Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Leukemia ; 29(11): 2208-20, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26183533

ABSTRACT

AC133 is a prominent surface marker of CD34+ and CD34- hematopoietic stem/progenitor cell (HSPC) subsets. AC133+ HSPCs contain high progenitor cell activity and are capable of hematopoietic reconstitution. Furthermore, AC133 is used for prospective isolation of tumor-initiating cells in several hematological malignancies. Nucleolin is a multifunctional factor of growing and cancer cells, which is aberrantly active in certain hematological neoplasms, and serves as a candidate molecular target for cancer therapy. Nucleolin is involved in gene transcription and RNA metabolism and is prevalently expressed in HSPCs, as opposed to differentiated hematopoietic tissue. The present study dissects nucleolin-mediated activation of surface AC133 and its cognate gene CD133, via specific interaction of nucleolin with the tissue-dependent CD133 promoter P1, as a mechanism that crucially contributes to AC133 expression in CD34+ HSPCs. In mobilized peripheral blood (MPB)-derived HSPCs, nucleolin elevates colony-forming unit (CFU) frequencies and enriches granulocyte-macrophage CFUs. Furthermore, nucleolin amplifies long-term culture-initiating cells and also promotes long-term, cytokine-dependent maintenance of hematopoietic progenitor cells. Active ß-catenin, active Akt and Bcl-2 levels in MPB-derived HSPCs are nucleolin-dependent, and effects of nucleolin on these cells partially rely on ß-catenin activity. The study provides new insights into molecular network relevant to stem/progenitor cells in normal and malignant hematopoiesis.


Subject(s)
Antigens, CD/physiology , Glycoproteins/physiology , Hematopoietic Stem Cells/physiology , Peptides/physiology , Phosphoproteins/physiology , RNA-Binding Proteins/physiology , AC133 Antigen , Antigens, CD/genetics , Cells, Cultured , Glycoproteins/genetics , Humans , Peptides/genetics , Promoter Regions, Genetic , beta Catenin/analysis , beta Catenin/physiology , Nucleolin
2.
Histol Histopathol ; 22(10): 1099-107, 2007 10.
Article in English | MEDLINE | ID: mdl-17616937

ABSTRACT

Heat shock proteins (HSPs) play an important role in the cellular response to environmental stress and exert a cytoprotective effect. Especially HSP70 is an effective inhibitor of apoptosis, suggesting a role of HSP70 in carcinogenesis and tumor progression. To explore the relevance of HSP70 in renal cell carcinomas (RCCs), we analyzed nuclear and cytoplasmic HSP70 protein expression in formalin-fixed tissue from 145 clear cell RCCs by immunohistochemistry as well as Western blot analysis. Nuclear HSP70 expression was found in all RCCs and 75% of the tumors also exhibited a cytoplasmic HSP70 staining. Importantly, RCCs showed significantly reduced cytoplasmic (p=0.001) and combined nuclear/cytoplasmic (p=0.0022) HSP70 expression when compared with their cells of origin. A significant (p=0.0176) decrease of nuclear HSP70 expression became evident from well to poorly differentiated clear cell RCCs. Quite similarly, a trend (p=0.0558) for reduced combined nuclear/cytoplasmic HSP70 expression was shown from early (pT1) to advanced (pT3) tumor stages. Nevertheless, no correlation between HSP70 expression and patients survival became evident. In conclusion, our investigation demonstrates a significant decrease of antiapoptotic HSP70 protein expression during carcinogenesis and during progression from well (G1) to poorly (G3) differentiated clear cell RCCs. Our results suggest that HSP70-mediated inhibition of apoptosis seems to be of minor importance for carcinogenesis and tumor progression in RCCs.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/metabolism , HSP70 Heat-Shock Proteins/metabolism , Kidney Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Cell Count , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cytoplasm/metabolism , Cytoplasm/pathology , Disease Progression , Female , Germany/epidemiology , Humans , Immunoenzyme Techniques , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Rate
3.
Klin Padiatr ; 219(3): 127-33, 2007.
Article in English | MEDLINE | ID: mdl-17525905

ABSTRACT

BACKGROUND: Survivin, a member of the inhibitor of apoptosis protein (IAP) family is transiently expressed at low levels during normal hematopoesis but profoundly overexpressed in adult leukemia potentially contributing to leukemogenesis due to deregulated apoptosis and defective cell cycle control. Alternative splicing results in four different mRNA variants survivin, survivin2B, survivin-DeltaExon3 and survivin-3B, with distinct cellular localization patterns and anti-apoptotic potential. Due to co-localization of survivin and survivin-2B in the cytoplasm survivin-2B may permit interactive fine-tuning of survivin actions and moreover play an attenuating role in its anti-apoptotic function. Lack of survivin-2B is associated with disease progression of malignomas suggesting a differential role of these isoforms in tumorigenesis. PATIENTS AND METHODS: We therefore determined the expression of the functional survivin splice variants performing RT- and real-time PCR in a purely pediatric cohort of 20 patients suffering from precursor B-ALL (BCP-ALL). RESULTS: Here, we demonstrate for the first time in pediatric patients with precursor B-ALL an association between lower survivin-2B expression and affiliation to the high risk group. CONCLUSION: The idea that survivin-2B may act as natural antagonist of survivin could potentially be used in novel approaches of anti-cancer treatment by influencing the proportional expression of the different splice variants.


Subject(s)
Apoptosis/genetics , Genetic Variation/genetics , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Isoforms/genetics , Child , Cohort Studies , Coumarins , Gene Expression Regulation, Leukemic , Humans , Inhibitor of Apoptosis Proteins , Leukocyte Count , Polymerase Chain Reaction , Prognosis , RNA, Messenger/genetics , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors , Survivin
4.
Br J Cancer ; 91(7): 1349-57, 2004 Oct 04.
Article in English | MEDLINE | ID: mdl-15328523

ABSTRACT

Dysregulation of apoptosis plays an important role in tumour progression and resistance to chemotherapy. The X-linked inhibitor of apoptosis (XIAP) is considered to be the most potent caspase inhibitor of all known inhibitor of apoptosis-family members. Only recently, an antagonist of XIAP has been identified, termed Smac/DIABLO. To explore the relevance of antiapoptotic XIAP and proapoptotic Smac/DIABLO for tumour progression in renal cell carcinomas (RCCs), we analysed XIAP and Smac/DIABLO mRNA and protein expression in the primary tumour tissue from 66 RCCs of all major histological types by quantitative real-time PCR, Western blot and ELISA. X-linked inhibitor of apoptosis and Smac/DIABLO mRNA expression was found in all RCCs. Importantly, the relative XIAP mRNA expression levels significantly increased from early (pT1) to advanced (pT3) tumour stages (P=0.0002) and also with tumour dedifferentiation (P=0.04). Western blot analysis confirmed the tumour stage-dependent increase of XIAP expression on the protein level. In contrast, mRNA and protein expression levels of Smac/DIABLO did not significantly change between early and advanced tumour stages or between low and high tumour grades. Consequently, the mRNA expression ratio between antiapoptotic XIAP and proapoptotic Smac/DIABLO markedly increased during progression from early (pT1) to advanced (pT3) tumour stages. Moreover, RCCs confined within the organ capsule (pT1 and pT2) exhibited a significantly lower XIAP to Smac/DIABLO expression ratio when compared with RCCs infiltrating beyond the kidney (pT3; P=0.01). Thus, our investigation demonstrates that the delicate balance between XIAP and Smac/DIABLO expression is gradually disturbed during progression of RCCs, resulting in a relative increase of antiapoptotic XIAP over proapoptotic Smac/DIABLO, thereby probably contributing to the marked apoptosis resistance of RCC.


Subject(s)
Apoptosis/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carrier Proteins/biosynthesis , Gene Expression Profiling , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mitochondrial Proteins/biosynthesis , Protein Biosynthesis , Proteins , Apoptosis Regulatory Proteins , Blotting, Western , Disease Progression , Enzyme-Linked Immunosorbent Assay , Humans , Intracellular Signaling Peptides and Proteins , Polymerase Chain Reaction , X-Linked Inhibitor of Apoptosis Protein , Zinc Fingers
5.
Br J Cancer ; 88(11): 1800-7, 2003 Jun 02.
Article in English | MEDLINE | ID: mdl-12771998

ABSTRACT

TNF-related apoptosis-inducing ligand (TRAIL APO-2L) is a member of the TNF family and induces apoptosis in cancer cells without affecting most non-neoplastic cells. The present investigation is focused on apoptosis induction by combined exposure to TRAIL and ionising radiation (IR) in human renal cell carcinoma (RCC) cell lines. Here, we demonstrate that all RCC cell lines coexpress TRAIL and the death-inducing receptors, TRAIL-R1 and TRAIL-R2. Exposure to TRAIL alone induced marked apoptosis in three out of eight RCC cell lines. Combined exposure to TRAIL and IR resulted in a sensitisation to TRAIL-induced apoptosis in one RCC cell line only. Enhanced apoptosis induction by TRAIL in combination with IR was paralleled by an increase in PARP cleavage and activation of executioner caspase-3, whereas caspases-6 and -7 were not involved. Moreover, exposure to TRAIL and/or IR resulted in a marked activation of initiator caspase-8, possibly augmented by the observed reduction of inhibitory c-FLIP expression. In contrast to other tumour types, activation of initiator caspase-9 was not detectable in our RCC model system after exposure to TRAIL and/or IR. This lack of caspase-9 activation might be related to an impaired 'crosstalk' with the caspase-8 pathway as suggested by the missing Bid cleavage and to the appearance of an XIAP cleavage product known to inhibit caspase-9 activation. Deficient activation of caspase-9, therefore, might contribute to the clinically known resistance of human RCC against IR and also argues against an effective combination therapy with TRAIL and IR in this tumour type.


Subject(s)
Adenocarcinoma, Clear Cell/pathology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Renal Cell/pathology , Caspases/deficiency , Intracellular Signaling Peptides and Proteins , Kidney Neoplasms/pathology , Membrane Glycoproteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Adenocarcinoma, Clear Cell/enzymology , Apoptosis Regulatory Proteins , BH3 Interacting Domain Death Agonist Protein , Blotting, Western , CASP8 and FADD-Like Apoptosis Regulating Protein , Carcinoma, Renal Cell/enzymology , Carrier Proteins/metabolism , Caspase 8 , Caspase 9 , Caspases/metabolism , DNA Primers/chemistry , Gamma Rays , Humans , Kidney Neoplasms/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Polymerase Chain Reaction , Proteins/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects , Tumor Suppressor Protein p53/metabolism , X-Linked Inhibitor of Apoptosis Protein
6.
Br J Cancer ; 88(6): 918-27, 2003 Mar 24.
Article in English | MEDLINE | ID: mdl-12644830

ABSTRACT

Tumour necrosis factor (TNF) related apoptosis-inducing ligand (TRAIL/APO2L) is a recently identified member of the TNF family, which induces programmed cell death in a variety of neoplastic cell types, but not in most nonneoplastic cells. In this study, we report on the identification of two novel alternative splice variants of TRAIL in neoplastic and non-neoplastic human cells lacking either exon 3 (TRAIL-beta) or exons 2 and 3 (TRAIL-gamma). In both splice variants, loss of exon 3 resulted in a frame shift generating a stop codon with consecutive extensive truncation in the extracellular domain. Ectopic expression revealed a loss of proapoptotic potential for both alternative splice variants. In contrast to the predominantly cytoplasmatic localisation of GFP-tagged TRAIL-alpha and TRAIL-beta, TRAIL-gamma showed an additional association with the cell surface and nuclear membrane. In conclusion, alternative splicing might be involved in fine tuning of TRAIL-induced apoptosis and underlines the complexity of the TRAIL system.


Subject(s)
Alternative Splicing , Apoptosis/drug effects , Membrane Glycoproteins/genetics , Tumor Necrosis Factor-alpha/genetics , Apoptosis Regulatory Proteins , Base Sequence , Carcinoma, Renal Cell/pathology , Cytoplasm/chemistry , Exons , Frameshift Mutation , Humans , Kidney Neoplasms/pathology , Ligands , Membrane Glycoproteins/pharmacology , Molecular Sequence Data , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor
7.
Cell Death Differ ; 9(12): 1334-42, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12478470

ABSTRACT

Survivin is an inhibitor of apoptosis protein (IAP) that is markedly overexpressed in most cancers. We identified two novel functionally divergent splice variants, i.e. non-antiapoptotic survivin-2B and antiapoptotic survivin-deltaEx3. Because survivin-2B might be a naturally occurring antagonist of antiapoptotic survivin variants, we analyzed the subcellular distribution of these proteins. PSORT II analysis predicted a preferential cytoplasmic localization of survivin and survivin-2B, but a preferential nuclear localization of survivin-deltaEx3. GFP-tagged survivin variants confirmed the predicted subcellular localization and additionally revealed a cell cycle-dependent nuclear accumulation of survivin-deltaEx3. Moreover, a bipartite nuclear localization signal found exclusively in survivin-deltaEx3 may support cytoplasmic clearance of survivin-deltaEx3. In contrast to the known association between survivin and microtubules or centromeres during mitosis, no corresponding co-localization became evident for survivin-deltaEx3 or survivin-2B. In conclusion, our study provided data on a differential subcellular localization of functionally divergent survivin variants, suggesting that survivin isoforms may perform different functions in distinct subcellular compartments and distinct phases of the cell cycle.


Subject(s)
Alternative Splicing/genetics , Cell Nucleus/metabolism , Eukaryotic Cells/metabolism , Microtubule-Associated Proteins/metabolism , Apoptosis/genetics , Cell Compartmentation/genetics , Cell Nucleus/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , Eukaryotic Cells/cytology , Green Fluorescent Proteins , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Luminescent Proteins , Microtubule-Associated Proteins/genetics , Molecular Sequence Data , Neoplasm Proteins , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Fusion Proteins , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Survivin
8.
Leukemia ; 16(11): 2205-12, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12399963

ABSTRACT

The t(5;14)(q33-34;q11) translocation constitutes a recurrent rearrangement in acute lymphoblastic leukemia involving the T cell receptor (TCR) delta locus on chromosome 14. Breakpoint sequences of the derivative chromosome 5 were isolated by application of a ligation-mediated PCR technique using TCR delta-specific primers to amplify genomic DNA from the leukemic cells of a patient with t(5;14). Through exon trap analysis, we identified various putative exons of the chromosome 5 target gene of the translocation; compilation of sequence information of trapped exons and available expressed sequence tags (ESTs) from the GenBank database allowed us to assemble 1.2 kb of the cDNA. Full-length cDNAs were isolated from a human testis cDNA library and sequence analysis predicted a putative Ran binding protein, a novel member of the importin-beta superfamily of nuclear transport receptors, called RanBP17. The t(5;14) breakpoint maps to the 3' coding region of the gene. The breakpoint of a second t(5;14) positive patient was mapped about 8 kb downstream of the most 3' RanBP17 exon and 2 kb upstream of the first exon of the orphan homeobox gene, Hox11L2. In both cases TCR delta enhancer sequences are juxtaposed downstream of the truncated or intact RanBP17 gene, respectively on the derivative chromosome.


Subject(s)
Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 5/genetics , Genes, T-Cell Receptor delta/genetics , Homeodomain Proteins/genetics , Oncogene Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Recombination, Genetic/genetics , ran GTP-Binding Protein/genetics , Acute Disease , Blotting, Southern , DNA Primers/chemistry , DNA, Neoplasm/analysis , Exons/genetics , Gene Library , Humans , Male , Polymerase Chain Reaction , Proto-Oncogene Proteins , RNA, Neoplasm/analysis , Testis/metabolism , Translocation, Genetic
9.
Br J Cancer ; 86(5): 737-43, 2002 Mar 04.
Article in English | MEDLINE | ID: mdl-11875736

ABSTRACT

Survivin is a novel member of the inhibitor of apoptosis family and determines the susceptibility of tumour cells to pro-apoptotic stimuli. Recently, we identified two novel alternative splice variants of survivin, differing in their anti-apoptotic properties: whereas the anti-apoptotic potential of survivin-DeltaEx3 is preserved, survivin-2B has lost its anti-apoptotic potential and may act as a naturally occurring antagonist of survivin. Because the in vivo expression of these alternative splice variants has not been explored so far, we analysed gastric carcinomas of different histological subtypes, grades and stages. Since no antibodies are currently available to determine the novel splice variants, quantitative reverse transcriptase polymerase chain reaction was performed, using RNA samples obtained from 30 different gastric carcinomas. Polymerase chain reactions products were quantified by densitometric evaluation. We found that all gastric carcinomas, irrespective of their histological types, grades or stages, express survivin-DeltaEx3, survivin-2B and survivin, the latter being the dominant transcript. Comparing the disease stages I+II with III+IV, expression of survivin and survivin-DeltaEx3 remained unchanged. In contrast, a significant (P=0.033) stage-dependent decrease in the expression of survivin-2B became evident. Our study demonstrates for the first time the expression of alternative splice variants in gastric carcinomas and provides a first clue to a role of survivin-2B in tumour progression.


Subject(s)
Apoptosis/genetics , Biomarkers, Tumor/analysis , Carcinoma/genetics , Carcinoma/pathology , Chromosomal Proteins, Non-Histone/biosynthesis , Gene Expression Regulation, Neoplastic , Microtubule-Associated Proteins , RNA Splice Sites , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , DNA Primers , DNA, Neoplasm/genetics , Disease Progression , Humans , Inhibitor of Apoptosis Proteins , Neoplasm Proteins , Neoplasm Staging , Polymerase Chain Reaction , RNA , Survivin
10.
Anticancer Res ; 21(5): 3509-17, 2001.
Article in English | MEDLINE | ID: mdl-11848517

ABSTRACT

BACKGROUND: Since no effective therapeutic approach is yet known for metastatic renal cell carcinoma (RCC), we analyzed the effects of topotecan (Hycamtin), a novel topoisomerase I-inhibitor, in RCC cell lines of the clear cell and papillary/chromophilic types. MATERIALS AND METHODS: The anti-proliferative and apoptosis-inducing effects of topotecan were analyzed in 20 RCC cell lines by MTT-assay and light microscopic apoptosis counting. Moreover, Bcl-2 and Bax expression was investigated by Northern blot and immunocytochemistry while the p53 mutation status was analyzed by DNA sequencing. RESULTS: Exposure to clinically relevant concentrations of topotecan (i.e. < or = 1 microg/ml) resulted in a significant (p<0.05) dose-dependent reduction of cell number in 17 out of 20 RCC cell lines. The reduction of cell number was paralleled by an increase in apoptotic cell death. Papillary/chromophilic RCCs exhibited a significantly (p<0.05) more pronounced responsiveness to topotecan than clear cell RCCs. Moreover, the effects of topotecan proved to be superior to those of 5-fluorouracil (5-FU), an anticancer drug currently used in the therapy of RCCs. No correlation became evident between responsiveness to topotecan and the expression levels of Bcl-2 and Bax. Moreover, the response to topotecan could not be correlated with the p53 mutation status of our RCC cell lines. CONCLUSION: Clinically relevant concentrations of topotecan induced apoptosis in RCC cell lines more effectively than 5-FU. Further testing will show whether topotecan-induced apoptosis can be exploited for the treatment of RCCs in vivo as well.


Subject(s)
Adenocarcinoma, Clear Cell/drug therapy , Antineoplastic Agents/pharmacology , Carcinoma, Papillary/drug therapy , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Topotecan/pharmacology , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/metabolism , Bleomycin/pharmacology , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Drug Screening Assays, Antitumor , Fluorouracil/pharmacology , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Mutation , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein
11.
Br J Pharmacol ; 131(3): 473-81, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11015297

ABSTRACT

The therapeutic use of the antifungal drug amphotericin B (AmB) is limited due to severe side effects like glomerular vasoconstriction and risk of renal failure during AmB administration. As nitric oxide (NO) has substantial functions in renal autoregulation, we have determined the effects of AmB on endothelial constitutive NO synthase (ecNOS) expression and activity in human and rat endothelial cell cultures. AmB used at concentrations of 0.6 to 1.25 microg ml(-1) led to increases in ecNOS mRNA and protein expression as well as NO production. This was the result of an increased ecNOS mRNA half-life. In contrast, incubation of cells with higher albeit subtoxic concentrations of AmB (2.5 - 5.0 microg ml(-1)) resulted in a decrease or respectively in completely abolished ecNOS mRNA and protein expression with a strongly reduced or inhibited ecNOS activity, due to a decrease of ecNOS mRNA half-life. None of the AmB concentrations affected promoter activity as found with a reporter gene construct stably transfected into ECV304 cells. Thus, our experiments show a concentration-dependent biphasic effect of AmB on expression and activity of ecNOS, an effect best explained by AmB influencing ecNOS mRNA stability. In view of the known renal accumulation of this drug the results reported here could help to elucidate its renal toxicity.


Subject(s)
Amphotericin B/pharmacology , Endothelium, Vascular/drug effects , Nitric Oxide Synthase/genetics , RNA Stability/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Cells, Cultured , Cytokines/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Humans , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Promoter Regions, Genetic/drug effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats
13.
Br J Cancer ; 82(11): 1851-9, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10839301

ABSTRACT

The pronounced resistance of human renal cell carcinoma (RCC) to anticancer-induced apoptosis has primarily been related to the expression of P-glycoprotein and effective drug detoxification mechanisms. Because the CD95 system has recently been identified as a key mediator of anticancer drug-induced apoptosis, we analysed the contribution of the CD95 system to chemotherapy-induced apoptosis in four newly established RCC cell lines. Here, we demonstrate that all RCC cell lines expressed CD95-receptor and -ligand. Exposure to agonistic anti-CD95 antibodies resulted in induction of apoptosis and significant (P < 0.05) reduction of cell number in three out of four cell lines, indicating that the essential components for CD95-mediated apoptosis were present and functionally intact in the majority of these RCC cell lines. Moreover, treatment of cultures with bleomycin or topotecan, a novel topoisomerase I inhibitor with little substrate affinity for P-glycoprotein, led to induction of apoptosis and significant (P < 0.05) dose-dependent reduction of cell number in all RCC cell lines. Both anticancer drugs also induced upregulation of CD95 ligand expression in all cell lines. Additionally, augmentation of CD95 receptor expression was found in three RCC cell lines, including one p53-mutated cell line, whereas another p53-mutated cell line showed no or only a weak CD95 receptor upregulation after exposure to topotecan or bleomycin, respectively. Despite this upregulation of CD95 receptor and ligand, antagonistic antibodies directed against CD95 receptors or ligands could not inhibit induction of apoptosis by topotecan and bleomycin in any cell line. Thus, although a functionally intact CD95 signalling cascade is present in most RCC cell lines, the anticancer drugs topotecan and bleomycin that induce upregulation of CD95 receptor and ligand fail to effectively activate CD95-mediated apoptosis. This deficient activation of CD95-mediated apoptosis might be an important additional factor for the multidrug resistance phenotype of human RCCs.


Subject(s)
Apoptosis/physiology , Carcinoma, Renal Cell/pathology , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Kidney Neoplasms/pathology , fas Receptor/physiology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Base Sequence , Carcinoma, Renal Cell/genetics , DNA Primers , Flow Cytometry , Genes, p53 , Humans , Kidney Neoplasms/genetics , Ligands , Mutation , Tumor Cells, Cultured , fas Receptor/genetics
14.
Cell Death Differ ; 7(11): 1127-36, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11139287

ABSTRACT

TRAIL (APO-2L) is a newly identified member of the TNF family and induces apoptosis in cancer cells without affecting most non-neoplastic cells, both in vitro and in vivo. Our study focused on the expression and function of TRAIL and its receptors in renal cell carcinoma (RCC) cell lines of all major histological types. Here, we demonstrate that all RCC cell lines express TRAIL as well as the death-inducing receptors TRAIL-R1 (DR4) and TRAIL-R2 (Killer/DR5). Exposure to TRAIL induced apoptosis in 10 of 16 RCC cell lines. Remarkably, five of six TRAIL-resistant RCC cell lines exhibited high levels of TRAIL expression. Topotecan, a novel topoisomerase I inhibitor, induced upregulation of TRAIL-R2 as well as downregulation of TRAIL. Neutralization of TRAIL with recombinant soluble TRAIL-R1-Fc and TRAIL-R2-Fc failed to inhibit topotecan-induced apoptosis indicating that topotecan-induced cell death can occur in a TRAIL-independent fashion. However, exposure to topotecan resulted in an enhancement of TRAIL-induced apoptosis in all primarily TRAIL-resistant RCC cell lines. This synergistic effect of cotreatment with Topotecan and TRAIL may provide the basis for a new therapeutic approach to induce apoptosis in otherwise unresponsive RCC.


Subject(s)
Apoptosis/drug effects , Carcinoma, Renal Cell/pathology , Enzyme Inhibitors/pharmacology , Kidney Neoplasms/pathology , Membrane Glycoproteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Topotecan/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Apoptosis/physiology , Apoptosis Regulatory Proteins , Carcinoma, Renal Cell/metabolism , Genes, p53 , Humans , Kidney Neoplasms/metabolism , Membrane Glycoproteins/genetics , Mutation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation
15.
Cancer Res ; 59(24): 6097-102, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10626797

ABSTRACT

Recently, a novel antiapoptosis gene, i.e., survivin, was identified as a structurally unique member of the inhibitor of apoptosis protein family. Survivin expression is turned off during fetal development and not found in non-neoplastic adult human tissues but is again turned on in the most common human cancers. The antiapoptotic properties of survivin might provide a significant growth advantage in tumors and possibly also contribute to chemoresistance of cancer. Therefore, we analyzed the expression of survivin in human renal cell carcinomas (RCCs), known to be largely resistant to chemotherapy. Northern blot analysis and RT-PCR revealed survivin expression in newly established RCC cell lines (n = 11) of all major histological types. Moreover, we identified two novel splice variants of survivin, lacking exon 3 (survivin-deltaEx3) or retaining a part of intron 2 as a cryptic exon (survivin-2B). Both sequence alterations cause marked changes in the structure of the corresponding proteins, including structural modifications of the baculovirus inhibitor of apoptosis protein repeat domain. The role of the novel isoforms in the regulation of apoptosis was assessed in transfection experiments, showing conservation of antiapoptotic properties for survivin-deltaEx3 and a markedly reduced antiapoptotic potential for survivin-2B. In conclusion, our observations suggest a complex regulatory balance between the different isoforms of survivin, which might determine the response to proapoptotic stimuli, not only in human RCCs but also in fetal tissues and other types of cancer.


Subject(s)
Alternative Splicing , Apoptosis , Microtubule-Associated Proteins , Proteins/genetics , Amino Acid Sequence , Base Sequence , Carcinoma, Renal Cell/metabolism , DNA, Neoplasm , Humans , Inhibitor of Apoptosis Proteins , Kidney Neoplasms/metabolism , Molecular Sequence Data , Neoplasm Proteins , Protein Biosynthesis , Protein Conformation , Protein Isoforms/biosynthesis , Protein Isoforms/chemistry , Protein Isoforms/genetics , Proteins/chemistry , Proteins/physiology , Sequence Homology, Amino Acid , Survivin , Tumor Cells, Cultured
16.
Lab Invest ; 79(12): 1521-34, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10616203

ABSTRACT

Disorders in negative growth control by CD95 (APO-1/Fas)-mediated apoptosis have been suggested to facilitate immune evasion of neoplastic cells and resistance to anticancer drugs. The present report describes the expression and function of CD95 receptor and ligand in human renal cell carcinomas (RCC) of all major histological types, presenting in vitro data on RCC cell lines (n = 30) and ex vivo observations in RCC tissue samples (n = 30). Using RT-PCR, flow cytometry and immunostaining, expression of CD95 receptor and ligand was found in human RCC of all major histological types. The expression levels of CD95 ligand, however, were rather low. Despite constitutive CD95 receptor expression, resistance to CD95-mediated apoptosis became evident from the weak response to agonistic anti-CD95 antibodies, which induced a low increase of apoptosis in only 9 of 30 RCC cell lines. After IFN-gamma pretreatment, however, apoptosis triggered by agonistic anti-CD95 antibodies was observed in 22 of 30 RCC cell lines. These data indicated that the machinery required for CD95-induced cell death was present, albeit inactive in most RCC. Inhibition of protein synthesis by cycloheximide resulted in increased sensitivity to agonistic anti-CD95 antibodies, suggesting a role for short-lived apoptosis-protective proteins in the resistance of RCC to CD95-mediated apoptosis. Moreover, we demonstrated the secretion of soluble CD95 receptor which might contribute to this resistance as well. In conclusion, resistance rather than responsiveness to CD95-mediated apoptosis is a key feature of human RCC. This resistance might facilitate evasion from negative growth control and contribute to the failure of cytotoxic drugs in the treatment of human RCC.


Subject(s)
Apoptosis/immunology , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , fas Receptor/immunology , Blotting, Northern , Blotting, Western , Cell Division/immunology , Enzyme-Linked Immunosorbent Assay , Fas Ligand Protein , Humans , Immunohistochemistry , Membrane Glycoproteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
17.
Leukemia ; 9(10): 1700-3, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7564513

ABSTRACT

Human acute lymphoblastic leukemia cell lines represent valuable tools to investigate distinct steps of the complex regulatory pathways underlying T cell receptor recombination and expression. A case in point are V delta 2D delta 3 and subsequent V delta 2D delta 3J alpha rearrangements observed in human leukemic pre-B cells as well as in normal lymphopoiesis. The functional expression of these unusual (VD) delta (JC) alpha hybrids is almost exclusively prevented by alternative splicing events. In this report we show that alternative splicing at cryptic splice donor sites within V elements is not a unique feature of hybrid TCR delta/alpha transcripts. Among seven V alpha families analyzed by RT-PCR, alternatively spliced products were observed in TCR alpha recombinations containing V alpha 1 or V alpha 14 elements. In contrast to normal peripheral blood cells and thymocytes, the leukemia cell line JM expressing functional V alpha 1J alpha 3C alpha transcripts lacked evidence of aberrant TCR alpha RNA species.


Subject(s)
Alternative Splicing , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Amino Acid Sequence , Base Sequence , Humans , Molecular Sequence Data , Oligonucleotide Probes/genetics , Sequence Analysis, DNA , Tumor Cells, Cultured
18.
Blood ; 85(7): 1888-96, 1995 Apr 01.
Article in English | MEDLINE | ID: mdl-7535593

ABSTRACT

Lymphocytes expressing alpha beta or gamma delta T-cell receptors (TCR) represent distinct T-cell populations. Because TCR delta genes lie within the TCR alpha locus, the rearrangement processes, transcription, and translation of TCR delta or TCR alpha variable domain exons require tight regulation. Human precursor B-cell leukemias (eg, the REH cell line) constitute an interesting model to study TCR delta/alpha recombination because they rearrange TCR delta/alpha loci along a hierarchically ordered pathway in which V delta 2D delta 3 segments are joined to the J alpha cluster. We now show for REH cells that chimeric TCR delta/alpha variable domain exons are posttranscriptionally modified by alternative splicing resulting in truncated V delta 2C alpha transcripts. This process also takes place during thymic differentiation. CD7+/CD3- T-cell precursors exhibit V delta 2D delta 3 rearrangements. Further differentiation into CD7+/CD3+ thymocytes is associated with the expression of a truncated V delta 2C alpha RNA species. In contrast, chimeric TCR delta/alpha rearrangements containing a V delta 1 segment (but no D delta sequences) are predominantly expressed as full-length V delta 1J alpha C alpha transcripts. These data suggest that alternative splicing constitutes a mechanism that restricts the production of distinct chimeric TCR alpha chains.


Subject(s)
Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor , Gene Rearrangement, delta-Chain T-Cell Antigen Receptor , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Biosynthesis , RNA Splicing , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Antigens, CD/analysis , Antigens, CD7 , Antigens, Differentiation, T-Lymphocyte/analysis , Base Sequence , CD3 Complex/analysis , Cell Differentiation , DNA, Neoplasm/genetics , Humans , Molecular Sequence Data , Neoplasm Proteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Sequence Deletion , T-Lymphocyte Subsets/chemistry , Thymus Gland/cytology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...