Carrier Proteins/genetics , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Hippocampus/metabolism , Neoplasm Proteins/genetics , Nerve Growth Factors , Neurons/drug effects , Neurons/metabolism , Proto-Oncogene Proteins c-rel/genetics , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit , Transcription, Genetic/physiology
19.
Cell Death Dis ; 5: e1391, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25165877

ABSTRACT

Accumulating evidence indicates that loss of physiologic amyloid precursor protein (APP) function leads to reduced neuronal plasticity, diminished synaptic signaling and enhanced susceptibility of neurons to cellular stress during brain aging. Here we investigated the neuroprotective function of the soluble APP ectodomain sAPPα (soluble APPα), which is generated by cleavage of APP by α-secretase along the non-amyloidogenic pathway. Recombinant sAPPα protected primary hippocampal neurons and SH-SY5Y neuroblastoma cells from cell death induced by trophic factor deprivation. We show that this protective effect is abrogated in neurons from APP-knockout animals and APP-depleted SH-SY5Y cells, but not in APP-like protein 1- and 2- (APLP1 and APLP2) depleted cells, indicating that expression of membrane-bound holo-APP is required for sAPPα-dependent neuroprotection. Trophic factor deprivation diminished the activity of the Akt survival pathway. Strikingly, both recombinant sAPPα and the APP-E1 domain were able to stimulate Akt activity in wild-type (wt) fibroblasts, SH-SY5Y cells and neurons, but failed to rescue in APP-deficient neurons or fibroblasts. The ADAM10 (a disintegrin and metalloproteinase domain-containing protein 10) inhibitor GI254023X exacerbated neuron death in organotypic (hippocampal) slice cultures of wt mice subjected to trophic factor and glucose deprivation. This cell death-enhancing effect of GI254023X could be completely rescued by applying exogenous sAPPα. Interestingly, sAPPα-dependent Akt induction was unaffected in neurons of APP-ΔCT15 mice that lack the C-terminal YENPTY motif of the APP intracellular region. In contrast, sAPPα-dependent rescue of Akt activation was completely abolished in APP mutant cells lacking the G-protein interaction motif located in the APP C-terminus and by blocking G-protein-dependent signaling with pertussis toxin. Collectively, our data provide new mechanistic insights into the physiologic role of APP in antagonizing neurotoxic stress: they suggest that cell surface APP mediates sAPPα-induced neuroprotection via G-protein-coupled activation of the Akt pathway.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/metabolism , ADAM10 Protein , Amino Acid Motifs , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/genetics , Animals , Cell Line , Cell Survival/drug effects , Dipeptides/pharmacology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Hydroxamic Acids/pharmacology , In Vitro Techniques , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pertussis Toxin/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction/drug effects
20.
Cell Death Dis ; 4: e629, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23681224

ABSTRACT

Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults. GBM cells are highly resistant to apoptosis induced by antitumor drugs and radiotherapy resulting in cancer progression. We assessed whether a systems medicine approach, analysing the ability of tumor cells to execute apoptosis could be utilized to predict the response of GBM patients to treatment. Concentrations of the key proapoptotic proteins procaspase-3, procaspase-9, Smac and Apaf-1 and the antiapopotic protein XIAP were determined in a panel of GBM cell lines and GBM patient tumor resections. These values were used as input for APOPTO-CELL, a systems biological based mathematical model built to predict cellular susceptibility to undergo caspase activation. The modeling was capable of accurately distinguishing between GBM cells that die or survive in response to treatment with temozolomide in 10 of the 11 lines analysed. Importantly the results obtained using GBM patient samples show that APOPTO-CELL was capable of stratifying patients according to their progression-free survival times and predicted the ability of tumor cells to support caspase activation in 16 of the 21 GBM patients analysed. Calculating the susceptibility to apoptosis execution may be a potent tool in predicting GBM patient therapy responsiveness and may allow for the use of APOPTO-CELL in a clinical setting.


Subject(s)
Algorithms , Brain Neoplasms/metabolism , Caspases/metabolism , Glioblastoma/metabolism , Adult , Aged , Antineoplastic Agents, Alkylating/toxicity , Apoptosis Regulatory Proteins , Apoptotic Protease-Activating Factor 1/metabolism , Brain Neoplasms/pathology , Caspase 3/metabolism , Caspase 9/metabolism , Cell Survival/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/toxicity , Disease-Free Survival , Female , Glioblastoma/pathology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Mitochondrial Proteins/metabolism , Temozolomide , X-Linked Inhibitor of Apoptosis Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL