Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
Apoptosis ; 16(6): 636-51, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21437721

ABSTRACT

Apoptosis is modulated by extrinsic and intrinsic signaling pathways through the formation of the death receptor-mediated death-inducing signaling complex (DISC) and the mitochondrial-derived apoptosome, respectively. Ino-C2-PAF, a novel synthetic phospholipid shows impressive antiproliferative and apoptosis-inducing activity. Little is known about the signaling pathway through which it stimulates apoptosis. Here, we show that this drug induces apoptosis through proteins of the death receptor pathway, which leads to an activation of the intrinsic apoptotic pathway. Apoptosis induced by Ino-C2-PAF and its glucosidated derivate, Glc-PAF, was dependent on the DISC components FADD and caspase-8. This can be inhibited in FADD--/-- and caspase-8--/-- cells, in which the breakdown of the mitochondrial membrane potential, release of cytochrome c and activation of caspase-9, -8 and -3 do not occur. In addition, the overexpression of crmA, c-Flip or dominant negative FADD as well as treatment with the caspase-8 inhibitor z-IETD-fmk protected against Ino-C2-PAF-induced apoptosis. Apoptosis proceeds in the absence of CD95/Fas-ligand expression and is independent of blockade of a putative death-ligand/receptor interaction. Furthermore, apoptosis cannot be inhibited in CD95/Fas--/-- Jurkat cells. Expression of Bcl-2 in either the mitochondria or the endoplasmic reticulum (ER) strongly inhibited Ino-C2-PAF- and Glc-PAF-induced apoptosis. In conclusion, Ino-C2-PAF and Glc-PAF trigger a CD95/Fas ligand- and receptor-independent atypical DISC that relies on the intrinsic apoptotic pathway via the ER and the mitochondria.


Subject(s)
Apoptosis/drug effects , Caspase 8/metabolism , Fas-Associated Death Domain Protein/metabolism , Mitochondria/metabolism , Neoplasms/physiopathology , Phospholipids/pharmacology , Signal Transduction , Caspase 8/genetics , Enzyme Activation/drug effects , Fas-Associated Death Domain Protein/genetics , Glycosylation , Humans , Jurkat Cells , Mitochondria/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Phospholipids/chemical synthesis , Proto-Oncogene Proteins c-bcl-2/metabolism
3.
J Mol Med (Berl) ; 88(6): 609-22, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20419447

ABSTRACT

Induction of cell death by p14(ARF) is mediated through a Bax/Bak-dependent mitochondrial apoptosis pathway. To investigate the upstream signaling events required for the activation of Bax and/or Bak and to determine the functional impact of de-regulated cell cycle restriction point control in this context, we genetically dissected the impact of BH3-only proteins and the role of the cyclin-dependent kinase (cdk) inhibitor p21(CDKN1). Using isogenic HCT116 colorectal cancer cells, either wild-type or homozygously deleted for the BH3-only protein Puma/bbc3 and/or p21(CDKN1) or p53-reconstituted DU145 prostate cancer cells, we show that p14(ARF)-induced apoptosis is attenuated in the absence of Puma. Upon expression of p14(ARF) in HCT116 cells, Puma is rapidly induced at both the mRNA and protein level. Puma-proficient HCT116 cells undergo apoptotic (nuclear) DNA fragmentation, which is preceded by the N-terminal conformational change of Bax, the breakdown of the mitochondrial membrane potential, and induction of caspase-9 (LEHD)-like and caspase-3/7 (DEVD)-like activities. In contrast, p14(ARF)-induced apoptosis is markedly attenuated in isogenic HCT116 cells bi-allelically deleted for puma. The sensitivity of Puma-deficient cells to p14(ARF)-induced apoptosis is fully restored by functional reconstitution of Puma using a conditional adenoviral expression vector. Notably, the concomitant deletion of p21(CDKN1) strongly enhances p14(ARF)-induced apoptosis in Puma-proficient cells, but not in isogenic Puma-deficient cells. These results indicate that p14(ARF)-induced mitochondrial apoptosis critically depends on the BH3-only protein Puma. In the presence of a functional p53/Puma/Bax-signaling axis, p14(ARF)-triggered apoptosis is enhanced by loss of p21(CDKN1)-mediated cell cycle checkpoint control.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cell Death/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Mitochondria/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p14ARF/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Caspases/metabolism , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/genetics , Enzyme Activation , HCT116 Cells , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
4.
J Cell Biol ; 188(6): 851-62, 2010 Mar 22.
Article in English | MEDLINE | ID: mdl-20308427

ABSTRACT

Tumor necrosis factor (alpha)-related apoptosis-inducing ligand (TRAIL) is a promising anticancer agent that preferentially kills tumor cells with limited cytotoxicity to nonmalignant cells. However, signaling from death receptors requires amplification via the mitochondrial apoptosis pathway (type II) in the majority of tumor cells. Thus, TRAIL-induced cell death entirely depends on the proapoptotic Bcl-2 family member Bax, which is often lost as a result of epigenetic inactivation or mutations. Consequently, Bax deficiency confers resistance against TRAIL-induced apoptosis. Despite expression of Bak, Bax-deficient cells are resistant to TRAIL-induced apoptosis. In this study, we show that the Bax dependency of TRAIL-induced apoptosis is determined by Mcl-1 but not Bcl-xL. Both are antiapoptotic Bcl-2 family proteins that keep Bak in check. Nevertheless, knockdown of Mcl-1 but not Bcl-xL overcame resistance to TRAIL, CD95/FasL and tumor necrosis factor (alpha) death receptor ligation in Bax-deficient cells, and enabled TRAIL to activate Bak, indicating that Mcl-1 rather than Bcl-xL is a major target for sensitization of Bax-deficient tumors for death receptor-induced apoptosis via the Bak pathway.


Subject(s)
Carcinoma/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , bcl-2 Homologous Antagonist-Killer Protein/antagonists & inhibitors , bcl-2-Associated X Protein/deficiency , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Myeloid Cell Leukemia Sequence 1 Protein , TNF-Related Apoptosis-Inducing Ligand/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics
5.
Oncogene ; 24(25): 4052-64, 2005 Jun 09.
Article in English | MEDLINE | ID: mdl-15806163

ABSTRACT

The death ligand TRAIL has been suggested as a suitable biological agent for the selective induction of cell death in cancer cells. Moreover, TRAIL synergizes with DNA-damaging therapies such as chemotherapeutic drugs or ionizing irradiation (IR). Here, we show that synergy of TRAIL and IR, that is, crosssensitization between TRAIL and IR for induction of apoptosis, entirely depends on Bax proficiency in human DU145 and HCT116 carcinoma cells. DU145 prostate carcinoma cells that have lost Bax protein expression due to mutation fail to activate caspase-3 and -9 when exposed to TRAIL and IR. In contrast, TRAIL sensitized for IR-induced apoptosis and vice versa upon reconstitution of Bax expression. Notably, both DU145 and HCT116 still express significant levels of the multidomain proapoptotic Bcl-2 homolog Bak. This indicates that Bak is not sufficient to mediate crosssensitization and synergism between IR and TRAIL. These data clearly establish distinct roles for Bax and Bak in linking the TRAIL death receptor pathway to the mitochondrial apoptosis signaling cascade upon DNA damage by IR.


Subject(s)
Apoptosis/radiation effects , Membrane Glycoproteins/physiology , Mitochondria/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Necrosis Factor-alpha/physiology , Apoptosis/physiology , Apoptosis Regulatory Proteins , Cell Death/radiation effects , Cell Line, Tumor , Cell Membrane Permeability , Gamma Rays , Humans , Intracellular Membranes/physiology , Male , Mitochondria/radiation effects , Prostatic Neoplasms , Proto-Oncogene Proteins c-bcl-2/radiation effects , Signal Transduction/radiation effects , TNF-Related Apoptosis-Inducing Ligand , bcl-2-Associated X Protein
6.
Oncogene ; 24(25): 4114-28, 2005 Jun 09.
Article in English | MEDLINE | ID: mdl-15750619

ABSTRACT

The human INK4a locus encodes two structurally unrelated tumor suppressor proteins, p16 INK4a and p14 ARF (p19 ARF in the mouse), which are frequently inactivated in human cancer. Both the proapoptotic and cell cycle-regulatory functions of p14 ARF were initially proposed to be strictly dependent on a functional p53/mdm-2 tumor suppressor pathway. However, a number of recent reports have implicated p53-independent mechanisms in the regulation of cell cycle arrest and apoptosis induction by p14 ARF. Here, we show that the G1 cell cycle arrest induced by p14 ARF entirely depends on both p53 and p21 in human HCT116 and DU145 carcinoma cells. In contrast, neither loss of p53 nor p21 impaired apoptosis induction by p14 ARF as evidenced by nuclear DNA fragmentation, phosphatidyl serine exposure, and caspase activation, which included caspase-3/7- and caspase-9-like activities. However, lack of functional p21 resulted in the accumulation of cells in G2/M phase of the cell cycle and markedly enhanced p14 ARF-induced apoptosis that was, nevertheless, efficiently inhibited by the cell permeable broad-spectrum caspase inhibitor zVAD-fmk (valyl-alanyl-aspartyl-(O)-methyl)-fluoromethylketone). Thus, loss of cell cycle restriction point control in the absence of p21 may interfere with p14 ARF-induced apoptosis. Finally, these data indicate that the signaling events required for G1 cell cycle arrest and apoptosis induction by p14 ARF dissociate upstream of p53.


Subject(s)
Apoptosis/physiology , Cell Cycle Proteins/genetics , G1 Phase/physiology , Tumor Suppressor Protein p14ARF/physiology , Adenoviridae/genetics , Bromodeoxyuridine , Carcinoma , Caspases/metabolism , Cell Cycle/physiology , Cell Line , Cell Line, Tumor , Colorectal Neoplasms , Cyclin-Dependent Kinase Inhibitor p21 , Gene Deletion , Genetic Vectors , Humans , Kidney
7.
J Biol Chem ; 280(8): 7118-30, 2005 Feb 25.
Article in English | MEDLINE | ID: mdl-15582998

ABSTRACT

The human INK4a gene locus encodes two structurally unrelated tumor suppressor proteins, p16(INK4a) and p14(ARF). Although primarily proposed to require a functional p53.Mdm-2 signaling axis, recently p14(ARF) has been implicated in p53-independent cell cycle regulation. Here we show that p14(ARF) preferentially induces a G(2) arrest in tumor cells lacking functional p53 and/or p21. Expression of p14(ARF) impaired mitotic entry and enforced a primarily cytoplasmic localization of p34(cdc2) that was associated with a decrease in p34(cdc2) kinase activity and reduced p34(cdc2) protein expression. A direct physical interaction between p14(ARF) and p34(cdc2) was, nevertheless, ruled out by lack of co-immunoprecipitation. The p14(ARF)-induced depletion of p34(cdc2) was associated with impaired cdc25C phosphatase expression and a prominent shift to inhibitory Tyr-15-phosphorylation in G(2)-arrested cells lacking either p53, p21, or both. Finally, reconstitution of p34(cdc2) using a constitutively active, phosphorylation-deficient p34(cdc2AF) mutant alleviated this p14(ARF)-induced G(2) arrest, thereby allowing cell cycle progression. Taken together, these data indicate that p14(ARF) arrests cells lacking functional p53/p21 in the G(2) phase of the cell cycle by targeting p34(cdc2) kinase. This may represent an important fail-safe mechanism by which p14(ARF) protects p53/p21-deficient cells from unrestrained proliferation.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/genetics , Down-Regulation , G2 Phase , Tumor Suppressor Protein p14ARF/physiology , Tumor Suppressor Protein p53/genetics , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21 , Gene Expression Regulation, Neoplastic , Humans , Phosphorylation , Tumor Suppressor Protein p53/deficiency , cdc25 Phosphatases/deficiency
8.
Oncogene ; 23(50): 8320-32, 2004 Oct 28.
Article in English | MEDLINE | ID: mdl-15467752

ABSTRACT

The death ligand TRAIL synergizes with DNA-damaging therapies such as chemotherapeutic drugs or ionizing irradiation. Here, we show that the synergism of TRAIL and 5-fluorouracil (5-FU) and cross-sensitization between TRAIL and 5-FU for induction of apoptosis, entirely depend on Bax proficiency in human DU145 and HCT116 carcinoma cells. DU145 prostate carcinoma cells that have lost Bax protein expression due to mutation fail to release cytochrome c and to activate caspase-3 and -9 when exposed to TRAIL and 5-FU. In contrast, TRAIL sensitized for 5-FU-induced apoptosis and vice versa upon reconstitution of Bax expression. Isobolographic analyses of ED50 doses for 5-FU at increasing TRAIL concentrations showed a clear synergism of TRAIL and 5-FU in Bax-expressing cells. In contrast, the effect was merely additive in DU145 cells lacking Bax. Notably, both DU145 and HCT116 Bax-deficient cells still express Bak. This indicates that Bak is not sufficient to mediate cross-sensitization and synergism between 5-FU and TRAIL. Stable overexpression of Bak in DU145 sensitized for epirubicin-induced apoptosis but failed to confer synergy between TRAIL and 5-FU. Moreover, we show by the use of EGFP-tagged Bax and Bak that TRAIL and 5-FU synergistically trigger oligomerization and clustering of Bax but not Bak. These data clearly establish distinct roles for Bax and Bak in linking the TRAIL death receptor pathway to the mitochondrial apoptosis signaling cascade and delineate a higher degree of specificity in signaling for cell death by multidomain Bcl-2 homologs.


Subject(s)
Apoptosis/physiology , Fluorouracil/pharmacology , Membrane Glycoproteins/physiology , Membrane Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Tumor Necrosis Factor-alpha/physiology , Apoptosis Regulatory Proteins , Blotting, Western , Caspases/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Enzyme Activation , Humans , Male , TNF-Related Apoptosis-Inducing Ligand , bcl-2 Homologous Antagonist-Killer Protein , bcl-2-Associated X Protein
9.
Oncogene ; 21(20): 3149-61, 2002 May 09.
Article in English | MEDLINE | ID: mdl-12082630

ABSTRACT

The human INK4a gene locus encodes two structurally unrelated tumor suppressor proteins, p16(INK4a) and p14(ARF), which are frequently inactivated in human cancer. Whereas p16(INK4a) acts through engagement of the Rb-cdk4/6-cyclin D pathway, both the pro-apoptotic and cell cycle-regulatory functions of p14(ARF) were shown to be primarily dependent on the presence of functional p53. Recent reports have also implicated p14(ARF) in p53-independent mechanisms of cell cycle regulation and apoptosis induction, respectively. To further explore the pro-apoptotic function of p14(ARF) in relation to functional cellular p53, we constructed a replication-deficient adenoviral vector for overexpression of p14(ARF) (Ad-p14(ARF)). As expected, Ad-p14(ARF) efficiently induced apoptosis in p53/Rb wild-type U-2OS osteosarcoma cells at low multiplicities of infection. Interestingly, Ad-p14(ARF) also induced apoptosis in both p53-deleted SAOS-2 osteosarcoma cells and HCT116 colon cancer cells with a bi-allelic knock-out of p53 (HCT116-p53(-/-)). Similarly, adenovirus-mediated overexpression of p14(ARF) induced apoptosis in p53/Bax-mutated DU145 prostate cancer cells as well as in HCT116 cells devoid of functional Bax (HCT116-Bax(-/-)). Restoration of Bax expression by retroviral gene transfer in DU145 cells did not further enhance p14(ARF)-triggered cell death. Infection with Ad-p14(ARF) induced activation of mitochondrial permeability shift transition, caspase activation and apoptotic DNA fragmentation irrespective of the presence or absence of either Bax or functional cellular p53. Nevertheless, overexpression of the anti-apoptotic Bcl-2 homolog Bcl-x(L) markedly inhibited p14(ARF)-induced apoptosis. This may indicate that p14(ARF) triggers a so far unknown activator of mitochondrial apoptosis which can be inhibited by Bcl-2 but which acts either independently or downstream of Bax. Taken together, this report demonstrates the participation of signaling pathways apart from the p53/Mdm-2 rheostat and Bax in p14(ARF)-mediated apoptosis.


Subject(s)
Apoptosis/physiology , Proto-Oncogene Proteins/physiology , Tumor Suppressor Protein p14ARF/physiology , Tumor Suppressor Protein p53/physiology , Adenocarcinoma/pathology , Adenoviridae/genetics , Apoptosis/genetics , Bone Neoplasms/pathology , Gene Expression Regulation, Viral , Genetic Vectors/genetics , Humans , Male , Mitochondria/physiology , Models, Biological , Neoplasm Proteins/physiology , Osteosarcoma/pathology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins c-bcl-2/physiology , Recombinant Fusion Proteins/physiology , Tumor Cells, Cultured , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p53/deficiency , bcl-2-Associated X Protein , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...