Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters










Publication year range
1.
Oncogene ; 33(31): 4036-8, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-24166507

ABSTRACT

The study of the small DNA tumor viruses continues to provide valuable new insights into oncogenesis and fundamental biological processes. Although much has already been revealed about how the human papillomaviruses (HPVs) can transform cells and contribute to cervical and oropharyngeal cancer, there clearly is much more to learn. In this issue of Oncogene, Pang et al., doi:10.1038/onc.2013.426, demonstrate that the high-risk HPV16 E7 oncogene can promote cellular proliferation by interacting with the DREAM (DP, RB-like, E2F and MuvB) complex at two distinct phases of the cell cycle. Consistent with earlier work, HPV16 E7 can bind to the retinoblastoma tumor suppressor (RB) family member p130 (RBL2) protein and promote its proteasome-mediated destruction thereby disrupting the DREAM complex and can prevent exit from the cell cycle into quiescence. In addition, they demonstrate that HPV16 E7 can bind to MuvB core complex in association with BMYB and FOXM1 and activate gene expression during the G2 and M phase of the cell cycle. Thus, HPV16 E7 acts to prevent exit from the cell cycle entry and promotes mitotic proliferation and may account for the high levels of FOXM1 often observed in poor-risk cervical cancers.


Subject(s)
Cell Cycle Proteins/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Human papillomavirus 16/metabolism , Nuclear Proteins/metabolism , Papillomavirus E7 Proteins/metabolism , Trans-Activators/metabolism , Tumor Suppressor Proteins/metabolism , Uterine Cervical Neoplasms/virology , Female , Forkhead Box Protein M1 , Humans
2.
Leukemia ; 17(6): 1104-11, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12764376

ABSTRACT

B-cell chronic lymphocytic leukemia (B-CLL) is characterized by the accumulation of growth arrested clonal B lymphocytes that undergo apoptosis when treated with fludarabine. To further explore the mechanism for the cell cycle arrest, we examined the expression and activity of cyclin-dependent kinases and inhibitors in primary B-CLL cells. We observed high levels of p27kip1, cyclin D2, cyclin E, cdk2, and cdk4 expression in freshly isolated B-CLL cells. Despite high levels of cyclins and cdks, little cdk2 or cdk4 activity was observed with p27kip1 in complex with cyclinD2/cdk4 and cyclin E/cdk2. Remarkably, when B-CLL cells were treated in vitro with fludarabine, p27kip1 underwent caspase-specific degradation accompanied by an increase in cdk4 activity. We conclude that the G0/G1 arrest of B-CLL cells may protect against apoptosis and that the decrease in p27kip1 expression by caspase cleavage may be a key step in chemotherapy-induced apoptosis in B-CLL.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , CDC2-CDC28 Kinases , Caspases/metabolism , Cell Cycle Proteins/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Proto-Oncogene Proteins , Tumor Suppressor Proteins/metabolism , Vidarabine/analogs & derivatives , Vidarabine/pharmacology , Blotting, Western , Caspase Inhibitors , Cell Cycle/drug effects , Cyclin D2 , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Enzyme Inhibitors/pharmacology , Humans , In Situ Nick-End Labeling , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Precipitin Tests , Protein Serine-Threonine Kinases/metabolism , Subcellular Fractions , Tumor Cells, Cultured
3.
Mol Cell Biol ; 21(5): 1854-65, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238922

ABSTRACT

Substrates of cyclin-cdk2 kinases contain two distinct primary sequence motifs: a cyclin-binding RXL motif and one or more phosphoacceptor sites (consensus S/TPXK/R or S/TP). To identify novel cyclin-cdk2 substrates, we searched the database for proteins containing both of these motifs. One such protein is human HIRA, the homologue of two cell cycle-regulated repressors of histone gene expression in Saccharomyces cerevisiae, Hir1p and Hir2p. Here we demonstrate that human HIRA is an in vivo substrate of a cyclin-cdk2 kinase. First, HIRA bound to and was phosphorylated by cyclin A- and E-cdk2 in vitro in an RXL-dependent manner. Second, HIRA was phosphorylated in vivo on two consensus cyclin-cdk2 phosphoacceptor sites and at least one of these, threonine 555, was phosphorylated by cyclin A-cdk2 in vitro. Third, phosphorylation of HIRA in vivo was blocked by cyclin-cdk2 inhibitor p21(cip1). Fourth, HIRA became phosphorylated on threonine 555 in S phase when cyclin-cdk2 kinases are active. Fifth, HIRA was localized preferentially to the nucleus, where active cyclin A- and E-cdk2 are located. Finally, ectopic expression of HIRA in cells caused arrest in S phase and this is consistent with the notion that it is a cyclin-cdk2 substrate that has a role in control of the cell cycle.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle Proteins , Cyclin A/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinases/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/physiology , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins , Transcription Factors/chemistry , Transcription Factors/physiology , Amino Acid Sequence , Blotting, Western , Cell Cycle , Cell Line , Cell Nucleus/metabolism , Cell Separation , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Flow Cytometry , Glutathione Transferase/metabolism , Histone Chaperones , Humans , Mass Spectrometry , Microscopy, Fluorescence , Molecular Sequence Data , Nuclear Proteins/metabolism , Peptides/chemistry , Phosphorylation , Plasmids/metabolism , Precipitin Tests , Protein Binding , Recombinant Fusion Proteins/metabolism , Repressor Proteins/chemistry , S Phase , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid , Threonine/chemistry , Transcription Factors/metabolism , Transfection
4.
Semin Cancer Biol ; 11(1): 15-23, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11243895

ABSTRACT

SV40 large T antigen (TAg) is a powerful oncoprotein capable of transforming a variety of cell types. The transforming activity of TAg is due in large part to its perturbation of the retinoblastoma (pRB) and p53 tumor suppressor proteins. In addition, TAg binds to several other cellular factors, including the transcriptional co-activators p300 and CBP, which may contribute to its transformation function. Several other features of TAg that appear to contribute to its full transformation potential are yet to be completely understood. Study of TAg therefore continues to provide new insights into the mechanism of cellular transformation.


Subject(s)
Antigens, Polyomavirus Transforming/physiology , Cell Transformation, Neoplastic/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Animals , CREB-Binding Protein , Humans , Nuclear Proteins/genetics , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Trans-Activators/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Nat Cell Biol ; 3(12): 1043-50, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11781565

ABSTRACT

The Mlu1-binding factor (MBF) from the fission yeast Schizosaccharomyces pombe contains the proteins Res1p and Res2p and binds to the Mlu1 cell-cycle box (MCB) element in DNA, activating the transcription of genes required for S phase. We report here that the cell-cycle-regulated expression of the cyclin cig2 gene is dependent on MBF. Deletion of MCB elements in the cig2 promoter perturbed the expression not only of cig2 but also of other MBF-dependent genes, indicating that Cig2p could regulate MBF activity. Cig2p can bind to Res2p, promote the phosphorylation of Res1p and inhibit MBF-dependent gene transcription. Cig2p thus forms an autoregulating feedback-inhibition loop with MBF which is important for normal regulation of the cell cycle.


Subject(s)
Cyclins/genetics , Cyclins/metabolism , DNA-Binding Proteins , Saccharomyces cerevisiae Proteins/metabolism , Schizosaccharomyces pombe Proteins , Transcription Factors/metabolism , Base Sequence , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Cyclin B , Feedback, Physiological/physiology , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Meiosis/physiology , Molecular Sequence Data , Mutagenesis/physiology , Phosphorylation , Promoter Regions, Genetic/physiology , Schizosaccharomyces
7.
Mol Cell Biol ; 20(23): 8889-902, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11073989

ABSTRACT

Control of proliferation and differentiation by the retinoblastoma tumor suppressor protein (pRB) and related family members depends upon their interactions with key cellular substrates. Efforts to identify such cellular targets led to the isolation of a novel protein, EID-1 (for E1A-like inhibitor of differentiation 1). Here, we show that EID-1 is a potent inhibitor of differentiation and link this activity to its ability to inhibit p300 (and the highly related molecule, CREB-binding protein, or CBP) histone acetylation activity. EID-1 is rapidly degraded by the proteasome as cells exit the cell cycle. Ubiquitination of EID-1 requires an intact C-terminal region that is also necessary for stable binding to p300 and pRB, two proteins that bind to the ubiquitin ligase MDM2. A pRB variant that can bind to EID1, but not MDM2, stabilizes EID-1 in cells. Thus, EID-1 may act at a nodal point that couples cell cycle exit to the transcriptional activation of genes required for differentiation.


Subject(s)
Adenovirus E1A Proteins/metabolism , Cell Cycle/physiology , Saccharomyces cerevisiae Proteins , Acetyltransferases/antagonists & inhibitors , Amino Acid Sequence , CREB-Binding Protein , Cell Cycle Proteins , Cell Differentiation , Cloning, Molecular , Down-Regulation , Histone Acetyltransferases , Molecular Sequence Data , Nuclear Proteins/antagonists & inhibitors , Protein Binding , Protein Processing, Post-Translational , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Repressor Proteins , Retinoblastoma Protein/metabolism , Trans-Activators/antagonists & inhibitors , Transcriptional Activation , Two-Hybrid System Techniques , Ubiquitins/metabolism
8.
Oncogene ; 19(44): 5116-22, 2000 Oct 19.
Article in English | MEDLINE | ID: mdl-11042701

ABSTRACT

The retinoblastoma family of proteins including pRB, p107 and p130 undergoes cell cycle dependent phosphorylation during the mid-G1 to S phase transition. This phosphorylation is dependent upon the activity of cyclin D/cdk4. In contrast to pRB and p107, p130 is phosphorylated during G0 and the early G1 phase of the cell cycle. We observed that p130 is specifically phosphorylated on serine and threonine residues in T98G cells arrested in G0 by serum deprivation or density arrest. Identification of the phospho-serine and phospho-threonine residues revealed that most were clustered within a short co-linear region unique to p130, defined as the Loop. Deletion of the Loop region resulted in a change in the phosphorylation status of p130 under growth arrest conditions. Notably, deletion of the Loop did not affect the ability of p130 to bind to E2F-4 or SV40 Large T antigen, to induce growth arrest in Saos-2 cells, and to become hyperphosphorylated during the proliferative phase of the cell cycle. p130 undergoes specific G0 phosphorylation in a manner that distinguishes it from pRB and p107.


Subject(s)
Phosphoproteins/metabolism , Proteins , Resting Phase, Cell Cycle/physiology , Amino Acid Sequence , Animals , Antigens, Polyomavirus Transforming/metabolism , Cell Division/physiology , Culture Media, Serum-Free , DNA-Binding Proteins/metabolism , E2F4 Transcription Factor , Glioblastoma/metabolism , Glioblastoma/pathology , Mice , Molecular Sequence Data , Phosphorylation , Protein Structure, Tertiary , Rats , Retinoblastoma-Like Protein p130 , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured
9.
Mol Cell Biol ; 20(20): 7624-33, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11003658

ABSTRACT

At least three domains of simian virus 40 large T antigen (TAg) participate in cellular transformation. The LXCXE motif of TAg binds to all members of the retinoblastoma protein (pRB) family of tumor suppressors. The N-terminal 70 residues of TAg have significant homology to the J domain of Hsp40/DnaJ and cooperate with the LXCXE motif to inactivate the pRB family. A bipartite C-terminal domain of TAg binds to p53 and thereby disrupts the ability of p53 to act as a sequence-specific transcription factor. The contribution of these three domains of TAg to cellular transformation was evaluated in cells that contained inactivating mutations in the pRB and p53 pathways. Cells that stably expressed wild-type or selected mutant forms of TAg were generated in mouse embryo fibroblasts (MEFs) containing homozygous deletions in the RB, INK4a, and ARF loci. It was determined that the J domain, the LXCXE motif, and the p53-binding domain of TAg were required for full transformation of wild-type and RB(-/-) MEFs. In contrast, INK4a(-/-) MEFs that lacked expression of p16(INK4a) and p19(ARF) and ARF(-/-) MEFs that lacked p19(ARF) but expressed p16(INK4a) acquired anchorage-independent growth when expressing wild-type TAg or mutant derivatives that disrupted either the pRB-binding or p53-binding domain. The expression and function of the pRB family members were not overly disrupted in ARF(-/-) MEFs expressing LXCXE mutants of TAg. These results suggest that inactivating mutations of p19(ARF) can relieve the requirement for the LXCXE motif in TAg-mediated transformation and that TAg may have additional functions in transformation.


Subject(s)
Antigens, Viral, Tumor/chemistry , Antigens, Viral, Tumor/metabolism , Cell Cycle Proteins , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins , Proteins/metabolism , Retinoblastoma Protein/metabolism , Amino Acid Motifs , Animals , Antigens, Viral, Tumor/genetics , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Count , Cell Division , Cell Line, Transformed , Contact Inhibition , Cyclin-Dependent Kinase Inhibitor p16 , E2F Transcription Factors , Fibroblasts , Gene Deletion , Gene Expression Regulation, Neoplastic , Genes, Reporter , Mice , Promoter Regions, Genetic , Protein Binding , Proteins/genetics , Retinoblastoma Protein/genetics , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/metabolism , Tumor Suppressor Protein p14ARF , Tumor Suppressor Protein p53/metabolism
10.
J Vasc Surg ; 32(3): 441-8; discussion 448-50, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10957650

ABSTRACT

OBJECTIVE: The purpose of this study was to determine the effect of patient compliance on a program of watchful waiting in cases of small abdominal aortic aneurysms and to document the proportion of patients who become prohibitive operative risks during follow-up. STUDY DESIGN: A retrospective review was conducted at a regional military veterans medical center. The subjects were 101 male military veterans with abdominal aortic aneurysms measuring less than 5 cm who did not have medical contraindications to operative repair. The main outcome measures were (1) the proportion of patients who missed three scheduled radiologic tests in a row despite written notifications mailed to their homes and (2) the proportion of compliant patients who had medical illnesses and became prohibitive operative risks during follow-up. RESULTS: During a follow-up (mean +/- SEM) of 34 +/- 2 months, 69 patients (69%) were fully compliant with the watchful waiting program and underwent a mean of 4.5 +/- 0.3 radiologic tests. There were no abdominal aortic aneurysm ruptures in this subgroup. Twenty-five patients (36%) had indications for abdominal aortic aneurysm repair, and 28 (41%) have not met the criteria for repair. Sixteen (23%) of the 69 compliant patients developed prohibitive medical risks during follow-up; eight (50%) of these 16 patients died, all of the causes unrelated to their abdominal aortic aneurysms. Thirty-two (32%) of the 101 study subjects were noncompliant with the watchful waiting program. Twenty-seven (84%) of the noncompliant patients did not keep any scheduled appointments, and five (16%) were lost after one or two examinations. Three of the noncompliant patients experienced documented abdominal aortic aneurysm rupture, and it is suspected in a fourth. Direct contact was made with 28 (88%) of these patients or their families; all acknowledged having received written notifications regarding their watchful waiting program tests and had decided not to continue with surveillance for a variety of socioeconomic reasons. Between the 69 compliant patients and the 32 noncompliant patients, there were no differences with respect to mean age (70 +/- 1 years vs 73 +/- 2 years), distance from home of record to the hospital (62 +/- 14 miles vs 73 +/- 23 miles), or abdominal aortic aneurysm size at initial detection (3.75 +/- 0.5 cm vs 3.8 +/- 0.5 cm). CONCLUSIONS: Watchful waiting programs are imperfect and highly reliant on the motivation levels and means of the individual patients. Watchful waiting is reasonable among compliant patients with abdominal aortic aneurysms, inasmuch as fewer than half will meet the criteria for intervention within a mean of 3 years. Approximately one fourth of these patients will have medical contraindications to abdominal aortic aneurysm repair during follow-up, and many of these will die of causes other than abdominal aortic aneurysm rupture. In our experience, one third of candidates for watchful waiting programs are unable to participate and are at risk of rupture. These patients need special attention so that the reasons for their noncompliance can be determined, and they may be candidates for earlier intervention.


Subject(s)
Aortic Aneurysm, Abdominal/surgery , Aged , Aortic Aneurysm, Abdominal/diagnosis , Aortic Aneurysm, Abdominal/mortality , Aortic Rupture/diagnosis , Aortic Rupture/mortality , Aortic Rupture/surgery , Female , Follow-Up Studies , Humans , Male , Patient Compliance , Patient Education as Topic , Risk Assessment , Survival Rate
11.
Mol Cell Biol ; 20(13): 4745-53, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10848600

ABSTRACT

Independent of its antiapoptotic function, Bcl-2 can, through an undetermined mechanism, retard entry into the cell cycle. Cell cycle progression requires the phosphorylation by cyclin-dependent kinases (Cdks) of retinoblastoma protein (pRB) family members to free E2F transcription factors. We have explored whether retarded cycle entry is mediated by the Cdk inhibitor p27 or the pRB family. In quiescent fibroblasts, enforced Bcl-2 expression elevated levels of both p27 and the pRB relative p130. Bcl-2 still slowed G(1) progression in cells deficient in pRB but not in those lacking p27 or p130. Hence, pRB is not required, but both p27 and p130 are essential mediators. The ability of p130 to form repressive complexes with E2F4 is implicated, because the retardation by Bcl-2 was accentuated by coexpressed E2F4. A plausible relevant target of p130/E2F4 is the E2F1 gene, because Bcl-2 expression delayed E2F1 accumulation during G(1) progression and overexpression of E2F1 overrode the Bcl-2 inhibition. Hence, Bcl-2 appears to retard cell cycle entry by increasing p27 and p130 levels and maintaining repressive complexes of p130 with E2F4, perhaps to delay E2F1 expression.


Subject(s)
Carrier Proteins , Cell Cycle/physiology , Microtubule-Associated Proteins/metabolism , Phosphoproteins/metabolism , Proteins , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , E2F4 Transcription Factor , Lymphocytes/cytology , Lymphocytes/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma-Binding Protein 1 , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , Transcription Factor DP1 , Transcription Factors/genetics
12.
Oncogene ; 19(24): 2820-7, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10851085

ABSTRACT

Cyclin dependent kinase 4 (cdk4) activity is controlled by the binding of regulatory subunits and inhibitory factors, as well as tyrosine and serine/threonine phosphorylation. More recently the influence of calcium levels have been demonstrated. Using transient transfections in Jurkat cells, we observed specific binding between cdk4 and the calcium and calmodulin activated serine/threonine phosphatase, calcineurin. Furthermore, we demonstrated that the inhibition of the phosphatase activity of calcineurin with FK506 and cyclosporin A resulted in an overall increase in cdk4 kinase activity, suggesting that the phosphatase activity of calcineurin was inhibitory to the kinase activity of cdk4. In contrast, we were not able to observe a similar effect on the kinase activity of either cdk6 or cdk2, indicating that the phosphatase activity of calcineurin was specific for cdk4. In addition, using an in vitro phosphatase assay for calcineurin, we observed that the exogenous addition of calcineurin resulted in the dephosphorylation of cdk4, an event that downregulated the kinase activity of cdk4. Calcineurin could, therefore, play an opposing role to the action of the cyclin activating kinase complex, an enzyme that upregulates the kinase activity of cdk4, an important G0/G1 checkpoint element in mammalian cells. Oncogene (2000) 19, 2820 - 2827


Subject(s)
Calcineurin/physiology , Cyclin-Dependent Kinases/physiology , Proto-Oncogene Proteins , Cyclin-Dependent Kinase 4 , Humans , Jurkat Cells , Mitosis , Phosphoric Monoester Hydrolases/physiology , Phosphorylation
13.
J Virol ; 74(2): 864-74, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10623749

ABSTRACT

Simian virus 40 (SV40) large T antigen (LT) can immortalize and transform many cell types. These activities are attributed in large part to the binding and functional inactivation by LT of two major tumor suppressors: p53 and the retinoblastoma protein, pRB. Most effects of LT on pRB have been shown to additionally require an intact J domain, which mediates binding to Hsc70. We show here that the J domain is not required for p53 override in full-length LT. Although LT binds p53, it was shown previously that overcoming a p53-induced cell cycle arrest requires binding to pRB family members (R. S. Quartin et al., J. Virol. 68:1334-1341). We demonstrate that an LT mutant defective for pRB family member binding (K1) can be complemented for efficient override of p53 arrest by a construct encoding the first 135 amino acids of LT with a J domain-inactivating mutation, H42Q. Hence, complementation does not require the J domain, and pRB binding by LT is important for more than dissociating pRB-E2F complexes, which is J dependent. In accordance with this notion, LT alleviates pRB small-pocket-mediated transcriptional repression independently of the J domain. The LT K1 mutant can also be complemented for p53 override by small t antigen (st) in a manner independent of its J domain. Our observations underscore the importance of multiple SV40 functions, two in LT and one in st, that act cooperatively to counteract p53 growth suppression.


Subject(s)
Antigens, Polyomavirus Transforming/physiology , Retinoblastoma Protein/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Antigens, Polyomavirus Transforming/genetics , Binding Sites , Cell Cycle , Cell Division , Cell Line , Cell Transformation, Viral/physiology , Genetic Complementation Test , Humans , Mutagenesis , Rats , Temperature , Tumor Cells, Cultured , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
14.
Mol Cell Biol ; 19(10): 6632-41, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10490602

ABSTRACT

Retinoblastoma (RB) tumor suppressor family proteins block cell proliferation in part by repressing certain E2F-specific promoters. Both histone deacetylase (HDAC)-dependent and -independent repression activities are associated with the RB "pocket." The mechanism by which these two repression functions occupy the pocket is unknown. A known RB-binding protein, RBP1, was previously found by our group to be an active corepressor which, if overexpressed, represses E2F-mediated transcription via its association with the pocket. We show here that RBP1 contains two repression domains, one of which binds all three known HDACs and represses them in an HDAC-dependent manner while the other domain functions independently of the HDACs. Thus, RB family members repress transcription by recruiting RBP1 to the pocket. RBP1, in turn, serves as a bridging molecule to recruit HDACs and, in addition, provides a second HDAC-independent repression function.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation , Histone Deacetylases/metabolism , Retinoblastoma Protein/metabolism , Binding Sites , Carrier Proteins/genetics , Models, Genetic , Mutation , Protein Binding , Sequence Deletion , Transcription, Genetic
15.
Biologicals ; 27(1): 23-8, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10441399

ABSTRACT

SV40 large T antigen (TAg)-mediated transformation is dependent on binding to p53 and the retinoblastoma tumor suppressor protein (pRB) and inactivating their growth suppressive functions. Transformation minimally requires three regions of TAg: a C-terminal domain that mediates binding to p53; the LXCXE motif (residues 103-107), necessary for binding to pRB and the related proteins p107 and p130; and an N-terminal domain (residues 1-82) that contains homology to the J domain found in cellular DnaJ/Hsp40 molecular chaperone proteins. We have found that the N-terminal J domain of T Ag cooperates with the LXCXE motif to inactivate the growth suppressive functions of the pRB-related proteins.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Cell Transformation, Viral , Simian virus 40/metabolism , Animals , Antigens, Polyomavirus Transforming/chemistry , Binding Sites/physiology , HSP70 Heat-Shock Proteins/metabolism , Humans , Protein Binding , Retinoblastoma Protein/metabolism , Simian virus 40/immunology
16.
J Virol ; 73(7): 5826-32, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10364334

ABSTRACT

Variations in the polyomavirus major capsid protein VP1 underlie important biological differences between highly pathogenic large-plaque and relatively nonpathogenic small-plaque strains. These polymorphisms constitute major determinants of virus spread in mice and also dictate previously recognized strain differences in sialyloligosaccharide binding. X-ray crystallographic studies have shown that these determinants affect binding to the sialic acids. Here we report results of further experiments designed to test the importance of specific contacts between VP1 and the carbohydrate moieties of the receptor. With minor exceptions, substitutions at positions predicted from crystallography to be important in binding the terminal alpha-2,3-linked sialic acid or the penultimate sugar (galactose) destroyed the ability of the virus to replicate in cell culture. Substitutions that prevented binding to a branched disialyloligosaccharide were found to result in viruses that were both viable in culture and tumorigenic in the mouse. Conversely, substitutions that allowed recognition and binding of the branched carbohydrate chain inhibited spread in the mouse, though the viruses remained viable in culture. Mice of five different inbred strains, all highly susceptible to large-plaque virus, showed resistance to the spread of polyomavirus strains bearing the VP1 type which binds the branched-chain receptor. We suggest that glycoproteins bearing the appropriate O-linked branched sialyloligosaccharide chains are effective pseudoreceptors in the host and that they block the spread of potentially tumorigenic or virulent virus strains.


Subject(s)
Capsid Proteins , Capsid/metabolism , N-Acetylneuraminic Acid/metabolism , Polyomavirus/metabolism , Polyomavirus/pathogenicity , Receptors, Virus/metabolism , 3T3 Cells , Amino Acid Substitution , Animals , Animals, Newborn , Cricetinae , Glutamic Acid , Mice , Mice, Inbred AKR , Mice, Inbred C3H , Mice, Inbred DBA , Polyomavirus/genetics , Polyomavirus Infections/pathology , Polyomavirus Infections/virology , Sialoglycoproteins/metabolism , Tumor Virus Infections/pathology , Tumor Virus Infections/virology , Virulence
17.
Mol Cell Biol ; 19(6): 4465-79, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10330186

ABSTRACT

cdc25C induces mitosis by activating the cdc2-cyclin B complex. The intracellular localization of cyclin B1 is regulated in a cell cycle-specific manner, and its entry into the nucleus may be required for the initiation of mitosis. To determine the cellular localization of cdc25C, monoclonal antibodies specific for cdc25C were developed and used to demonstrate that in human cells, cdc25C is retained in the cytoplasm during interphase. A deletion analysis identified a 58-amino-acid region (amino acids 201 to 258) in cdc25C that was required for the cytoplasmic localization of cdc25C. This region contained a specific binding site for 14-3-3 proteins, and mutations in cdc25C that disrupted 14-3-3 binding also disrupted the cytoplasmic localization of cdc25C during interphase. cdc25C proteins that do not contain a binding site for 14-3-3 proteins showed a pancellular localization and an increased ability to induce premature chromosome condensation. The cytoplasmic localization of cdc25C was not altered by gamma irradiation or treatment with the nuclear export inhibitor leptomycin B. These results suggest that 14-3-3 proteins may negatively regulate cdc25C function by sequestering cdc25C in the cytoplasm.


Subject(s)
Cell Cycle Proteins/analysis , Cytoplasm/chemistry , Interphase , Mitosis , Phosphoprotein Phosphatases/analysis , cdc25 Phosphatases , Antibodies, Monoclonal , Blotting, Western , Cell Cycle , Cell Cycle Proteins/immunology , Cell Cycle Proteins/physiology , Cyclin B/metabolism , Cyclin B1 , Fibroblasts/metabolism , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Humans , Mimosine/pharmacology , Nuclear Localization Signals , Osteosarcoma/metabolism , Phosphoprotein Phosphatases/immunology , Phosphoprotein Phosphatases/physiology , Plasmids , Precipitin Tests , Protein Binding , Proto-Oncogene Proteins c-myc/metabolism , Recombinant Fusion Proteins , Subcellular Fractions , Time Factors , Transfection , Tumor Cells, Cultured
18.
Cancer Res ; 59(6): 1192-5, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10096546

ABSTRACT

Previous studies have demonstrated that gamma-irradiation (IR)-induced apoptosis in multiple myeloma (MM) is associated with activation of stress-activated protein kinase (SAPK). In the present study, we examined the molecules downstream of SAPK/C-Jun N-terminal kinase (JNK), focusing on the role of retinoblastoma protein (Rb) during IR-induced MM cell apoptosis. The results demonstrate that IR activates SAPK/JNK, which associates with Rb both in vivo and in vitro. Far Western blot analysis confirms that SAPK/JNK binds directly to Rb. IR-activated SAPK/JNK phosphorylates Rb, and deletion of the phosphorylation site in the COOH terminus domain of Rb abrogates phosphorylation of Rb by SAPK/JNK. Taken together, our results suggest that Rb is a target protein of SAPK/JNK and that the association of SAPK/JNK and Rb mediates IR-induced apoptosis in MM cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinases , Multiple Myeloma/metabolism , Retinoblastoma Protein/metabolism , Apoptosis/radiation effects , Enzyme Activation/radiation effects , Gamma Rays , Humans , JNK Mitogen-Activated Protein Kinases , Multiple Myeloma/enzymology , Phosphorylation/radiation effects , Protein Binding , Signal Transduction/physiology , Tumor Cells, Cultured
19.
Proc Natl Acad Sci U S A ; 96(1): 208-13, 1999 Jan 05.
Article in English | MEDLINE | ID: mdl-9874797

ABSTRACT

Steroid receptor coactivator-1 (SRC-1) family members interact with steroid receptors, including estrogen receptor alpha (ERalpha) and progesterone receptor (PR), to enhance ligand-dependent transcription. However, the expression of ERalpha and SRC-1 was found to be segregated in distinct subsets of cells within the epithelium of the estrogen-responsive rat mammary gland. This finding was in contrast to the finding for the stroma, where significant numbers of cells coexpressed ERalpha and SRC-1. Treatment of animals with estrogen induced PR expression in the ERalpha-expressing mammary epithelial cells in the absence of detectable SRC-1 and did not affect the segregated pattern of SRC-1 and ERalpha expression. PR was neither expressed nor induced by estrogen treatment in stroma, despite the coexpression of ERalpha and SRC-1. These results suggest that SRC-1 is not necessary for ERalpha-mediated induction of PR in mammary epithelial cells and is also not sufficient for PR induction in stromal cells expressing both ERalpha and SRC-1. Furthermore, the expression of SRC-1 in a subpopulation of mammary epithelial cells distinct from those expressing ERalpha or PR raises the possibility that SRC-1 has cell type-specific functions other than simply to act as coactivator for ERalpha or PR in the mammary epithelium.


Subject(s)
Mammary Glands, Animal/chemistry , Receptors, Steroid/isolation & purification , Transcription Factors/isolation & purification , Animals , Epithelial Cells/chemistry , Estrogens/pharmacology , Female , Gene Expression , Histone Acetyltransferases , Immunohistochemistry , Mammary Glands, Animal/drug effects , Nuclear Receptor Coactivator 1 , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/isolation & purification , Receptors, Progesterone/isolation & purification , Stromal Cells/chemistry
20.
Mol Cell Biol ; 18(11): 6316-24, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9774648

ABSTRACT

p73 is a recently identified member of the p53 family. Previously it was shown that p73 can, when overproduced in p53-defective tumor cells, activate p53-responsive promoters and induce apoptosis. In this report we describe the generation of anti-p73 monoclonal antibodies and confirm that two previously described p73 isoforms are produced in mammalian cells. Furthermore, we show that these two isoforms can bind to canonical p53 DNA-binding sites in electrophoretic mobility shift assays. Despite the high degree of similarity between p53 and p73, we found that adenovirus E1B 55K, simian virus 40 T, and human papillomavirus E6 do not physically interact with p73. The observation that viral oncoproteins discriminate between p53 and p73 suggests that the functions of these two proteins may differ under physiological conditions. Furthermore, they suggest that inactivation of p73 may not be required for transformation.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Tumor Suppressor Protein p53/metabolism , Adenoviridae/metabolism , Antibodies, Monoclonal/metabolism , Binding Sites/genetics , DNA-Binding Proteins/immunology , Genes, Tumor Suppressor , Humans , Nuclear Proteins/immunology , Papillomaviridae/metabolism , Recombinant Fusion Proteins/genetics , Simian virus 40/metabolism , Transfection/genetics , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...