Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
J Exp Med ; 177(5): 1257-68, 1993 May 01.
Article in English | MEDLINE | ID: mdl-8386741

ABSTRACT

A novel Hodgkin cell line, designated HD-MyZ, was established from the pleural effusion of a 29-yr-old patient with Hodgkin's disease (HD) of nodular sclerosing type. The majority of cells grow adherently and display typical morphological characteristics of Reed-Sternberg (RS) and Hodgkin (H) cells, i.e., large multi- and mononucleated cells with prominent nucleoli. Immunofluorescence analysis revealed a myelomonocytoid immunophenotype (expression of CD13 and CD68, and lack of lymphoid markers). HD-MyZ cells strongly expressed restin, a recently described intermediate filament-associated protein, the expression of which is restricted to H cells, RS cells, and in vitro cultivated peripheral blood monocytes. In addition mRNA expression of c-fms (colony-stimulating factor 1 receptor) could be induced in HD-MyZ cells by phorbol myristate acetate (PMA) stimulation. Southern blot analysis did not detect rearrangement of T cell receptor beta and immunoglobulin H loci, thus demonstrating the lack of lymphoid commitment. HD-MyZ cells were also devoid of Epstein-Barr virus genomes. HD-MyZ cells constitutively express mRNAs for interleukin 1 alpha (IL-1 alpha), IL-1 beta, IL-5, IL-6, IL-7, IL-8, IL-10, IL-1 receptor (type I), and IL-6 receptor. Stimulation of cells with PMA increased mRNA expression as well as the secretion of IL-1 beta, IL-6, and IL-8, and induced the de novo expression of IL-8 receptors. Xenotransplantation into severe combined immunodeficient (SCID) mice by intravenous or subcutaneous inoculation led to development of disseminated tumors with infiltrative and destructive growth. In addition lymphadenopathy, pleural effusion, and infiltration of spleen were observed. Morphological and immunological analysis of tumor cells revealed the same features as HD-MyZ cells. This cell line might be an important tool for understanding the pathogenesis and biology of HD. In addition the SCID mice model might prove helpful in developing new therapeutic strategies.


Subject(s)
Hodgkin Disease/pathology , Tumor Cells, Cultured , Adult , Animals , Antigens, Differentiation, Myelomonocytic/biosynthesis , Base Sequence , Cytokines/metabolism , Genome, Viral , Herpesvirus 4, Human/genetics , Hodgkin Disease/genetics , Hodgkin Disease/immunology , Humans , Immunophenotyping , Mice , Mice, SCID , Molecular Sequence Data , Neoplasm Transplantation , Oligonucleotides , Polymerase Chain Reaction , RNA, Messenger , Reed-Sternberg Cells
2.
J Exp Med ; 183(3): 1205-13, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8642262

ABSTRACT

The transcription factor E2F is regulated during the cell cycle through interactions with the product of the retinoblastoma susceptibility gene and related proteins. It is thought that E2F-mediated gene regulation at the G1/S boundary and during S phase may be one of the rate-limiting steps in cell proliferation. It was reported that in vivo overexpression of E2F-1 in fibroblasts induces S phase entry and leads to apoptosis. This observation suggests that E2F plays a role in both cell cycle regulation and apoptosis. To further understand the role of E2F in cell cycle progression, cell death, and tumor development, we have blocked endogenous E2F activity in HBL-100 cells, derived from nonmalignant human breast epithelium, using dominant-negative mutants under the control of a tetracycline-dependent expression system. We have shown here that induction of dominant-negative mutants led to strong downregulation of transiently transfected E2F-dependent chloramphenicol acetyl transferase reporter constructs and of endogenous c-myc, which has been described as a target gene of the transcription factor E2F/DP. In addition, we have shown that blocking of E2F could efficiently protect from apoptosis induced by serum starvation within a period of 10 d, whereas control cells started to die after 24 h. Surprisingly, blocking of E2F did not alter the rate of proliferation or of DNA synthesis of these cells; this finding indicates that cell-cycle progression could be driven in an E2F-independent manner. In addition, we have been able to show that blocking of endogenous E2F in HBL-100 cells led to rapid induction of tumor growth in severe combined immunodeficiency mice. No tumor growth could be observed in mice that received mock-transfected clones or tetracycline to block expression of the E2F mutant constructs in vivo. Thus, it appears that E2F has a potential tumor-suppressive function under certain circumstances. Furthermore, we provide evidence that dysregulation of apoptosis may be an important step in tumorigenesis.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carrier Proteins , Cell Cycle Proteins , Cell Cycle , DNA-Binding Proteins , Gene Expression Regulation , Genes, myc , Transcription Factors/metabolism , Animals , Apoptosis , Base Sequence , Cell Division , Cell Line , Chloramphenicol O-Acetyltransferase/biosynthesis , DNA Primers , DNA Replication , E2F Transcription Factors , E2F1 Transcription Factor , Female , Fibroblasts , Genes, Dominant , Humans , Kinetics , Mice , Molecular Sequence Data , Neoplasm Transplantation , Polymerase Chain Reaction , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/biosynthesis , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Recombinant Proteins/biosynthesis , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis , Transfection
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
J Clin Invest ; 97(11): 2651-9, 1996 Jun 01.
Article in English | MEDLINE | ID: mdl-8647960

ABSTRACT

We have studied the expression of members of the bcl-2 family in human breast cancer. The expression pattern of these genes in breast cancer tissue samples was compared with the expression pattern in normal breast epithelium. No marked difference with regard to bcl-2 and bcl-xL expression was observed between normal breast epithelium and cancer tissue. In contrast, bax-alpha, a splice variant of bax, which promotes apoptosis, is expressed in high amounts in normal breast epithelium, whereas only weak or no expression could be detected in 39 out of 40 cancer tissue samples examined so far. Of interest, downregulation of bax-alpha was found in different histological subtypes. Furthermore, we transfected bax-alpha into breast cancer cell lines under the control of a tetracycline-dependent expression system. We were able to demonstrate for the first time that induction of bax expression in breast cancer cell lines restores sensitivity towards both serum starvation and APO-I/Fas-triggered apoptosis and significantly reduces tumor growth in SCID mice. Therefore, we propose that dysregulation of apoptosis might contribute to the pathogenesis of breast cancer at least in part due to an imbalance between members of the bcl-2 gene family.


Subject(s)
Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins/biosynthesis , Animals , Base Sequence , Breast/metabolism , Cell Division , Cell Line , DNA Primers , DNA, Neoplasm/biosynthesis , Epithelium/metabolism , Female , Humans , Mice , Mice, SCID , Molecular Sequence Data , Polymerase Chain Reaction , Proto-Oncogene Proteins c-bcl-2 , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Recombinant Proteins/biosynthesis , Reference Values , Transcription, Genetic , Transfection , Tumor Cells, Cultured , bcl-2-Associated X Protein , fas Receptor/physiology
12.
Oncogene ; 10(2): 269-77, 1995 Jan 19.
Article in English | MEDLINE | ID: mdl-7838527

ABSTRACT

The tax protein of Human T-cell leukemia virus type 1 (HTLV-1) is important for the transforming properties of this virus in vitro and is considered to be responsible for the early stages of leukemogenesis in infected hosts. To address the early consequences of HTLV-1 tax function, we have constructed fusion proteins containing tax sequence either aminoterminal (taxER) or carboxy-terminal (ERtax) of the hormone binding domain of the human estrogen receptor (ER). Addition of estrogen or the antagonist hydroxytamoxifen to Jurkat T-cells expressing these constructs led to the trans-activation or responsive promoters and upregulation of cell surface markers CD28, CD69 and CD5 but not CD25 (IL2R-alpha subunit) or B7 (ligand for CD28). Additional stimulation of the T-cell receptor CD3 complex, led to the upregulation of CD25. B7 was upregulated by concomittent activation of ERtax and CD3 or CD28 pathways. These events were in part reversible upon withdrawal of hormone and inactivation of ERtax. Severe inhibition of proliferation, and apoptosis was observed with cells which had been subjected to short term (3 days) activation of the tax fusion proteins and the CD3 complex. Induction of ERtax activity for longer than 3 days promoted cell death independently of CD3 stimulation. Co-stimulation through the CD28 cell surface molecule did not suppress induction of apoptosis.


Subject(s)
Apoptosis , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/metabolism , T-Lymphocytes/immunology , CD28 Antigens/biosynthesis , CD3 Complex/biosynthesis , Cell Division , Estrogens/pharmacology , Flow Cytometry , Gene Products, tax/genetics , Humans , Lymphocyte Activation , Receptors, Estrogen/genetics , Recombinant Proteins , Time Factors , Transcriptional Activation , Tumor Cells, Cultured
13.
Oncogene ; 20(22): 2749-60, 2001 May 17.
Article in English | MEDLINE | ID: mdl-11420687

ABSTRACT

In this study, we asked whether overexpression of caspase-3, a central downstream executioner of apoptotic pathways, might sensitize breast cancer cells with acquired drug resistance (MT1/ADR) to drug-induced apoptosis. As control, we employed caspase-3 negative and caspase-3-transfected MCF-7 cells. Whereas mock-transfected MCF-7 cells were resistant to epirubicin, etoposide and paclitaxel (taxol), the same drugs led to breakdown of nuclear DNA in caspase-3-transfected MCF-7 cells. MT1/ADR cells express low levels of wild type caspase-3 but show defective caspase activation and apoptosis upon drug exposure. These cells also display a less efficient activation of the mitochondrial permeability transition. Caspase-3-transfected MT1/ADR clones showed a 2.8-fold increase in the protein level and a 3.7-fold higher specific enzyme activity. Procaspase-3 overexpression was not toxic and did not affect background apoptosis. Interestingly, procaspase-3-transfected MT1/ADR cells were more sensitive to cytotoxic drugs as compared with vector-transfected controls and DNA fragmentation nearly reached the levels of the original drug sensitive MT1 cells. Thus, overexpression of caspase-3 enhances chemosensitivity especially in situations where activation of the mitochondrial apoptosome is disturbed.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Caspases/metabolism , Neoplasm Proteins/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Caspase 3 , Caspases/genetics , DNA Fragmentation , DNA, Neoplasm , Drug Resistance, Neoplasm , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , Female , Flow Cytometry , Genetic Vectors , Humans , Neoplasm Proteins/genetics , Transfection , Tumor Cells, Cultured/drug effects
14.
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
15.
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
16.
J Clin Oncol ; 19(8): 2272-81, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11304781

ABSTRACT

PURPOSE: We have previously shown that loss of BAX expression is a negative prognostic factor in metastatic colorectal cancer. In the present study, we addressed the prognostic relevance of BAX and its upstream regulator p53 in squamous cell carcinoma (SCC) of the esophagus. Analysis of p16(ink4a/CDKN2) was included because p16(ink4a/CDKN2) and p53 were shown previously to cooperate during induction of cell cycle arrest and apoptosis. PATIENTS AND METHODS: Retrospective analysis of 53 patients with curative intended R0 resection of esophageal SCC was done. Protein expression of BAX, p53, and p16(ink4a/CDKN2) was investigated by immunohistochemistry. In addition, tumor DNA was screened for BAX frameshift mutations by fragment length analysis and for p53 mutations by single-strand conformation polymorphism-polymerase chain reaction. RESULTS: Overall median survival was 13.7 months. Patients with high BAX protein expression had a median survival of 19.5 months versus 8.0 months with low BAX expression (P <.005). High p16(ink4a/CDKN2) protein expression was associated with a median survival of 23.8 months versus 9.7 months with low p16(ink4a/CDKN2) (P =.011). The best survival (median, 45.8 months) was seen in a subgroup of 12 patients whose tumors bore the combination of both favorite phenotypes (ie, high BAX and high p16(ink4a/CDKN2) protein expression). CONCLUSION: In this retrospective investigation, the combined analysis of BAX and p16(ink4a/CDKN2) shows subgroups in SCC of the esophagus with favorable (p16(ink4a/CDKN2)/BAX high expressing) or poor prognosis (loss of p16(ink4a/CDKN2)/loss of BAX). We suggest that such a multimarker analysis of apoptosis pathways could be useful for individualization of therapeutic strategies in the future, and suggest prospective studies to confirm these results.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genes, p53/genetics , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/genetics , Adult , Aged , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Female , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Analysis , bcl-2-Associated X Protein
17.
J Clin Oncol ; 17(5): 1364-74, 1999 May.
Article in English | MEDLINE | ID: mdl-10334520

ABSTRACT

PURPOSE: To determine the prognostic value of the central downstream apoptosis effector BAX in relation to its upstream regulator p53 in R0-resected hepatic metastases of colorectal cancer. PATIENTS AND METHODS: Retrospective analysis of 41 patients who underwent potentially curative resection of liver metastases from colarectal cancer was performed. Tumor DNA was screened for p53 mutations by single-stranded conformational polymorphism polymerase chain reaction and for BAX frameshift mutations by fragment length analysis. Protein expression of BAX, p21, and p53 was investigated by immunohistochemistry. RESULTS: Overall median survival was 40.2 months. Tumors with BAX frameshift mutations were considered microsatellite mutator phenotype-positive and were excluded from further prognostic analyses. Patients with high BAX protein expression had a median survival of 53.6 months compared with 35.4 months for patients with low BAX expression (P < .05). The negative prognostic value of low BAX expression was more evident in those patients with wild-type p53 (median survival, 54.0 v 23.3 months for BAX-negative tumors; P < .01). Low BAX expression was an independent negative prognostic marker in multivariate regression analysis for all patients independent of the p53 status (relative risk, 3.03, P = .03), especially for p53 wild-type tumors (relative risk, 8.21; P = .0095). CONCLUSION: We conclude that low BAX expression is an independent negative prognostic marker in patients with hepatic metastases of colorectal cancer. The best survival was seen in patients with an intact p53-to-BAX pathway; ie, wild-type p53- and BAX-positive tumors. Thus, analysis of apoptosis signaling pathways (here, p53 in concert with its downstream death effector, BAX) might yield more prognostic power in future studies as compared with analysis of single genes such as p53 alone.


Subject(s)
Colorectal Neoplasms/genetics , Frameshift Mutation/genetics , Genes, p53/genetics , Liver Neoplasms/genetics , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p53/metabolism , Adult , Aged , Analysis of Variance , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , Female , Follow-Up Studies , Humans , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Male , Middle Aged , Neoplasm Proteins/metabolism , Prognosis , Proto-Oncogene Proteins/metabolism , Retrospective Studies , Survival Analysis , bcl-2-Associated X Protein
18.
Leukemia ; 14(12): 2035-44, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11187890

ABSTRACT

This report summarizes recent findings in the field of basic and translational apoptosis research which were presented at the 1st Conference on 'Mechanisms of Cell Death and Disease: Advances in Therapeutic Intervention' organized by the European School of Hematology and the University of Texas MD Anderson Cancer Center, 13-17 May, in Dublin, Ireland, and puts them in the context of the literature. Recent discoveries have significantly advanced the understanding of biochemical and genetic requirements of distinct apoptosis pathways (ie mitochondrial, death-receptor and endoplasmic reticulum-mediated apoptosis) and their dysregulation in disease. Progress has been made especially in the elucidation of the mechanisms of action of the Bcl-2 family members, in detail the formation of channels and their regulation in the mitochondrial membranes, conformational changes in Bax and Bak, and crosstalk of death receptor-triggered apoptosis to the mitochondria by activation of Bax via Bid. In addition, novel insights have been gained about the regulation of caspases and novel caspase signaling pathways, such as activation of caspase-12 by the endoplasmic reticulum stress response. Therapeutic applications of apoptosis manipulation include (1) the inhibition of caspases in acute and chronic neurodegenerative diseases, ie stroke, Alzheimer's or Huntington's disease by drugs and (2) sensitization of cancer cells for drug/radiation-induced apoptosis by modulation of survival signals and viral transfer of apoptosis promoting genes.


Subject(s)
Apoptosis , Animals , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Signal Transduction
19.
Leukemia ; 15(7): 1022-32, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11455969

ABSTRACT

Death receptors and their ligands exert important regulatory functions in the maintenance of tissue homeostasis and the physiological regulation of programmed cell death. Currently, six different death receptors are known including tumor necrosis factor (TNF) receptor-1, CD95 (Fas/APO-1), TNF receptor-related apoptosis-mediating protein (TRAMP), TNF-related apoptosis-inducing ligand (TRAIL) receptor-1 and -2, and death receptor-6 (DR6). The signaling pathways by which these receptors induce apoptosis are similar and rely on oligomerization of the receptor by death ligand binding, recruitment of an adapter protein through homophilic interaction of cytoplasmic domains, and subsequent activation of an inducer caspase which initiates execution of the cell death programme. The ability of these receptors and their ligands to kill malignant cells was discovered early and helped to coin the term 'tumor necrosis factor' for the first identified death ligand. This review summarizes the current and rapidly expanding knowledge about the signaling pathways triggered by death receptor/ligand systems, their potency in experimental cancer therapy, and their therapeutic limitations, especially regarding their toxicity for non-malignant cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , Membrane Glycoproteins/physiology , Neoplasms/therapy , fas Receptor/physiology , Animals , Antigens, CD/physiology , Carrier Proteins/physiology , Caspases/physiology , Fas Ligand Protein , Fas-Associated Death Domain Protein , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Member 25 , Receptors, Tumor Necrosis Factor, Type I
20.
Leukemia ; 8(11): 1825-33, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7526086

ABSTRACT

The monoclonal antibody anti-APO-1 induces apoptosis upon triggering the cell surface molecule APO-1 (CD95), a novel member of the tumor necrosis factor/nerve growth factor receptor superfamily. We tested the efficacy of APO-1 mediated apoptosis in a model system of human leukemia in SCID mice. T-ALL cells recovered from SCID mice were sensitive towards anti-APO-1 mediated apoptosis when tested in vitro. In vivo, treatment of leukemia-bearing SCID mice with anti-APO-1 induced programmed cell death in a substantial fraction of T-ALL cells, thus leading to significantly prolonged survival. Anti-APO-1 treatment, however, failed to completely eliminate all leukemic cells. This may be due to resistance towards anti-APO-1 mediated apoptosis in a fraction of T-ALL cells. Thus, identification of cellular programs which determine sensitivity and resistance towards apoptosis may provide new perspectives for rational therapeutic interventions.


Subject(s)
Antigens, Surface/physiology , Apoptosis , Leukemia-Lymphoma, Adult T-Cell/pathology , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, Surface/immunology , Humans , Immunophenotyping , Immunotherapy , Leukemia-Lymphoma, Adult T-Cell/therapy , Mice , Mice, SCID , Neoplasm Transplantation , Survival Analysis , Transplantation, Heterologous , fas Receptor
SELECTION OF CITATIONS
SEARCH DETAIL