Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell Biol ; 21(12): 4032-45, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11359910

ABSTRACT

The retinoblastoma tumor suppressor protein (RB) is a negative regulator of the cell cycle that inhibits both G(1) and S-phase progression. While RB-mediated G(1) inhibition has been extensively studied, the mechanism utilized for S-phase inhibition is unknown. To delineate the mechanism through which RB inhibits DNA replication, we generated cells which inducibly express a constitutively active allele of RB (PSM-RB). We show that RB-mediated S-phase inhibition does not inhibit the chromatin binding function of MCM2 or RPA, suggesting that RB does not regulate the prereplication complex or disrupt early initiation events. However, activation of RB in S-phase cells disrupts the chromatin tethering of PCNA, a requisite component of the DNA replication machinery. The action of RB was S phase specific and did not inhibit the DNA damage-mediated association of PCNA with chromatin. We also show that RB-mediated PCNA inhibition was dependent on downregulation of CDK2 activity, which was achieved through the downregulation of cyclin A. Importantly, restoration of cyclin-dependent kinase 2 (CDK2)-cyclin A and thus PCNA activity partially restored S-phase progression in the presence of active RB. Therefore, the data presented identify RB-mediated regulation of PCNA activity via CDK2 attenuation as a mechanism through which RB regulates S-phase progression. Together, these findings identify a novel pathway of RB-mediated replication inhibition.


Subject(s)
CDC2-CDC28 Kinases , Cyclin-Dependent Kinases/antagonists & inhibitors , Proliferating Cell Nuclear Antigen/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Retinoblastoma Protein/metabolism , S Phase/physiology , Animals , Base Sequence , Cell Line , Chromatin/metabolism , Cyclin A/metabolism , Cyclin-Dependent Kinase 2 , DNA Primers/genetics , DNA Replication , DNA-Binding Proteins/metabolism , Rats , Replication Protein A , Retinoblastoma Protein/genetics , Signal Transduction
2.
Mol Cell Biol ; 20(20): 7751-63, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11003670

ABSTRACT

The retinoblastoma tumor suppressor protein (RB) is a potent inhibitor of cell proliferation. RB is expressed throughout the cell cycle, but its antiproliferative activity is neutralized by phosphorylation during the G(1)/S transition. RB plays an essential role in the G(1) arrest induced by a variety of growth inhibitory signals. In this report, RB is shown to also be required for an intra-S-phase response to DNA damage. Treatment with cisplatin, etoposide, or mitomycin C inhibited S-phase progression in Rb(+/+) but not in Rb(-/-) mouse embryo fibroblasts. Dephosphorylation of RB in S-phase cells temporally preceded the inhibition of DNA synthesis. This S-phase dephosphorylation of RB and subsequent inhibition of DNA replication was observed in p21(Cip1)-deficient cells. The induction of the RB-dependent intra-S-phase arrest persisted for days and correlated with a protection against DNA damage-induced cell death. These results demonstrate that RB plays a protective role in response to genotoxic stress by inhibiting cell cycle progression in G(1) and in S phase.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA Damage , DNA-Binding Proteins , Retinoblastoma Protein/metabolism , S Phase , Animals , Cell Death/drug effects , Cisplatin/pharmacology , Cyclin A/antagonists & inhibitors , Cyclin A/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/deficiency , Cyclins/genetics , Cyclins/physiology , DNA/biosynthesis , DNA Damage/drug effects , DNA Replication/drug effects , E2F Transcription Factors , Etoposide/pharmacology , Fibroblasts , Flow Cytometry , Fluorescent Antibody Technique , Gene Deletion , Mice , Mice, Knockout , Mitomycin/pharmacology , Mutagenicity Tests , Phosphorylation/drug effects , Rats , Retinoblastoma Protein/genetics , Retinoblastoma-Binding Protein 1 , S Phase/drug effects , Transcription Factor DP1 , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
3.
Cell Growth Differ ; 11(7): 361-72, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10939590

ABSTRACT

Prostate cells are dependent on androgen for proliferation, but during tumor progression prostate cancer cells achieve independence from the androgen requirement. We report that androgen withdrawal fails to inhibit cell cycle progression or influence the expression of cyclin-dependent kinase (CDK)/cyclins in androgen-independent prostate cancer cells, indicating that these cells signal for cell cycle progression in the absence of androgen. However, phosphorylation of the retinoblastoma tumor suppressor protein (RB) is still required for G1-S progression in androgen-independent cells, since the expression of constitutively active RB (PSM-RB) or p16ink4a caused cell cycle arrest and mimicked the effects of androgen withdrawal on downstream targets in androgen-dependent LNCaP cells. Since Ras is known to mediate mitogenic signaling to RB, we hypothesized that active V12Ras would induce androgen-independent cell cycle progression in LNCaP cells. Although V12Ras was able to stimulate ERK phosphorylation and induce cyclin D1 expression in the absence of androgen, it was not sufficient to promote androgen-independent cell cycle progression. Similarly, ectopic expression of CDK4/cyclin D1, which stimulated RB phosphorylation in the presence of androgen, was incapable of inactivating RB or driving cell cycle progression in the absence of androgen. We show that androgen regulates both CDK4/cyclin D1 and CDK2 complexes to inactivate RB and initiate cell cycle progression. Together, these data show that androgen independence is achieved via deregulation of the androgen to RB signal, and that this signal can only be partially initiated by the Ras pathway in androgen-dependent cells.


Subject(s)
Adenocarcinoma/metabolism , Androgens/metabolism , CDC2-CDC28 Kinases , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins , Retinoblastoma Protein/metabolism , Signal Transduction , ras Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Androgens/pharmacology , Animals , Bromodeoxyuridine/pharmacology , Cell Cycle , Cell Line , Culture Media , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Electrophoresis, Polyacrylamide Gel , Fibroblasts , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Immunoblotting , Male , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Phosphorylation , Plasmids/genetics , Plasmids/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Rats , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , ras Proteins/genetics
4.
Proc Natl Acad Sci U S A ; 97(14): 7748-53, 2000 Jul 05.
Article in English | MEDLINE | ID: mdl-10884406

ABSTRACT

The antiproliferative action of the retinoblastoma tumor suppressor protein, RB, is disrupted in the majority of human cancers. Disruption of RB activity occurs through several disparate mechanisms, including viral oncoprotein binding, deregulated RB phosphorylation, and mutation of the RB gene. Here we report disruption of RB-signaling in tumor cells through loss of a critical cooperating factor. We have previously reported that C33A cells fail to undergo cell cycle inhibition in the presence of constitutively active RB (PSM-RB). To determine how C33A cells evade RB-mediated arrest, cell fusion experiments were performed with RB-sensitive cells. The resulting fusions were arrested by PSM-RB, indicating that C33A cells lack a factor required for RB-mediated cell cycle inhibition. C33A cells are deficient in BRG-1, a SWI/SNF family member known to stimulate RB activity. Consistent with BRG-1 deficiency underlying resistance to RB-mediated arrest, we identified two other BRG-1-deficient cell lines (SW13 and PANC-1) and demonstrate that these tumor lines are also resistant to cell cycle inhibition by PSM-RB and p16ink4a, which activates endogenous RB. In cell lines lacking BRG-1, we noted a profound defect in RB-mediated repression of the cyclin A promoter. This deficiency in RB-mediated transcriptional repression and cell cycle inhibition was rescued through ectopic coexpression of BRG-1. We also demonstrate that 3T3-derived cells, which inducibly express a dominant-negative BRG-1, arrest by PSM-RB and p16ink4a in the absence of dominant-negative BRG-1 expression; however, cell cycle arrest was abrogated on induction of dominant-negative BRG-1. These findings demonstrate that BRG-1 loss renders cells resistant to RB-mediated cell cycle progression, and that disruption of RB signaling through loss of cooperating factors occurs in cancer cells.


Subject(s)
Cyclin A/metabolism , Nuclear Proteins/metabolism , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Cell Cycle , Cell Fusion , Cyclin A/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Nuclear Proteins/genetics , Promoter Regions, Genetic , Signal Transduction , Transcription Factors/genetics , Tumor Cells, Cultured
5.
Oncogene ; 19(15): 1857-67, 2000 Apr 06.
Article in English | MEDLINE | ID: mdl-10773875

ABSTRACT

Phosphorylation/inactivation of RB is typically required for cell cycle progression. However, we have identified a tumor cell line, C33A, which progresses through the cell cycle in the presence of an active allele of RB (PSM-RB). To determine how C33A cells evade RB-mediated arrest, we compared RB signaling to downstream effectors in this resistant cell line to that of the RB-sensitive SAOS-2 cell line. Although introduction of PSM-RB repressed E2F-mediated transcription in both C33A and SAOS-2 cells, PSM-RB failed to repress Cyclin A promoter activity in C33A. Ectopic expression of PSM-RB in SAOS-2 cells resulted in a decrease in both Cyclin A and Cdk2 protein levels without affecting Cyclin E or Cdk4. In contrast, over-expression of PSM-RB in C33A cells did not alter endogenous Cyclin A, Cyclin E, or Cdk2 protein levels or impact Cdk2 kinase activity, indicating that signaling from RB to down-stream targets is abrogated in this cell line. The importance of Cdk2 activity was demonstrated by p27Kip1, which attenuated Cdk2 activity and inhibited cell cycle progression in C33A cells. Since RB signaling to Cdk2 is disrupted in these tumor cells, we co-expressed two proteins that cooperate with RB in transcriptional repression, AHR and BRG-1, in an attempt to correct this signaling dysfunction. Co-expression of AHR/BRG-1 with PSM-RB attenuated Cyclin A and Cdk2 expression as well as Cdk2-associated kinase activity, resulting in cell cycle inhibition of C33A cells. Importantly, ectopic expression of Cyclin A was able to reverse the arrest mediated by co-expression of AHR/BRG-1 with PSM-RB. These results indicate that down-regulation of Cdk2 activity is requisite for RB-mediated cell cycle arrest. Thus, this study reveals a new mechanism through which tumor cells evade anti-proliferative signals, and provides insight into how RB-signaling is mediated.


Subject(s)
CDC2-CDC28 Kinases , Carrier Proteins , Cell Cycle Proteins , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/metabolism , Cell Cycle , Cyclin A/genetics , Cyclin-Dependent Kinase 2 , DNA Helicases , Down-Regulation , E2F Transcription Factors , Humans , Mutation , Nuclear Proteins/pharmacology , Promoter Regions, Genetic , Receptors, Aryl Hydrocarbon/metabolism , Retinoblastoma-Binding Protein 1 , Signal Transduction , Transcription Factor DP1 , Transcription Factors/metabolism , Transcription Factors/pharmacology , Tumor Cells, Cultured
6.
J Biol Chem ; 274(39): 27632-41, 1999 Sep 24.
Article in English | MEDLINE | ID: mdl-10488103

ABSTRACT

Although RB inhibits the G(1)-S transition, the mechanism through which RB prevents cell cycle advancement remains unidentified. To delineate the mechanism(s) utilized by RB to exert its anti-proliferative activity, constitutively active RB proteins (which cannot be inactivated by phosphorylation) or p16ink4a (which prevents RB inactivation) were utilized. Both proteins inhibited the G(1)-S transition, whereas wild-type RB did not. We show that active RB acts to attenuate cyclin A promoter activity, and that overexpression of cyclin E reverses RB-mediated repression of the cyclin A promoter. Although cyclin A is an E2F-regulated gene, and it has been long hypothesized that RB mediates cell cycle advancement through binding to E2F and attenuating its transactivation potential, cyclin E does not reverse dominant negative E2F-mediated repression of the cyclin A promoter. Although active RB repressed both cyclin A and two other paradigm E2F-regulated promoters, only cyclin A transcription was restored upon co-expression of cyclin E. Additionally, we show that RB but not dominant negative E2F regulates the cyclin A promoter through the CCRE element. These data identify cyclin A as a downstream target of RB-mediated arrest. Consistent with this idea, ectopic expression of cyclin A reversed RB-mediated G(1) arrest. The findings presented suggest a pathway wherein cyclin A is a downstream target of RB, and cyclin E functions to antagonize this aspect of RB-mediated G(1)-S inhibition.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cell Cycle/physiology , Cyclin A/genetics , DNA-Binding Proteins , Retinoblastoma Protein/physiology , Animals , Cell Division , Cell Line , Cyclin A/physiology , Cyclin E/genetics , Cyclin E/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , E2F Transcription Factors , G1 Phase , Gene Expression Regulation , Genes, Reporter , Luciferases/genetics , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Retinoblastoma-Binding Protein 1 , S Phase , Tetrahydrofolate Dehydrogenase/genetics , Transcription Factors/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...