Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
5.
J Biol Chem ; 286(22): 19576-88, 2011 Jun 03.
Article in English | MEDLINE | ID: mdl-21482821

ABSTRACT

Daxx is involved in transcriptional control and apoptosis. It comprises several domains, including a regulatory C terminus that is responsible for the interaction with numerous proteins such as p53, promyelocytic leukemia protein (PML), and Hsp27. Here, we describe the identification and characterization of two novel variants of Daxx termed Daxx-ß and Daxx-γ, which are generated by alternative splicing. Alternative splicing results in a truncated regulatory C terminus in both proteins. As a consequence, Daxx-ß and Daxx-γ show a markedly decreased affinity to PML, which in turn is associated with a different subnuclear localization of these proteins compared with Daxx. Although Daxx is localized mainly in PML-oncogenic domains (PODs) Daxx-ß and Daxx-γ display a distinct distribution pattern. Furthermore, Daxx-ß and Daxx-γ show a decreased affinity to p53 also due to the truncated C terminus. We provide evidence that the p53 recruitment into PODs is Daxx isoform-dependent. The decreased affinity of Daxx-ß/-γ to p53 and PML results in a diffuse localization of p53 throughout the nucleus. In contrast to Daxx, Daxx-ß and Daxx-γ are unable to repress p53-mediated transcription. Therefore, alternative splicing of Daxx might indicate an additional level in the cellular apoptosis network.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Alternative Splicing/physiology , Apoptosis/physiology , Cell Nucleus/metabolism , Nuclear Proteins/biosynthesis , Repressor Proteins/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Cell Nucleus/genetics , Co-Repressor Proteins , HEK293 Cells , HeLa Cells , Humans , Molecular Chaperones , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Structure, Tertiary , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
Cell Signal ; 23(8): 1358-65, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21440621

ABSTRACT

Retinoblastoma protein (Rb) controls cell proliferation, differentiation, survival and gene expression and it has a central role in the signaling network that provides a cell cycle checkpoint in the G1 phase of the cell cycle. Studies in mice have shown that Rb regulates interactions between hematopoietic stem cells and their bone marrow microenvironment and it acts as a critical regulator of hematopoietic stem and progenitor cells under stress. In human hematopoiesis, the CD34 protein is expressed on a subset of progenitor cells capable of self-renewal, multilineage differentiation, and hematopoietic reconstitution, and CD34 has a role in the differentiation of hematopoietic cells. Here we find that, in CD34-positive hematopoietic cells, Rb controls the human CD34 promoter region by antagonizing the CD34 promoter factor nucleolin to provide a mechanism that links expression of endogenous CD34 to cell cycle progression. Our study suggests a direct involvement of Rb in the transcriptional program of human CD34-positive hematopoietic stem/progenitor cells, thus providing further insights into the molecular network relevant to the features of these cells.


Subject(s)
Antigens, CD34/metabolism , Phosphoproteins/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Retinoblastoma Protein/metabolism , Antigens, CD34/genetics , Cell Line , G1 Phase , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Phosphoproteins/metabolism , Promoter Regions, Genetic , RNA-Binding Proteins/metabolism , S Phase , Nucleolin
7.
Front Biosci (Elite Ed) ; 2(3): 1042-53, 2010 06 01.
Article in English | MEDLINE | ID: mdl-20515774

ABSTRACT

Differential expression of p53 isoforms might participate in the marked resistance towards conventional chemotherapy of renal cell carcinomas (RCCs). Therefore, we analysed their differential expression and regulation in RCCs. RCCs expressed a more p53 activating isoform pattern during tumor initiation and progression, in vivo. In vitro, two cell lines exhibiting a similar sensitivity towards Topotecan-induced cell death revealed a similar induction of p53 target genes but strongly differed in their extent of apoptosis. Furthermore, they strongly differed in their basal expression patterns and differential regulation of the isoforms. In conclusion, our study examined for the first time the differential expression and regulation of all p53 isoforms in a tumor in vivo. Furthermore, novel results in our in vitro studies show that p53 isoforms are strongly differentially regulated by chemotherapy in RCCs and that expression and regulation of so-called "p53-target genes" are obviously at least in part regulated by other transcription factors. In addition, our original findings show that p53 isoform expression in RCC cell lines is of minor importance for sensitivity towards chemotherapy.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Protein Isoforms/metabolism , Tumor Suppressor Protein p53/metabolism , Base Sequence , Blotting, Western , Carcinoma, Renal Cell/pathology , DNA Primers , Humans , Kidney Neoplasms/pathology , Polymerase Chain Reaction , Protein Isoforms/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/genetics
8.
Cell Oncol ; 31(6): 437-56, 2009.
Article in English | MEDLINE | ID: mdl-19940360

ABSTRACT

Renal cell carcinoma (RCC) is a prototype of a chemo refractory tumour. It remains the most lethal of the common urologic cancers and is highly resistant to conventional therapy. Here, we confirmed the efficiency of anti-Fas monoclonal antibody (CH11) as alternative therapeutic approach for the treatment of RCC and investigated the molecular mechanism(s), whereby CH11 induces apoptosis of RCC cells. The present study shows an essential role for apoptosis signal-regulating kinase 1 (ASK1), together with both c-jun-N-terminal kinase (JNK) and p38 pathways, and caspase-8 in this process. Furthermore, CH11-dependent induction of the ASK1-JNK/p38 pathways was found to activate the transcription factors AP-1 and ATF-2, and FADD-caspase-8-Bid signalling, resulting in the translocation of both Bax and Bak proteins, and subsequently mitochondrial dysregulation that is characterized by the loss of mitochondrial membrane potential (DeltaPsim), cytochrome c release and cleavage of caspase-9, caspase-3 and PARP. Thus, the described molecular mechanisms of CH11-induced apoptosis suggest the reliability of Fas activation as an alternative therapeutic approach for the treatment of patients with advanced renal cell carcinoma.


Subject(s)
Apoptosis/physiology , Caspase 8/metabolism , MAP Kinase Kinase Kinase 5/metabolism , fas Receptor/physiology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Murine-Derived , Apoptosis/drug effects , Blotting, Western , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , DNA Damage , Enzyme Activation/drug effects , Flow Cytometry , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , MAP Kinase Kinase Kinase 5/genetics , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Models, Biological , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , fas Receptor/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
9.
J Stem Cells ; 4(3): 141-6, 2009.
Article in English | MEDLINE | ID: mdl-20232599

ABSTRACT

Divisions of somatic stem cells are required for the maintenance and regeneration of normal tissues, while divisions of cancerous stem cells likely underlie the existence of certain malignant diseases. Studies of recent years suggest that molecular mechanisms governing stem cell self-renewal can be subverted in tumorigenesis to maintain cancerous growth. This is exemplified by the proto-oncogene BMI-1 that is involved in the maintenance of somatic stem cells and in carcinogenesis within the same tissues. BMI-1 interferes with the central cellular tumor suppressor pathways linked to retinoblastoma protein (Rb) and p53. These signaling pathways control the cell cycle, cell differentiation, cellular senescence and cell death. While the roles of the pathways associated with Rb and p53 in cancer are broadly established, further elucidation thereof in stem cells might have implications in cancer research, stem cell biology and regenerative medicine.


Subject(s)
Cell Division , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction , Stem Cells/metabolism , Animals , Humans , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1 , Proto-Oncogene Mas , Regeneration , Tumor Suppressor Proteins/metabolism
10.
J Cancer Res Clin Oncol ; 134(6): 689-95, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18074150

ABSTRACT

PURPOSE: Metastasizing epithelioid sarcoma (ES) is an extremely aggressive tumor, because conventional chemotherapy and irradiation are largely ineffective. Here, we analyzed the impact of the CD95-mediated drug-induced apoptosis in ES cell lines. METHODS: The effects of paclitaxel (Taxol) and 5-FU were determined by MTT assay. The extent of apoptosis was analyzed by light microscopy and Annexin V staining (flow cytometry). The expression of death receptors and ligands was defined by RT-PCR, Western blotting and flow cytometry. RESULTS: All cell lines expressed CD95, but not the CD95 ligand. The CD95 activation resulted in apoptosis and cell death in all cell lines. Both paclitaxel and 5-FU are able to trigger apoptosis, and furthermore, to upregulate CD95, whereas only paclitaxel increases CD95 ligand expression. Neutralizing antibodies directed against CD95 ligand effectively inhibited paclitaxel-induced cell death, thereby providing evidence for a direct involvement of the CD95 system in paclitaxel-induced apoptosis. CONCLUSIONS: Concomitant upregulation of CD95 receptor and ligand may significantly enhance the response of ES to anticancer drugs. As evident from the differential response of our clonal ES subpopulations to paclitaxel and 5-FU, effective activation of the CD95 system depends on intrinsic properties of both the chemotherapeutic agent and target cell population.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Fas Ligand Protein/physiology , Paclitaxel/pharmacology , Sarcoma/drug therapy , Cell Line, Tumor , Fas Ligand Protein/genetics , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Humans , RNA, Messenger/analysis , Sarcoma/metabolism , Sarcoma/pathology , Signal Transduction , Up-Regulation , fas Receptor/genetics , fas Receptor/physiology
11.
Cell Signal ; 19(12): 2428-33, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17651940

ABSTRACT

Self-renewing divisions of normal and cancerous stem cells are responsible for the initiation and maintenance of normal and certain cancerous tissues, respectively. Recent findings suggest that tumor surveillance mechanisms can reduce regenerative capacity and frequency of normal stem cells, thereby contributing to tissue aging. Signaling pathways promoting self-renewal of stem cells can also drive proliferation in cancer. The BMI-1 proto-oncogene is required for the maintenance of tissue-specific stem cells and is involved in carcinogenesis within the same tissues. BMI-1 promotes self-renewal of stem cells largely by interfering with two central cellular tumor suppressor pathways, p16(Ink4a)/retinoblastoma protein (Rb) and ARF/p53, whose disruption is a hallmark of cancer. Nucleolin, an Rb-associated protein, is abundant in proliferating cancerous cells and likely contributes to the maintenance of human CD34-positive stem/progenitor cells of hematopoiesis. Elucidation of the involvement of proto-oncogenes and tumor suppressors in the maintenance of stem cells might have therapeutic implications.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Proliferation , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Signal Transduction , Stem Cells/metabolism , ADP-Ribosylation Factors/metabolism , Animals , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Neoplasms/pathology , Phosphoproteins/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Nucleolin
12.
J Biol Chem ; 282(17): 12439-49, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17255095

ABSTRACT

CD34 glycoprotein in human hematopoiesis is expressed on a subset of progenitor cells capable of self-renewal, multilineage differentiation, and hematopoietic reconstitution. Nucleolin is an abundant multifunctional phosphoprotein of growing eukaryotic cells, involved in regulation of gene transcription, chromatin remodeling, and RNA metabolism, whose transcripts are enriched in murine hematopoietic stem cells, as opposed to differentiated tissue. Here we show that, in human CD34-positive hematopoietic cells, nucleolin activates endogenous CD34 and Bcl-2 gene expression, and cell surface CD34 protein expression is thereby enhanced by nucleolin. Nucleolin-mediated activation of CD34 gene transcription results from direct sequence-specific interactions with the CD34 promoter region. Nucleolin expression prevails in CD34-positive cells mobilized into peripheral blood (PB), as opposed to CD34-negative peripheral blood mononuclear cells (PBMCs). Therefore, in intact CD34-positive mobilized PB cells, a recruitment of nucleolin to the CD34 promoter region takes place, accompanied by nucleosomal determinants of gene activity, which are absent from the CD34 promoter region in CD34-negative PBMCs. Our data show that nucleolin acts as a component of the gene regulation program of CD34-positive hematopoietic cells and provide further insights into processes by which human CD34-positive hematopoietic stem/progenitor cells are maintained.


Subject(s)
Antigens, CD34/biosynthesis , Chromatin Assembly and Disassembly/physiology , Hematopoietic Stem Cells/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Response Elements , Cell Line , Hematopoietic Stem Cells/cytology , Humans , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Transcription, Genetic , Nucleolin
13.
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
14.
J Exp Med ; 196(8): 1067-78, 2002 Oct 21.
Article in English | MEDLINE | ID: mdl-12391018

ABSTRACT

High risk human papillomaviruses (HPVs) are central to the development of cervical cancer and the deregulated expression of high risk HPV oncogenes is a critical event in this process. Here, we find that the cell protein nucleolin binds in a sequence-specific manner to the HPV18 enhancer. The DNA binding activity of nucleolin is primarily S phase specific, much like the transcription of the E6 and E7 oncoproteins of HPV18 in cervical cancer cells. Antisense inactivation of nucleolin blocks E6 and E7 oncogene transcription and selectively decreases HPV18(+) cervical cancer cell growth. Furthermore, nucleolin controls the chromatin structure of the HPV18 enhancer. In contrast, HPV16 oncogene transcription and proliferation rates of HPV16(+) SiHa cervical cancer cells are independent of nucleolin activity. Moreover, nucleolin expression is altered in HPV18(+) precancerous and cancerous tissue from the cervix uteri. Whereas nucleolin was homogeneously distributed in the nuclei of normal epithelial cells, it showed a speckled nuclear phenotype in HPV18(+) carcinomas. Thus, the host cell protein nucleolin is directly linked to HPV18-induced cervical carcinogenesis.


Subject(s)
Oncogene Proteins, Viral/genetics , Oncogenes , Papillomaviridae/genetics , Phosphoproteins/physiology , RNA-Binding Proteins/physiology , Uterine Cervical Neoplasms/virology , Base Sequence , Cell Division , Female , HeLa Cells , Humans , Immunohistochemistry , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Nucleolin
15.
Oncogene ; 21(10): 1485-92, 2002 Feb 28.
Article in English | MEDLINE | ID: mdl-11896576

ABSTRACT

Sp1 binding sites have been identified in enhancer/promoter regions of several growth and cell cycle regulated genes, and it has been shown that Sp1 is increasingly phosphorylated in G1 phase of the cell cycle. Interactions of Sp1 with proteins involved in control of cell cycle and tumor formation have been reported. Here we show that expression of Sp1 protein predominates in the G1 phase of the cell cycle in epithelial cells. This is achieved by proteasome-dependent degradation. Inhibition of endogeneous Sp1 activity by a dominant-negative Sp1 mutant was associated with a cell cycle arrest in G1 phase, a strongly reduced expression of cyclin D1, the EGF-receptor and increased levels of p27Kip1. We have thus identified Sp1 as an important regulator of the cell cycle in G1 phase.


Subject(s)
Epithelial Cells/metabolism , Sp1 Transcription Factor/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Clone Cells , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Cysteine Endopeptidases/metabolism , Electrophoretic Mobility Shift Assay , Epithelial Cells/cytology , ErbB Receptors/metabolism , G1 Phase , Genes, Reporter , HeLa Cells , Humans , Immunohistochemistry , Kinetics , Multienzyme Complexes/metabolism , Mutation , Proteasome Endopeptidase Complex , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/immunology , Transcription, Genetic , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...