Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cancer Med ; 7(4): 1359-1368, 2018 04.
Article in English | MEDLINE | ID: mdl-29542868

ABSTRACT

Wilms tumors (WT) with WT1 mutations do not respond well to preoperative chemotherapy by volume reduction, suggesting resistance to chemotherapy. The histologic pattern of this tumor subtype indicates an intrinsic mesenchymal differentiation potential. Currently, it is unknown whether cytotoxic treatments can induce a terminal differentiation state as a direct comparison of untreated and chemotherapy-treated tumor samples has not been reported so far. We conducted gene expression profiling of 11 chemotherapy and seven untreated WT1-mutant Wilms tumors and analyzed up- and down-regulated genes with bioinformatic methods. Cell culture experiments were performed from primary Wilms tumors and genetic alterations in WT1 and CTNNB1 analyzed. Chemotherapy induced MYF6 165-fold and several MYL and MYH genes more than 20-fold and repressed many genes from cell cycle process networks. Viable tumor cells could be cultivated when patients received less than 8 weeks of chemotherapy but not in two cases with longer treatments. In one case, viable cells could be extracted from a lung metastasis occurring after 6 months of intensive chemotherapy and radiation. Comparison of primary tumor and metastasis cells from the same patient revealed up-regulation of RELN and TBX2, TBX4 and TBX5 genes and down-regulation of several HOXD genes. Our analyses demonstrate that >8 weeks of chemotherapy can induce terminal myogenic differentiation in WT1-mutant tumors, but this is not associated with volume reduction. The time needed for all tumor cells to achieve the terminal differentiation state needs to be evaluated. In contrast, prolonged treatments can result in genetic alterations leading to resistance.


Subject(s)
Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mutation , WT1 Proteins/genetics , Wilms Tumor/genetics , Wilms Tumor/pathology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor , Cell Cycle/genetics , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/secondary , Muscle Development/genetics , Reelin Protein , Transcription, Genetic , Wilms Tumor/drug therapy
2.
Biochem Pharmacol ; 97(2): 147-57, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26239805

ABSTRACT

Low molecular weight heparin (LMWH) is routinely used for antithrombotic treatment of cancer patients. Preclinical- and clinical data suggest that LMWH has beneficial effects for cancer patients beyond the prevention of thrombosis, i.e. by inhibiting metastasis. It is, however, unclear whether heparin has an impact on the efficiency of chemotherapy in cancer patients. Here we show that a therapeutic dosage of LMWH tinzaparin reverses cisplatin resistance of A2780cis human ovarian cancer cells to the level of sensitive cells. This novel activity of tinzaparin is associated with intense transcriptional reprogramming. Our gene expression profiling experiments revealed that 3776 genes responded to tinzaparin treatment. For this reason tinzaparin has a complex impact on diverse biological processes. We discovered that tinzaparin inhibits the expression of genes that mediate cisplatin resistance of A2780cis cells. In contrast tinzaparin induced the expression of genes that antagonize drug resistance. This activity of tinzaparin is mediated by cell surface proteoglycans, since enzymatic cleavage of heparan sulfates prevented the reversal of cisplatin resistance. These data indicate that cell surface heparan sulfate proteoglycans play an important role for chemotherapy resistance. The results of this study shed a new light on LMWH application in cancer therapy and suggest tinzaparin as promising treatment option of ovarian cancer patients in combination with anticancer drugs. Future clinical trials are needed to validate these findings.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Heparin, Low-Molecular-Weight/therapeutic use , Ovarian Neoplasms , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Female , Heparin, Low-Molecular-Weight/pharmacology , Humans , Ovarian Neoplasms/drug therapy , Tinzaparin
3.
Hum Mol Genet ; 23(15): 3958-74, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24619359

ABSTRACT

The WT1 gene encodes a zinc finger transcription factor important for normal kidney development. WT1 is a suppressor for Wilms tumour development and an oncogene for diverse malignant tumours. We recently established cell lines from primary Wilms tumours with different WT1 mutations. To investigate the function of mutant WT1 proteins, we performed WT1 knockdown experiments in cell lines with a frameshift/extension (p.V432fsX87 = Wilms3) and a stop mutation (p.P362X = Wilms2) of WT1, followed by genome-wide gene expression analysis. We also expressed wild-type and mutant WT1 proteins in human mesenchymal stem cells and established gene expression profiles. A detailed analysis of gene expression data enabled us to classify the WT1 mutations as gain-of-function mutations. The mutant WT1(Wilms2) and WT1(Wilms3) proteins acquired an ability to modulate the expression of a highly significant number of genes from the G2/M phase of the cell cycle, and WT1 knockdown experiments showed that they are required for Wilms tumour cell proliferation. p53 negatively regulates the activity of a large number of these genes that are also part of a core proliferation cluster in diverse human cancers. Our data strongly suggest that mutant WT1 proteins facilitate expression of these cell cycle genes by antagonizing transcriptional repression mediated by p53. We show that mutant WT1 can physically interact with p53. Together the findings show for the first time that mutant WT1 proteins have a gain-of-function and act as oncogenes for Wilms tumour development by regulating Wilms tumour cell proliferation.


Subject(s)
Gene Expression Regulation, Neoplastic , Mutation , Tumor Suppressor Protein p53/genetics , WT1 Proteins/genetics , Wilms Tumor/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Profiling , Gene Knockdown Techniques , Gene Regulatory Networks , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Molecular Sequence Annotation , Primary Cell Culture , Protein Interaction Mapping , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Suppressor Protein p53/metabolism , WT1 Proteins/metabolism , Wilms Tumor/metabolism , Wilms Tumor/pathology
4.
Anticancer Res ; 34(1): 525-30, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24403511

ABSTRACT

The frequent development of cellular resistance to cisplatin in cancer patients is a serious limitation for clinical drug therapy. However, cisplatin resistance is incompletely understood. We have shown that cisplatin-resistant A2780 ovarian cancer cells (A2780cis) can efficiently be eliminated by liposomal cisplatin, which displayed similar cytotoxicity towards both A2780 and A2780cis cells. This may, at least in part, be related to a higher intracellular accumulation of the drug within the resistant cells after liposomal entry. However, the superior cytotoxicity of the liposomal drug was not reflected by DNA platination. This suggests a more complex mode of action of liposomal cisplatin, most likely affecting different signaling pathways. To gain insight into the resistance gene signature, a whole-genome gene expression analysis was performed for A2780cis cells, untreated or treated with half-minimal inhibitory concentration (IC50) of free and liposomal cisplatin. Strong differences in the functional networks affected by free and liposomal cisplatin became evident. p53 was identified as a key factor directing differences in the apoptotic processes. While free cisplatin induced the intrinsic pathway of apoptosis, liposomal cisplatin induced expression of genes of DNA damage pathways and of the extrinsic pathway of apoptosis. These predictions from gene expression data were confirmed at the protein and function level. This sheds new light on liposomal drug carrier approaches in cancer and suggests liposomal cisplatin as a promising strategy for the treatment of cisplatin-resistant ovarian carcinoma.


Subject(s)
Apoptosis/drug effects , Cisplatin/pharmacology , Drug Carriers , Drug Delivery Systems , Drug Resistance, Neoplasm , Liposomes , Ovarian Neoplasms/drug therapy , Caspases/metabolism , Cisplatin/administration & dosage , Female , Humans , Ovarian Neoplasms/pathology , Tumor Cells, Cultured
6.
Cell Commun Signal ; 11: 63, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-24103640

ABSTRACT

BACKGROUND: The Y-box protein-1 (YB-1) fulfills pleiotropic functions relating to gene transcription, mRNA processing, and translation. It remains elusive how YB-1 shuttling into the nuclear and cytoplasmic compartments is regulated and whether limited proteolysis by the 20S proteasome releases fragments with distinct function(s) and subcellular distribution(s). RESULTS: To address these questions, mapping of domains responsible for subcellular targeting was performed. Three nuclear localization signals (NLS) were identified. NLS-1 (aa 149-156) and NLS-2 (aa 185-194) correspond to residues with unknown function(s), whereas NLS-3 (aa 276-292) matches with a designated multimerization domain. Nuclear export signal(s) were not identified. Endoproteolytic processing by the 20S proteasome before glycine 220 releases a carboxy-terminal fragment (CTF), which localized to the nucleus, indicating that NLS-3 is operative. Genotoxic stress induced proteolytic cleavage and nuclear translocation of the CTF. Co-expression of the CTF and full-length YB-1 resulted in an abrogated transcriptional activation of the MMP-2 promoter, indicating an autoregulatory inhibitory loop, whereas it fulfilled similar trans-repressive effects on the collagen type I promoter. CONCLUSION: Compartmentalization of YB-1 protein derivatives is controlled by distinct NLS, one of which targets a proteolytic cleavage product to the nucleus. We propose a model for an autoregulatory negative feedback loop that halts unlimited transcriptional activation.


Subject(s)
Y-Box-Binding Protein 1/metabolism , Animals , Cell Line , Cell Line, Tumor , Cells, Cultured , Humans , Mesangial Cells/metabolism , Nuclear Export Signals , Nuclear Localization Signals/metabolism , Protein Structure, Tertiary , Proteolysis , Rats , Transcription, Genetic , Y-Box-Binding Protein 1/chemistry
7.
Biochem Pharmacol ; 85(8): 1077-90, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23396090

ABSTRACT

Previously we reported that liposomal cisplatin (CDDP) overcomes CDDP resistance of ovarian A2780cis cancer cells (Krieger et al., Int. J. Pharm. 389, 2010, 10-17). Here we find that the cytotoxic activity of liposomal CDDP is not associated with detectable DNA platination in resistant ovarian cancer cells. This suggests that the mode of action of liposomal CDDP is different from the free drug. To gain insight into mechanisms of liposomal CDDP activity, we performed a transcriptome analysis of untreated A2780cis cells, and A2780cis cells in response to exposure with IC50 values of free or liposomal CDDP. A process network analysis of upregulated genes showed that liposomal CDDP induced a highly different gene expression profile in comparison to the free drug. p53 was identified as a key player directing transcriptional responses to free or liposomal CDDP. The free drug induced expression of essential genes of the intrinsic (mitochondrial) apoptosis pathway (BAX, BID, CASP9) most likely through p38MAPK activation. In contrast, liposomal CDDP induced expression of genes from DNA damage pathways and several genes of the extrinsic pathway of apoptosis (TNFRSF10B-DR5, CD70-TNFSF7). It thus appears that liposomal CDDP overcomes CDDP resistance by inducing DNA damage and in consequence programmed cell death by the extrinsic pathway. Predictions from gene expression data with respect to apoptosis activation were confirmed at the protein level by an apoptosis antibody array. This sheds new light on liposomal drug carrier approaches in cancer and suggests liposomal CDDP as promising strategy for the treatment of CDDP resistant ovarian carcinomas.


Subject(s)
Antineoplastic Agents/administration & dosage , Cisplatin/administration & dosage , Gene Expression Profiling , Ovarian Neoplasms/drug therapy , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Female , Humans , Hydroxamic Acids/pharmacology , Liposomes , Tumor Suppressor Protein p53/physiology
8.
Int J Radiat Biol ; 88(12): 961-71, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22494164

ABSTRACT

PURPOSE: We studied the response of human embryonic stem cells (hESC) to the ß-emitter (131)I, which affects the entire cell and to the Auger electron emitter (125)I-deoxyuridine ((125)I-dU), primarily affecting the deoxyribonuleic acid (DNA). The effects were also studied in keratinocytes as a prototype for somatic cells. METHODS: HESC (H1) and human keratinocytes (HaCaT, human) were exposed to (125)I-dU (5 × 10(-5) - 5 MBq/ml) and (131)I-iodide (5 × 10(-5) - 12.5 MBq/ml) and apoptosis was measured by DNA-fragmentation. Cell morphology was studied by light microscopy and electron microscopy. Transcriptional profiling was done on the Agilent oligonucleotide microarray platform. RESULTS: Auger-process induced no apoptosis but a strong transcriptional response in hESC. In contrast, HaCaT cells showed a pronounced induction of apoptosis but only a moderate transcriptional response. Transcriptional response of hESC was similar after (125)I-dU and (131)I treatments, whereas HaCaT cells expressed a much more pronounced response to (125)I-dU than to (131)I. A striking radiation-induced down-regulation of pluripotency genes was observed in hESC whereas in keratinocytes the enriched gene annotations were related primarily to apoptosis, cell division and proliferation. CONCLUSIONS: Human embryonic stem cells respond to ionizing radiation by (125)I-dU and (131)I in a different way compared to keratinocytes. Transcriptional response and gene expression appear to facilitate an escape from programmed cell death by striking a new path which probably leads to cell differentiation.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/radiation effects , Keratinocytes/cytology , Keratinocytes/radiation effects , Apoptosis/radiation effects , Cell Line , DNA Breaks, Double-Stranded/radiation effects , DNA Breaks, Single-Stranded/radiation effects , DNA Fragmentation/radiation effects , Deoxyuridine/chemistry , Dose-Response Relationship, Radiation , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/ultrastructure , Humans , Iodine Radioisotopes , Keratinocytes/metabolism , Keratinocytes/ultrastructure , Transcription, Genetic/radiation effects , Transcriptome/radiation effects
9.
Mol Inform ; 30(11-12): 918-21, 2011 Dec.
Article in English | MEDLINE | ID: mdl-27468146

ABSTRACT

UNLABELLED: Analysis of gene expression profiles is no longer exclusively a task for bioinformatic experts. However, gaining statistically significant results is challenging and requires both biological knowledge and computational know-how. Here we present a novel, user-friendly microarray reporting tool called maRt. The software provides access to bioinformatic resources, like gene ontology terms and biological pathways by use of the DAVID and the BioMart web-service. Results are summarized in structured HTML reports, each presenting a different layer of information. In these report, contents of diverse sources are integrated and interlinked. To speed up processing, maRt takes advantage of the multi-core technology of modern desktop computers by using parallel processing. Since the software is built upon a RCP infrastructure it might be an outset for developers aiming to integrate novel R based applications. AVAILABILITY: Installer, documentation and various kinds of tutorials are available under LGPL license at the website of our institute http://www.pharma.uni-bonn.de/www/mart. This software is free for academic use.

10.
PLoS Genet ; 6(12): e1001231, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-21170361

ABSTRACT

Transcriptional signatures are an indispensible source of correlative information on disease-related molecular alterations on a genome-wide level. Numerous candidate genes involved in disease and in factors of predictive, as well as of prognostic, value have been deduced from such molecular portraits, e.g. in cancer. However, mechanistic insights into the regulatory principles governing global transcriptional changes are lagging behind extensive compilations of deregulated genes. To identify regulators of transcriptome alterations, we used an integrated approach combining transcriptional profiling of colorectal cancer cell lines treated with inhibitors targeting the receptor tyrosine kinase (RTK)/RAS/mitogen-activated protein kinase pathway, computational prediction of regulatory elements in promoters of co-regulated genes, chromatin-based and functional cellular assays. We identified commonly co-regulated, proliferation-associated target genes that respond to the MAPK pathway. We recognized E2F and NFY transcription factor binding sites as prevalent motifs in those pathway-responsive genes and confirmed the predicted regulatory role of Y-box binding protein 1 (YBX1) by reporter gene, gel shift, and chromatin immunoprecipitation assays. We also validated the MAPK-dependent gene signature in colorectal cancers and provided evidence for the association of YBX1 with poor prognosis in colorectal cancer patients. This suggests that MEK/ERK-dependent, YBX1-regulated target genes are involved in executing malignant properties.


Subject(s)
Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Genes, Regulator , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Y-Box-Binding Protein 1/metabolism , Cell Line, Tumor , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Gene Expression Profiling , Humans , Mitogen-Activated Protein Kinase Kinases/genetics , Y-Box-Binding Protein 1/genetics
11.
Cancer Res ; 70(7): 2840-51, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20332234

ABSTRACT

Y-box binding protein-1 (YB-1) is an oncogenic transcription/translation factor expressed in >40% of breast cancers, where it is associated with poor prognosis, disease recurrence, and drug resistance. We questioned whether this may be linked to the ability of YB-1 to induce the expression of genes linked to cancer stem cells such as CD44 and CD49f. Herein, we report that YB-1 binds the CD44 and CD49f promoters to transcriptionally upregulate their expressions. The introduction of wild-type (WT) YB-1 or activated P-YB-1(S102) stimulated the production of CD44 and CD49f in MDA-MB-231 and SUM 149 breast cancer cell lines. YB-1-transfected cells also bound to the CD44 ligand hyaluronan more than the control cells. Similarly, YB-1 was induced in immortalized breast epithelial cells and upregulated CD44. Conversely, silencing YB-1 decreased CD44 expression as well as reporter activity in SUM 149 cells. In mice, expression of YB-1 in the mammary gland induces CD44 and CD49f with associated hyperplasia. Further, activated mutant YB-1(S102D) enhances self-renewal, primary and secondary mammosphere growth, and soft-agar colony growth, which were reversible via loss of CD44 or CD49f. We next addressed the consequence of this system on therapeutic responsiveness. Here, we show that paclitaxel induces P-YB-1(S102) expression, nuclear localization of activated YB-1, and CD44 expression. The overexpression of WT YB-1 promotes mammosphere growth in the presence of paclitaxel. Importantly, targeting YB-1 sensitized the CD44(High)/CD24(Low) cells to paclitaxel. In conclusion, YB-1 promotes cancer cell growth and drug resistance through its induction of CD44 and CD49f.


Subject(s)
Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Hyaluronan Receptors/biosynthesis , Integrin alpha6/biosynthesis , Nuclear Proteins/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Growth Processes/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/genetics , Integrin alpha6/genetics , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Paclitaxel/pharmacology , Y-Box-Binding Protein 1
12.
Hum Mol Genet ; 19(9): 1651-68, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20106868

ABSTRACT

Wilms tumors (WTs) are genetically heterogeneous kidney tumors whose cells of origin are unknown. Tumors with WT1 mutations and concomitant loss of the wild-type allele represent a distinct subgroup, frequently associated with mutations in CTNNB1. Here, we describe the establishment and characterization of long-term cell cultures derived from five individual WTs with WT1 mutations. Three of these tumor cell lines also had CTNNB1 mutations and an activated canonical Wnt signaling pathway as measured by beta-catenin/T cell-specific transcription factor (TCF) transcriptional activity. Four of the five Wilms cell lines had a stable normal karyotype for at least 25 passages, and four lines showed loss of heterozygosity of chromosome 11p due to mitotic recombination in 11p11. Gene expression profiling revealed that the WT cell lines are highly similar to human mesenchymal stem cells (MSCs) and FACS analysis demonstrated the expression of MSC-specific surface proteins CD105, CD90 and CD73. The stem cell like nature of the WT cells is further supported by their adipogenic, chondrogenic, osteogenic and myogenic differentiation potentials. By generating multipotent mesenchymal precursors from paraxial mesoderm (PAM) in tissue culture using embryonal stem cells, gene expression profiles of PAM and MSCs were described. Using these published gene sets, we found coexpression of a large number of genes in WT cell lines, PAM and MSCs. Lineage plasticity is indicated by the simultaneous expression of genes from the mesendodermal and neuroectodermal lineages. We conclude that WTs with WT1 mutations have specific traits of PAM, which is the source of kidney stromal cells.


Subject(s)
Cell Line, Tumor/cytology , Gene Expression Regulation, Neoplastic/genetics , Genes, Wilms Tumor , Kidney Neoplasms/genetics , Mesenchymal Stem Cells/cytology , Mesoderm/metabolism , Wilms Tumor/genetics , Amino Acid Sequence , Cell Lineage/genetics , Chromosomes, Human, Pair 11/genetics , Flow Cytometry , Gene Expression Profiling , Humans , Karyotyping , Loss of Heterozygosity , Lymphoid Enhancer-Binding Factor 1/metabolism , Mesoderm/cytology , Molecular Sequence Data , Mutation/genetics , beta Catenin/genetics
13.
Int J Pharm ; 389(1-2): 10-7, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20060458

ABSTRACT

The clinical application of cisplatin to treat solid tumours is often limited by the development of tumour cell resistance against this cytostatic agent. Although liposomal carriers of cisplatin are currently in clinical development, approaches to functionally overcome cisplatin resistance by liposomes have hardly been reported. We prepared PEGylated cisplatin-containing liposomes with diameters of about 110 nm and targetability to transferrin receptors (TfR) to correlate cisplatin cell uptake with cytotoxicity in sensitive and cisplatin resistant ovarian cancer cells A2780 compared to the free drug. Whereas the cell entry of free cisplatin was reduced by factor 4 after 24h in resistant cells, liposomal uptake was similar in both cell lines and not affected by resistance. Cytotoxicity was clearly related to intracellular platinum levels, which were even higher for liposomal vs. free cisplatin in the resistant cells after 24, 48, and 72 h and slightly lower in the sensitive cells. However, TfR targeting was of less impact on activity in comparison to non-targeted liposomes. Detection of cellular ATP levels within 24h allowed postulations on the intracellular fate of the liposomes. Altogether, this study strongly supports approaches to overcome cisplatin resistance by a liposomal application of the drug.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Delivery Systems , Ovarian Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Biological Transport , Cell Line, Tumor , Chemistry, Pharmaceutical/methods , Cisplatin/administration & dosage , Cisplatin/pharmacokinetics , Drug Carriers/chemistry , Drug Resistance, Neoplasm , Female , Humans , Liposomes , Ovarian Neoplasms/pathology , Polyethylene Glycols/chemistry , Receptors, Transferrin/metabolism , Time Factors
14.
J Pharmacol Exp Ther ; 332(1): 238-47, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19815812

ABSTRACT

The G protein-coupled P2Y(11) receptor is involved in immune system modulation. In-depth physiological evaluation is hampered, however, by a lack of selective and potent ligands. By screening a library of sulfonic and phosphonic acid derivatives at P2Y(11) receptors recombinantly expressed in human 1321N1 astrocytoma cells (calcium and cAMP assays), the selective non-nucleotide P2Y(11) agonist NF546 [4,4'-(carbonylbis(imino-3,1-phenylene-carbonylimino-3,1-(4-methyl-phenylene)carbonylimino))-bis(1,3-xylene-alpha,alpha'-diphosphonic acid) tetrasodium salt] was identified. NF546 had a pEC(50) of 6.27 and is relatively selective for P2Y(11) over P2Y(1), P2Y(2), P2Y(4), P2Y(6), P2Y(12), P2X(1), P2X(2), and P2X(2)-X(3). Adenosine-5'-O-(3-thio)triphosphate (ATPgammaS), a nonhydrolyzable analog of the physiological P2Y(11) agonist ATP, and NF546 use a common binding site as suggested by molecular modeling studies and their competitive behavior toward the nanomolar potency antagonist NF340 [4,4'-(carbonylbis(imino-3,1-(4-methyl-phenylene)carbonylimino))bis(naphthalene-2,6-disulfonic acid) tetrasodium salt] in Schild analysis. The pA(2) of NF340 was 8.02 against ATPgammaS and 8.04 against NF546 (calcium assays). NF546 was further tested for P2Y(11)-mediated effects in monocyte-derived dendritic cells. Similarly to ATPgammaS, NF546 led to thrombospondin-1 secretion and inhibition of lipopolysaccharide-stimulated interleukin-12 release, whereas NF340 inhibited these effects. Further, for the first time, it was shown that ATPgammaS or NF546 stimulation promotes interleukin 8 (IL-8) release from dendritic cells, which could be inhibited by NF340. In conclusion, we have described the first selective, non-nucleotide agonist NF546 for P2Y(11) receptors in both recombinant and physiological expression systems and could show a P2Y(11)-stimulated IL-8 release, further supporting the immunomodulatory role of P2Y(11) receptors.


Subject(s)
Dendritic Cells/drug effects , Diphosphonates/pharmacology , Interleukin-8/metabolism , Naphthalenesulfonates/pharmacology , Purinergic P2 Receptor Agonists , Calcium/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cloning, Molecular , Cyclic AMP/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dose-Response Relationship, Drug , Humans , Ligands , Protein Binding , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X , Recombinant Proteins , Transfection
15.
J Clin Oncol ; 27(36): 6144-51, 2009 Dec 20.
Article in English | MEDLINE | ID: mdl-19901122

ABSTRACT

PURPOSE: To investigate the potential of Y-box-binding protein YB-1, a multifunctional protein linked to tumor aggressiveness and multidrug resistance, to identify patients with breast cancer likely to benefit from dose-intensified chemotherapy regimens. PATIENTS AND METHODS: YB-1 was immunohistochemically determined in 211 primary tumors from the prospective, randomized West German Study Group WSG-AM-01 trial in high-risk (> or = 10 involved lymph-nodes) breast cancer (HRBC). Predictive impact of YB-1 was assessed by multivariate survival analysis, including time-varying factor-therapy interactions. RESULTS: At median follow-up of 61.7 months, patients receiving rapidly cycled tandem high-dose therapy (HD; two cycles [2x] epirubicin 90 mg/m(2) and cyclophosphamide 600 mg/m(2) every 14 days, followed by 2x epirubicin 90 mg/m(2), cyclophosphamide 3,000 mg/m(2), and thiotepa 400 mg/m(2) every 21 days) had better disease-free survival (DFS; hazard ratio [HR] = 0.62; 95% CI, 0.44 to 0.89) and overall survival (OS; HR = 0.59; 95% CI, 0.4 to 0.89) than those receiving conventional dose-dense chemotherapy (DD; 4x epirubicin 90 mg/m(2) and cyclophosphamide 600 mg/m(2), followed by 3x cyclophosphamide 600 mg/m(2), methotrexate 40 mg/m(2), and fluorouracil 600 mg/m(2) every 14 days). High YB-1 was associated with aggressive tumor phenotype (negative steroid hormone receptor status, positive human epidermal growth factor receptor 2 and p53 status, high MIB-1, unfavorable tumor grade) and poor OS (median 78 v 97 months; P = .01). In patients with high YB-1, HD yielded a 63-month median DFS (P = .001) and a 46-month median OS advantage (P = .002) versus DD. In multivariate models, patients with high B-1 receiving HD (v DD) had one third the hazard rate after 20 months for DFS and one sixth after 40 months for OS. CONCLUSION: In a randomized prospective cancer therapy trial, for the first time, a strong predictive impact of YB-1 on survival has been demonstrated: enhanced benefit from HD (v DD) therapy occurs in HRBC with high YB-1. Future trials could therefore address optimal chemotherapeutic strategies,taking YB-1 into account.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Humans , Immunohistochemistry , Middle Aged , Multivariate Analysis , Prospective Studies , Risk Factors , Survival Rate , Treatment Outcome , Y-Box-Binding Protein 1
16.
Cancer Res ; 69(7): 2996-3003, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19318572

ABSTRACT

Platinum plays a central role in the therapy of ovarian cancer, and the emergence of platinum resistance is a major obstacle for clinical management of the disease. We treated A2780 ovarian cancer cells by weekly cycles of cisplatin over a period of 6 months and unveiled that enhanced insulin-like growth factor I receptor (IGF-IR) expression and autocrine IGF-I are associated with hyperactivation of the IGF-IR and phosphatidylinositol-3-OH kinase (PI3K) pathways in cisplatin-resistant cells. IGF-IR expression levels increased during treatment cycles and correlated with cisplatin resistance. Purified IGF-I induced cisplatin resistance in diverse ovarian cancer cell lines, and small molecule inhibitors proved that IGF-IR and PI3K are essential for cisplatin resistance. Similar results were obtained with BG-1 ovarian cancer cells. Cytogenetic and array comparative genomic hybridization analyses revealed selection and de novo formation of chromosomal alterations during resistance development. An analysis of gene expression profiles of primary ovarian carcinomas identified the regulatory subunit PIK3R2 of PI3-kinase as a significant negative prognosis factor for ovarian cancer. We conclude that targeting the IGF-IR and the PI3K pathways is a promising new strategy to treat cisplatin-resistant ovarian carcinomas.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Receptor, IGF Type 1/metabolism , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Chromosome Aberrations , Drug Resistance, Neoplasm , Female , Gene Expression , Humans , In Situ Hybridization, Fluorescence , Insulin-Like Growth Factor I/metabolism , Ovarian Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Receptor, IGF Type 1/biosynthesis , Receptor, IGF Type 1/genetics , Signal Transduction
17.
J Biol Chem ; 283(2): 739-50, 2008 Jan 11.
Article in English | MEDLINE | ID: mdl-17942395

ABSTRACT

The use of platinum complexes for the therapy of breast cancer is an emerging new treatment modality. To gain insight into the mechanisms underlying cisplatin resistance in breast cancer, we used estrogen receptor-positive MCF-7 cells as a model system. We generated cisplatin-resistant MCF-7 cells and determined the functional status of epidermal growth factor receptor (EGFR), MAPK, and AKT signaling pathways by phosphoreceptor tyrosine kinase and phospho-MAPK arrays. The cisplatin-resistant MCF-7 cells are characterized by increased EGFR phosphorylation, high levels of AKT1 kinase activity, and ERK1 phosphorylation. In contrast, the JNK and p38 MAPK modules of the MAPK signaling pathway were inactive. These conditions were associated with inactivation of the p53 pathway and increased BCL-2 expression. We investigated the expression of genes encoding the ligands for the ERBB signaling cascade and found a selective up-regulation of amphiregulin expression, which occurred at later stages of cisplatin resistance development. Amphiregulin is a specific ligand of the EGFR (ERBB1) and a potent mitogen for epithelial cells. After exposure to cisplatin, the resistant MCF-7 cells secreted amphiregulin protein over extended periods of time, and knockdown of amphiregulin expression by specific short interfering RNA resulted in a nearly complete reversion of the resistant phenotype. To demonstrate the generality and importance of our findings, we examined amphiregulin expression and cisplatin resistance in a variety of human breast cancer cell lines and found a highly significant correlation. In contrast, amphiregulin levels did not significantly correlate with cisplatin resistance in a panel of lung cancer cell lines. We have thus identified a novel function of amphiregulin for cisplatin resistance in human breast cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , ErbB Receptors/physiology , Glycoproteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Amphiregulin , Breast Neoplasms , Bromodeoxyuridine/pharmacology , Cell Division/drug effects , Cell Line, Tumor , EGF Family of Proteins , Enzyme-Linked Immunosorbent Assay , ErbB Receptors/drug effects , Female , Humans , Neoplasm Invasiveness , Phosphorylation , Protein Kinases/drug effects , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
18.
Blood ; 111(7): 3714-22, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18006704

ABSTRACT

Current knowledge about molecular mechanisms underlying disease progression and drug resistance in multiple myeloma (MM) is still limited. Here, we analyzed the potential pathogenetic role of the Y-box binding protein YB-1 in MM. YB-1 is a member of the cold-shock domain protein superfamily and involved in various cellular functions such as proliferation. Immunohistochemical analyses revealed that neither normal bone marrow (BM) plasma cells (PCs), premalignant PCs of patients with monoclonal gammopathy of unknown significance (MGUS), nor MM cells with a mature morphology showed expression of YB-1 in situ. In contrast, YB-1 was strongly expressed in situ in normal PC precursor blasts as well as in a MM subset and in vitro in all of the evaluated MM cell lines. The YB-1-expressing MM cells were characterized by an immature morphology and a highly proliferative phenotype as defined by Ki 67 expression. We observed that siRNA-mediated knockdown of YB-1 decreased proliferation and induced apoptosis in MM cells even in the presence of BM stromal cells. Furthermore, we found that overexpression of YB-1 mediated resistance toward doxorubicin-induced apoptosis in MM cells. Thus, YB-1 contributes to disease progression, survival, and drug resistance in MM and might therefore provide an attractive therapeutic target.


Subject(s)
Cell Proliferation , DNA-Binding Proteins/biosynthesis , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Lymphoid Progenitor Cells/metabolism , Multiple Myeloma/metabolism , Nuclear Proteins/biosynthesis , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Ki-67 Antigen/biosynthesis , Ki-67 Antigen/genetics , Lymphoid Progenitor Cells/pathology , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Plasma Cells/metabolism , Plasma Cells/pathology , RNA, Small Interfering/genetics , Stromal Cells/metabolism , Stromal Cells/pathology , Y-Box-Binding Protein 1
19.
Genes Chromosomes Cancer ; 46(12): 1119-28, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17823930

ABSTRACT

Isolated deletions of the long arm of chromosome 5, del(5q), are observed in 10% of myelodysplastic syndromes (MDS) and are associated with a more favorable prognosis, although the clinical course varies considerably. If one or more additional chromosomal aberrations are present, this correlates with a significantly shorter overall survival. To assess the frequency of hidden abnormalities in cases with an isolated cytogenetic del(5q), we have performed a genome wide high resolution 44 K 60mer oligonucleotide array comparative genomic hybridization (aCGH) study using DNA from bone marrow cells of 12 MDS and one AML patient. In one case a single additional hidden 5.6 Mb deletion of 13q14 and in another case multiple larger aberrations involving many chromosomes were found. Fluorescence in situ hybridization demonstrated that aberrations present in 35% of the bone marrow cells can be detected by aCGH. Furthermore with oligonucleotide aCGH the deletion end points in 5q were mapped precisely, revealing a cluster of proximal breakpoints in band q14.3 (n = 8) and a distal cluster between bands q33.2 and q34 (n = 11). This study shows the high resolution of oligonucleotide CGH arrays for precisely mapping genomic alterations and for refinement of deletion end points. In addition, the high sensitivity of this method enables the study of whole bone marrow cells from MDS patients, a disease with a low blast count.


Subject(s)
Chromosome Aberrations , Chromosome Deletion , Chromosomes, Human, Pair 5 , Genome, Human , Myelodysplastic Syndromes/genetics , Oligonucleotide Array Sequence Analysis/methods , Adult , Aged , Bone Marrow Cells/metabolism , Female , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged
20.
J Biol Chem ; 281(31): 22223-22235, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16698799

ABSTRACT

Retinoblastoma protein (Rb) is a multifunctional tumor suppressor, frequently inactivated in certain types of human cancer. Nucleolin is an abundant multifunctional phosphoprotein of proliferating and cancerous cells, recently identified as cell cycle-regulated transcription activator, controlling expression of human papillomavirus type 18 (HPV18) oncogenes in cervical cancer. Here we find that nucleolin is associated with Rb in intact cells in the G1 phase of the cell cycle, and the complex formation is mediated by the growth-inhibitory domain of Rb. Association with Rb inhibits the DNA binding function of nucleolin and in consequence the interaction of nucleolin with the HPV18 enhancer, resulting in Rb-mediated repression of the HPV18 oncogenes. The intracellular distribution of nucleolin in epithelial cells is Rb-dependent, and an altered nucleolin localization in human cancerous tissues results from a loss of Rb. Our findings suggest that deregulated nucleolin activity due to a loss of Rb contributes to tumor development in malignant diseases, thus providing further insights into the molecular network for the Rb-mediated tumor suppression.


Subject(s)
Cell Cycle , Cell Transformation, Neoplastic , Neoplasms/pathology , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Retinoblastoma Protein/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Human papillomavirus 18/genetics , Humans , Protein Binding , Tumor Suppressor Proteins/metabolism , Nucleolin
SELECTION OF CITATIONS
SEARCH DETAIL
...