Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mutat Res ; 488(3): 233-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11397651

ABSTRACT

Two genes, TSC1 and TSC2, have been shown to be responsible for tuberous sclerosis (TSC). The detection of loss of heterozygosity of TSC1 or TSC2 in hamartomas, the growths characteristically occurring in TSC patients, suggested a tumor suppressor function for their gene products hamartin and tuberin. Studies analyzing ectopically modulated expression of TSC2 in human and rodent cells together with the finding that a homolog of TSC2 regulates the Drosophila cell cycle suggest that TSC is a disease of proliferation/cell cycle control. We discuss this question including very recent data obtained from analyzing mice expressing a modulated TSC2 transgene, and from studying the effects of deregulated TSC1 expression. Elucidation of the cellular functions of these proteins will form the basis of a better understanding of how mutations in these genes cause the disease and for the development of new therapeutic strategies.


Subject(s)
Cell Cycle/genetics , Cell Division/genetics , Drosophila Proteins , Genes, Tumor Suppressor , Proteins/physiology , Repressor Proteins/physiology , Tuberous Sclerosis/genetics , Tumor Suppressor Proteins , Active Transport, Cell Nucleus , Animals , Carcinoma, Renal Cell/genetics , Cell Compartmentation , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Cell Size/genetics , Chromosomes, Human, Pair 16/genetics , Chromosomes, Human, Pair 9/genetics , Cyclin-Dependent Kinase Inhibitor p27 , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Genes, Dominant , Hamartoma/genetics , Humans , Insect Proteins/genetics , Insect Proteins/physiology , Kidney Neoplasms/genetics , Loss of Heterozygosity , Macromolecular Substances , Proteins/genetics , Rats , Rats, Mutant Strains , Repressor Proteins/genetics , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein
2.
Oncogene ; 18(3): 843-8, 1999 Jan 21.
Article in English | MEDLINE | ID: mdl-9989838

ABSTRACT

The transcription factors c-Myc and E2F-1 have been shown to harbour both mitogenic and apoptotic properties. Both factors have been implicated in the regulation of the transition from the G1 phase to the S phase in the mammalian cell cycle. However, whether cell death triggered by these molecules is dependent on the cell's position in the ongoing cell cycle remained elusive. Using centrifugal elutriation we here show for the first time that c-Myc induces apoptosis in G1 and in G2 phase, whereas E2F-1-induced apoptosis specifically occurs in G1. S phase cells are resistant to cell death triggered by these factors. We demonstrate that this is not a general phenomenon, since S phase cells are susceptible to apoptosis induced by treatment with actinomycin D and to the anti-apoptotic activity of Bcl-2. Our data indicate that S phase cells harbour specific protective activities against c-Myc- and E2F-1-induced apoptosis. Our results demonstrate that these transcription factors, although probably sharing specific apoptotic pathways, also take distinct routes to induce cell death and that apoptosis can occur at different phases of the cell cycle depending on the apoptotic stimulus. In this report we present the usefulness of a new approach to determine the regulation of apoptosis in the ongoing unperturbated cell cycle. This approach has clear implications for the identification of target genes involved in the regulation of cell death.


Subject(s)
Apoptosis , Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Proto-Oncogene Proteins c-myc/biosynthesis , Transcription Factors/biosynthesis , Animals , Cell Cycle , Cell Line , E2F Transcription Factors , E2F1 Transcription Factor , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-myc/genetics , Rats , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/genetics
3.
Mutat Res ; 436(1): 1-9, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9878675

ABSTRACT

In the mammalian cell cycle, the transition from the G1 phase to S phase, in which DNA replication occurs, is dependent on tight cell size control and has been shown to be regulated by the cyclin-dependent kinases (Cdks) 2, 3, 4 and 6. Activities of Cdks are controlled by association with cyclins and reversible phosphorylation reactions. An additional level of regulation is provided by inhibitors of Cdks. G1-S and S phase substrates of these enzymes include proteins implicated in replication and transcription. Whereas the regulation and role of Cdk2, 4 and 6 has intensively been studied, less is known about Cdk3. Recent data provide first insights into the regulation of Cdk3-associate kinase activity and suggest a model how Cdk3 participates in the regulation of the G1-S transition. Although it has been shown that these G1-Cdks are absolutely essential for a proper transition into S phase, their physiological activation is not sufficient to directly initiate replication independently of cell size. Evidence obtained from yeast and Xenopus indicate the initiation of DNA replication to be a two-step process: the origin recognition complex, Cdc6 and Mcm proteins are required for establishing the prereplicative complex and the activities of Cdks and of Cdc7 kinase then trigger the G1-S transition. Recent findings provide evidence that the overall mechanism of initiation of replication is conserved in mammalian cells.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cell Cycle , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins , Animals , Cyclin-Dependent Kinases/antagonists & inhibitors , DNA Replication , E2F Transcription Factors , G1 Phase , Retinoblastoma-Binding Protein 1 , S Phase , Transcription Factors/physiology
4.
J Biol Chem ; 273(48): 32213-21, 1998 Nov 27.
Article in English | MEDLINE | ID: mdl-9822699

ABSTRACT

LR7/8B is a member of the low density lipoprotein receptor gene family that is specifically synthesized in the brain. Here we have functionally expressed in 293 cells the splice variant harboring eight ligand binding repeats (LR8B). As assessed by confocal microscopy, the expressed receptor is localized to the plasma membrane. Importantly, in cell binding experiments, we demonstrate that this protein is a receptor for activated alpha2-macroglobulin. Because to date low density lipoprotein receptor-related protein (LRP) has been shown to be the only alpha2-macroglobulin receptor in brain, we became interested in the expression pattern of both proteins at the cellular level in the brain. LR7/8B is expressed in large neurons and Purkinje cells of the cerebellum and in cells constituting brain barrier systems such as the epithelial cells of the choroid plexus, the arachnoidea, and the endothelium of penetrating blood vessels. Anti-LR7/8B antibody stains the plasma membrane, dendrites, and vesicular structures close to the cell membrane of neurons, especially of Purkinje cells. In contrast, LRP is present in patchy regions around large neurons and most prominently in the glomeruli of the stratum granulare of the cerebellum. This suggests that, contrary to LR7/8B, LRP is expressed in synaptic regions of the neurons; furthermore, there is a striking difference in the expression patterns of LR7/8B and LRP in the choroid plexus. Whereas LRP shows baso-lateral and apical localization in the epithelial cells, LR7/8B is restricted to the apical cell aspect facing the cerebrospinal fluid. Finally, these studies were extended to cultured primary rat neurons, where double immunofluorescence labeling with anti-LR7/8B and anti-microtubuli-associated protein 2 (MAP2) confirmed the somatodendritic expression of the receptor. Based upon these data, we propose that LR7/8B is involved in the clearance of alpha2-macroglobulin.proteinase complexes and/or of other substrates bound to alpha2-macroglobulin from the cerebrospinal fluid and from the surface of neurons.


Subject(s)
Alternative Splicing , Brain/metabolism , Multigene Family , Receptors, Immunologic/genetics , Receptors, LDL/genetics , Animals , Binding Sites , Cell Line , Chickens , Genetic Variation , Humans , Immunohistochemistry , Kinetics , Low Density Lipoprotein Receptor-Related Protein-1 , Rats , Receptors, Immunologic/analysis , Receptors, Immunologic/metabolism , Receptors, LDL/analysis , Receptors, LDL/metabolism , Recombinant Proteins/metabolism , Repetitive Sequences, Amino Acid , Transfection , alpha-Macroglobulins/metabolism
6.
DNA Cell Biol ; 16(6): 737-47, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9212167

ABSTRACT

Although a remarkable number of genes has been identified that are either activated or repressed via c-Myc, only few of them obviously contribute to Myc's biological effect--the induction of proliferation. We found that in logarithmically growing cells overexpression of Myc specifically induces thymidine kinase (TK) mRNA expression and enzyme activity, whereas loss of one allele of Myc causes downregulation of this enzyme. We show that activation of Myc triggers high levels of this normally strictly S-phase-regulated DNA metabolism enzyme in serum arrested G0 cells and causes high and constant levels of TK expression throughout the entire ongoing cell cycle. Induction of TK by Myc requires an intact transcriptional activation domain. Myc-induced deregulation of this enzyme is paralleled by alterations of protein binding at the E2F-site of the TK promoter. We further show that cell growth arrest by the cyclin-dependent kinase inhibitor p16 is abrogated by overexpression of Myc and that co-overexpression of p16 cannot inhibit the Myc-induced up-regulation of TK expression. Our data demonstrate TK to be a cellular target of Myc independently of the status of cell proliferation and provide evidence that the transcription factor E2F might be involved in this process.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins , Proto-Oncogene Proteins c-myc/physiology , Thymidine Kinase/metabolism , Animals , Carrier Proteins/genetics , Cell Division/genetics , Cell Line , Cyclin-Dependent Kinase Inhibitor p16 , E2F Transcription Factors , Enzyme Activation/physiology , Promoter Regions, Genetic , RNA, Messenger/genetics , Rats , Retinoblastoma-Binding Protein 1 , Thymidine Kinase/genetics , Transcription Factor DP1 , Transcription Factors/metabolism , Up-Regulation
7.
Oncogene ; 14(19): 2251-7, 1997 May 15.
Article in English | MEDLINE | ID: mdl-9178900

ABSTRACT

The transcription factor E2F activates genes required for S phase, such as cyclin E and cyclin A. We show that, contrary to long term effects of E2F-1 overexpression, short ectopic overexpression of this transcription factor in logarithmically growing cells does neither affect the cell cycle distribution nor the cell size, but heavily induces cyclin E and A expression as well as cyclin E- and A-dependent kinase activities. We further separated logarithmically growing E2F-1-overexpressing cells according to their different cell cycle phases by centrifugal elutriation. These experiments revealed that deregulated E2F-1 expression triggers high levels of cyclin E and A expression and kinase activities in small early G1 cells, normally not exhibiting these activities. These effects on the regulation of cyclin E- and A-associated kinases are not accompanied by any detectable alteration in the rate of progression through the cell cycle, suggesting that these changes are independent of any mitogenic properties of E2F-1.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cell Cycle , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , DNA-Binding Proteins , Transcription Factors/genetics , Animals , Cell Line , E2F Transcription Factors , E2F1 Transcription Factor , Gene Expression Regulation , RNA, Messenger/genetics , Rats , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1
8.
FEBS Lett ; 385(3): 143-8, 1996 May 06.
Article in English | MEDLINE | ID: mdl-8647239

ABSTRACT

We analysed cyclin D1 mRNA and protein expression in several different cell types after separating these cells according to their different cell cycle phases by centrifugal elutriation. In normal human and rat fibroblasts cyclin D1 expression is high in early to mid G1 and decreases about 6-7 fold before onset of replication. It has been demonstrated that specific transforming events, such as loss of functional retinoblastoma protein, overexpression of c-myc, and transfection with the human papillomavirus oncoproteins E6 and E7 cause transcriptional downregulation of cyclin D1 expression in logarithmically growing cells. We found that such transformed cells exhibit loss of the cell cycle-dependent cyclin D1 fluctuation accompanied with reduced upregulation of cyclin D1 in G1 phase. The data presented here provide the experimental support for a recently suggested model involving the function of the retinoblastoma protein in cyclin D1 cell cycle regulation.


Subject(s)
Cell Cycle , Cell Transformation, Neoplastic , Cell Transformation, Viral , Cyclins/metabolism , Oncogene Proteins/metabolism , Animals , Cell Line, Transformed , Cells, Cultured , Cyclin D1 , Cyclins/genetics , Fibroblasts , Gene Expression Regulation , Humans , Interphase , Mitosis , Oncogene Proteins/genetics , RNA, Messenger/metabolism , Rats , S Phase , Thymidine Kinase/metabolism , Tumor Cells, Cultured
9.
Oncogene ; 12(8): 1635-43, 1996 Apr 18.
Article in English | MEDLINE | ID: mdl-8622883

ABSTRACT

The role of alterations of the MTS1 tumor suppressor gene on chromosome 9p21, which encodes p16, the inhibitor of cyclin-dependent-kinase-4 and 6, in tumorigenesis is not yet clear. Phosphorylation of the retinoblastoma protein by cyclin-dependent kinases 4 and 6 prevents its interaction with the transcription factor E2F, which subsequently promotes the expression of S phase regulated genes, such as thymidine kinase. Although a role of p16 in this regulation has been presumed, there is no proof so far that loss of this tumor suppressor gene really affects E2F-mediated regulations. We investigated the regulation of thymidine kinase in phytohemagglutinin-stimulated normal human lymphocytes and in the p16-negative human acute lymphoblastic leukemia cell lines, MOLT-4 and CEM. Compared to normal lymphocytes, MOLT-4 and CEM cells exhibited an altered cell cycle regulation of thymidine kinase, a much higher intracellular activity of this enzyme, and higher thymidine kinase mRNA expression. Transient expression of p16 in normal human lymphocytes caused arrest in G1, but was without effect on the cell growth of MOLT-4 and CEM cells, although all of them express functional retinoblastoma protein. Nevertheless, in the two leukemia cell lines transient overexpression of p16 reestablished the normal regulation of thymidine kinase, paralleled by an increase of the underphosphorylated form of retinoblastoma protein and decrease of free E2F bound to its motif in the thymidine kinase promoter. We demonstrate that loss of p16 causes upregulation of this DNA precursor pathway enzyme via activation of E2F by a mechanism involving retinoblastoma protein.


Subject(s)
Carrier Proteins/physiology , Cell Cycle Proteins , DNA-Binding Proteins , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Thymidine Kinase/biosynthesis , Transcription Factors/physiology , Cell Cycle/genetics , Cell Division/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16 , Deoxycytidine Kinase/biosynthesis , Down-Regulation , E2F Transcription Factors , G1 Phase , HeLa Cells , Humans , Lymphocytes/cytology , Lymphocytes/drug effects , Lymphocytes/metabolism , Phosphorylation , Phytohemagglutinins/metabolism , Phytohemagglutinins/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma Protein/physiology , Retinoblastoma-Binding Protein 1 , Thymidine Kinase/genetics , Transcription Factor DP1 , Transcription Factors/genetics , Transcription, Genetic , Transfection , Tumor Cells, Cultured , Up-Regulation
10.
DNA Cell Biol ; 15(1): 41-51, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8561896

ABSTRACT

Homozygous deletions of the tumor suppressor gene p16/MTS1 were reported in a wide variety of tumors and tumor cell lines. Its product inhibits the phosphorylation of the retinoblastoma protein (pRb) by CDK4 and CDK6. Because phosphorylation of pRb is a major regulatory event in the activation of the transcription factor E2F, a role for p16 in the regulation of E2F-dependent transcription was presumed. We investigated the effect of the loss of p16 on E2F-mediated transcription in a tumor progression model consisting of three cell lines originating from a common precursor cell--one p16-positive cell line established from the primary biopsy and two lines derived from more advanced stages of the tumor representing the same cell clone after loss of p16. We observed up- and deregulation of E2F-dependent transcription during the cell cycle of the p16-negative cell clones, which returned to normal after transient expression of p16. This p16-dependent regulation affects a set of enzymes necessary for the activation of all four DNA precursors; it is paralleled by the interconversion of transcriptionally active free E2F and transcriptionally inactive higher molecular complexes of E2F and is dependent on the existence of endogenous pRb. Furthermore, we show that p16-negative cell clones exhibit a growth advantage compared to their p16-positive counterparts. One might speculate that one feature of tumor progression could be deregulation of E2F-dependent transcription caused by loss of p16.


Subject(s)
Carrier Proteins/genetics , Cell Cycle Proteins , DNA-Binding Proteins , DNA/metabolism , Enzymes/genetics , Transcription Factors/metabolism , Carrier Proteins/metabolism , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p16 , E2F Transcription Factors , Enzyme Activation , Enzymes/metabolism , Gene Deletion , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/genetics , HeLa Cells , Humans , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1
11.
J Biol Chem ; 270(44): 26139-45, 1995 Nov 03.
Article in English | MEDLINE | ID: mdl-7592817

ABSTRACT

Rapidly growing oocytes in the laying hen are, in addition to the liver, targets of the so-called "reverse cholesterol transport" (RCT) (Vieira, P.M., Vieira, A.V., Sanders, E.J., Steyrer, E., Nimpf, J., and Schneider, W.J. (1995) J. Lipid Res. 36, 601-610), pointing to the importance of this process in nonplacental reproduction. We have begun to delineate the details of this unique transport pathway branch by molecular characterization of the first nonmammalian lecithin-cholesterol acyltransferase (LCAT), the enzyme that catalyzes an early step in RCT. The biological significance of the enzyme is underscored by the high degree of protein sequence identity (73%) maintained from chicken to man. Interestingly, the conservation extends much less to the cysteine residues; in fact, two of the cysteines thought to be important in mammalian enzymes (residues 31 and 184 in man) are absent from the chicken enzyme, providing proof of their dispensability for enzymatic activity. Antibodies prepared against a chicken LCAT fusion protein cross-react with human LCAT and identify a 64-kDa protein present in enzymatically active fractions obtained by hydrophobic chromatography of chicken serum. The developmental and tissue distribution pattern of LCAT in females is striking; during embryogenesis and adolescence, LCAT expression is extremely high in liver but undetectable in brain. Upon onset of laying, however, brain LCAT mRNA increases suddenly and is maintained at levels 5 times higher than in liver, in stark contrast to most mammals. In adult roosters, the levels of LCAT transcripts in brain are lower than in liver. Together with the molecular characterization of chicken LCAT, these newly discovered developmental changes and gender differences in its expression establish the avian oocyte/liver system as a powerful model to delineate in vivo regulatory elements of RCT.


Subject(s)
Oocytes/enzymology , Phosphatidylcholine-Sterol O-Acyltransferase/biosynthesis , Phosphatidylcholine-Sterol O-Acyltransferase/chemistry , Amino Acid Sequence , Animals , Antibodies , Base Sequence , Binding Sites , Blotting, Northern , Brain/enzymology , Chick Embryo , Chickens , Cross Reactions , Female , Gene Expression , Humans , Liver/enzymology , Male , Mammals , Molecular Sequence Data , Organ Specificity , Oviposition , Papio , Phosphatidylcholine-Sterol O-Acyltransferase/immunology , Rats , Recombinant Fusion Proteins/immunology , Sequence Homology, Amino Acid , Swine
12.
FEBS Lett ; 373(2): 164-9, 1995 Oct 09.
Article in English | MEDLINE | ID: mdl-7589458

ABSTRACT

It has been demonstrated that protein expression of p16, the inhibitor of cyclin-dependent kinase 4 and 6, increases 4 fold at the G1/S transition when serum-arrested cells are restimulated to logarithmic growth. We examined the cell cycle regulation of this cyclin-dependent kinase inhibitor in cells separated according to their cell cycle phases by centrifugal elutriation. Neither p16 mRNA nor its protein expression are regulated during the cell cycle of normal phytohemagglutinin-stimulated lymphocytes, retinoblastoma protein-negative cells, papilloma virus-transformed cells, and acute promyelocytic leukemia cells. p16 mRNA is constitutively expressed in cells in which we detected the normal E2F-dependent S-phase specific expression of thymidine kinase mRNA. We further observed a G1-phase specific expression of cyclin D1 mRNA in the same cells separated by centrifugal elutriation.


Subject(s)
Carrier Proteins/biosynthesis , Cell Cycle , Gene Expression , Blotting, Western , Carrier Proteins/analysis , Cell Division , Cyclin D1 , Cyclin-Dependent Kinase Inhibitor p16 , Cyclins/biosynthesis , Enzyme Inhibitors/analysis , Eye Neoplasms , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Leukemia, Promyelocytic, Acute , Oncogene Proteins/biosynthesis , Protein Kinase Inhibitors , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Retinoblastoma , Retinoblastoma Protein/analysis , Retinoblastoma Protein/biosynthesis , Thymidine Kinase/biosynthesis , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...