Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Oncogene ; 29(36): 5083-94, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20581871

ABSTRACT

Oncogene-mediated premature senescence has emerged as a potential tumor-suppressive mechanism in early cancer transitions. Many studies showed that Ras and p38 mitogen-activated protein kinase (MAPK) participate in premature senescence. Our previous work indicated that the HMG box-containing protein 1 (HBP1) transcription factor is involved in Ras- and p38 MAPK-induced premature senescence, but the mechanism of which has not yet been identified. Here, we showed that the p16(INK4A) cyclin-dependent kinase inhibitor is a novel target of HBP1 participating in Ras-induced premature senescence. The promoter of the p16(INK4A) gene contains an HBP1-binding site at position -426 to -433 bp from the transcriptional start site. HBP1 regulates the expression of the endogenous p16(INK4A) gene through direct sequence-specific binding. With HBP1 expression and the subsequent increase of p16(INK4A) gene expression, Ras induces premature senescence in primary cells. The data suggest a model in which Ras and p38 MAPK signaling engage HBP1 and p16(INK4A) to trigger premature senescence. In addition, we report that HBP1 knockdown is also required for Ras-induced transformation. All the data indicate that the mechanism of HBP1-mediated transcriptional regulation is important for not only premature senescence but also tumorigenesis.


Subject(s)
Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , High Mobility Group Proteins/physiology , Oncogene Protein p21(ras)/physiology , Repressor Proteins/physiology , Animals , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Expression Regulation , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Models, Biological , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Time Factors , Transcription Factors/metabolism , Transcription Factors/physiology , Transplantation, Heterologous , Up-Regulation
2.
Br J Cancer ; 92(12): 2153-9, 2005 Jun 20.
Article in English | MEDLINE | ID: mdl-15942628

ABSTRACT

The reduction or loss of plakoglobin expression in late-stage bladder cancer has been correlated with poor survival where upregulation of this catenin member by histone deacetylase inhibitors has been shown to accompany tumour suppression in an in vivo model. In this study, we directly addressed the question of the role of plakoglobin in bladder tumorigenesis following restoration, or knockdown of expression in bladder carcinoma cell lines. Restoration of plakoglobin expression resulted in a reduction in migration and suppression of tumorigenic potential in vivo. Immunocytochemistry revealed cytoplasmic and membranous localisation of plakoglobin in transfectants with < 1% of cells displaying detectable nuclear localisation of plakoglobin. siRNA knockdown experiments targeting plakoglobin, revealed enhanced migration in all cell lines in the presence and absence of E-cadherin expression. In bladder cell lines expressing low levels of plakoglobin and desmoglein-2, elevated levels of desmoglein-2 were detected following restoration of plakoglobin expression in transfected cell lines. Analysis of wnt signalling revealed no activation event associated with plakoglobin expression in the bladder model. These results show that plakoglobin acts as a tumour suppressor gene in bladder carcinoma cells and the silencing of plakoglobin gene expression in late-stage bladder cancer is a primary event in tumour progression.


Subject(s)
Cytoskeletal Proteins/genetics , Genes, Tumor Suppressor/physiology , Neoplasm Invasiveness/genetics , Urinary Bladder Neoplasms/genetics , Animals , Cadherins/biosynthesis , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Cytoskeletal Proteins/biosynthesis , Desmoglein 2 , Desmogleins , Desmoplakins , Disease Progression , Down-Regulation , Gene Expression , Gene Silencing , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Models, Animal , Neoplasm Staging , Signal Transduction/genetics , Transfection , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Wnt Proteins , gamma Catenin
3.
EMBO J ; 20(16): 4500-11, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11500377

ABSTRACT

In certain cancers, constitutive Wnt signaling results from mutation in one or more pathway components. The result is the accumulation and nuclear localization of beta-catenin, which interacts with the lymphoid enhancer factor-1 (LEF)/T-cell factor (TCF) family of HMG-box transcription factors, which activate important growth regulatory genes, including cyclin D1 and c-myc. As exemplified by APC and axin, the negative regulation of beta-catenin is important for tumor suppression. Another potential mode of negative regulation is transcriptional repression of cyclin D1 and other Wnt target genes. In mammals, the transcriptional repressors in the Wnt pathway are not well defined. We have previously identified HBP1 as an HMG-box repressor and a cell cycle inhibitor. Here, we show that HBP1 is a repressor of the cyclin D1 gene and inhibits the Wnt signaling pathway. The inhibition of Wnt signaling and growth requires a common domain of HBP1. The apparent mechanism is an inhibition of TCF/LEF DNA binding through a physical interaction with HBP1. These data suggest that the suppression of Wnt signaling by HBP1 may be a mechanism to prevent inappropriate proliferation.


Subject(s)
Cytoskeletal Proteins/metabolism , Gene Expression Regulation, Neoplastic , High Mobility Group Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction , Trans-Activators , Zebrafish Proteins , Caco-2 Cells , Cell Line, Transformed , Cyclin D1/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , Growth Inhibitors , High Mobility Group Proteins/genetics , Humans , Lymphoid Enhancer-Binding Factor 1 , Repressor Proteins/genetics , TCF Transcription Factors , Transcription Factor 7-Like 2 Protein , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Wnt Proteins , beta Catenin
4.
Mol Cell Biol ; 21(17): 5723-32, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11486012

ABSTRACT

We previously isolated HBP1 as a target of the retinoblastoma (RB) and p130 family members and as the first of the HMG box transcriptional repressors. Our subsequent work demonstrated that HBP1 coordinates differentiation in cell culture models. In the present study, we show that HBP1 regulates proliferation in a differentiated tissue of an animal. Using transgenic mice in which HBP1 expression was specifically increased in hepatocytes under control of the transthyretin promoter, we determined the impact of HBP1 on synchronous cell cycle reentry following partial hepatectomy. Modest overexpression of HBP1 yielded a detectable cell cycle phenotype. Following a mitogenic stimulus induced by two-thirds partial hepatectomy, mice expressing the HBP1 transgene showed a 10- to 12-h delay in progression through G(1) to the peak of S phase. There was a concomitant delay in mid-G(1) events, such as the induction of cyclin E. While the delay in G(1) and S phases correlated with the slight overexpression of transgenic HBP1, the level of the endogenous HBP1 protein itself declined in S phase. In contrast, the onset of the immediate-early response following partial hepatectomy was unchanged in HBP1 transgenic mice. This observation indicated that the observed delay in S phase did not result from changes in signaling pathways leading into the G(0)-to-G(1) transition. Finally, transgenic mice expressing a mutant HBP1 lacking the N-terminal RB interacting domain showed a stronger S-phase response following partial hepatectomy. These results provide the first evidence that HBP1 can regulate cell cycle progression in differentiated tissues.


Subject(s)
High Mobility Group Proteins/metabolism , Liver/cytology , Repressor Proteins/metabolism , Animals , Cell Differentiation , Cell Division , Female , G1 Phase , Gene Expression , Genes, Immediate-Early , Hepatectomy , High Mobility Group Proteins/genetics , Liver Regeneration , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/genetics , Repressor Proteins/genetics , S Phase
6.
Mol Cell Biol ; 19(3): 2400-7, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10022926

ABSTRACT

Human cyclin A1, a newly discovered cyclin, is expressed in testis and is thought to function in the meiotic cell cycle. Here, we show that the expression of human cyclin A1 and cyclin A1-associated kinase activities was regulated during the mitotic cell cycle. In the osteosarcoma cell line MG63, cyclin A1 mRNA and protein were present at very low levels in cells at the G0 phase. They increased during the progression of the cell cycle and reached the highest levels in the S and G2/M phases. Furthermore, the cyclin A1-associated histone H1 kinase activity peaked at the G2/M phase. We report that cyclin A1 could bind to important cell cycle regulators: the Rb family of proteins, the transcription factor E2F-1, and the p21 family of proteins. The in vitro interaction of cyclin A1 with E2F-1 was greatly enhanced when cyclin A1 was complexed with CDK2. Associations of cyclin A1 with Rb and E2F-1 were observed in vivo in several cell lines. When cyclin A1 was coexpressed with CDK2 in sf9 insect cells, the CDK2-cyclin A1 complex had kinase activities for histone H1, E2F-1, and the Rb family of proteins. Our results suggest that the Rb family of proteins and E2F-1 may be important targets for phosphorylation by the cyclin A1-associated kinase. Cyclin A1 may function in the mitotic cell cycle in certain cells.


Subject(s)
CDC2-CDC28 Kinases , Carrier Proteins , Cell Cycle Proteins , Cell Cycle , Cyclin A/physiology , DNA-Binding Proteins , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins , Animals , Cyclin A/genetics , Cyclin A/metabolism , Cyclin A1 , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Cyclins/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , Gene Expression Regulation , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitosis , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rabbits , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Tumor Cells, Cultured
7.
J Neurol Sci ; 159(2): 140-4, 1998 Aug 14.
Article in English | MEDLINE | ID: mdl-9741397

ABSTRACT

We studied four patients with subacute to chronic varicella zoster virus (VZV) infection of the central nervous system (CNS). VZV infection was verified by detecting antibody to VZV in the cerebrospinal fluid (CSF). VZV caused myelitis in two patients and encephalitis in two patients. In one of the patients with VZV encephalitis, in addition to VZV IgM antibody, VZV DNA was found in the CSF. Among the four patients with VZV infection of the CNS, CSF antibody to VZV was the crucial diagnostic laboratory test which corroborated the clinical features, and indicated that VZV caused neurologic disease. In addition to looking for amplifiable VZV DNA in the CSF of patients with neurologic disease whose clinical and radiologic features suggest VZV infection, we also recommend a search for CSF antibody to VZV, particularly in patients with intervals of weeks to months between zoster and the onset of neurologic disease, or in those patients without rash in whom the tempo of illness is unknown.


Subject(s)
Antibodies, Viral/cerebrospinal fluid , Encephalitis, Viral/diagnosis , Herpesvirus 3, Human/immunology , Myelitis/diagnosis , AIDS-Related Opportunistic Infections/cerebrospinal fluid , AIDS-Related Opportunistic Infections/diagnosis , AIDS-Related Opportunistic Infections/immunology , Adolescent , Adult , Encephalitis, Viral/cerebrospinal fluid , Encephalitis, Viral/immunology , Female , Humans , Male , Middle Aged , Myelitis/cerebrospinal fluid , Myelitis/immunology , Polymerase Chain Reaction , Predictive Value of Tests
8.
Mol Cell Biol ; 18(8): 4732-43, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9671483

ABSTRACT

Differentiation is a coordinated process of irreversible cell cycle exit and tissue-specific gene expression. To probe the functions of the retinoblastoma protein (RB) family in cell differentiation, we isolated HBP1 as a specific target of RB and p130. Our previous work showed that HBP1 was a transcriptional repressor and a cell cycle inhibitor. The induction of HBP1, RB, and p130 upon differentiation in the muscle C2C12 cells suggested a coordinated role. Here we report that the expression of HBP1 unexpectedly blocked muscle cell differentiation without interfering with cell cycle exit. Moreover, the expression of MyoD and myogenin, but not Myf5, was inhibited in HBP1-expressing cells. HBP1 inhibited transcriptional activation by the MyoD family members. The inhibition of MyoD family function by HBP1 required binding to RB and/or p130. Since Myf5 might function upstream of MyoD, our data suggested that HBP1 probably blocked differentiation by disrupting Myf5 function, thus preventing expression of MyoD and myogenin. Consistent with this, the expression of each MyoD family member could reverse the inhibition of differentiation by HBP1. Further investigation implicated the relative ratio of RB to HBP1 as a determinant of whether cell cycle exit or full differentiation occurred. At a low RB/HBP1 ratio cell cycle exit occurred but there was no tissue-specific gene expression. At elevated RB/HBP1 ratios full differentiation occurred. Similar changes in the RB/HBP1 ratio have been observed in normal C2 differentiation. Thus, we postulate that the relative ratio of RB to HBP1 may be one signal for activation of the MyoD family. We propose a model in which a checkpoint of positive and negative regulation may coordinate cell cycle exit with MyoD family activation to give fidelity and progression in differentiation.


Subject(s)
Cell Differentiation , DNA-Binding Proteins , High Mobility Group Proteins/metabolism , Repressor Proteins/metabolism , Retinoblastoma Protein/metabolism , Trans-Activators , Animals , Cell Line , Gene Expression Regulation , High Mobility Group Proteins/genetics , Mice , Muscle Proteins/genetics , MyoD Protein/genetics , Myogenic Regulatory Factor 5 , Repressor Proteins/genetics , Retinoblastoma Protein/genetics , Transcriptional Activation
9.
Front Biosci ; 3: D532-47, 1998 Jun 08.
Article in English | MEDLINE | ID: mdl-9616132

ABSTRACT

Cell differentiation is a coordinated process that includes cell cycle exit and the expression of unique genes to specify tissue identity. The focus of this review is the recent progress in understanding the functions of the RB family (RB, p130,p107) in cell differentiation. Much work has focused on the functions of RB in G1 regulation. However, much evidence now suggests a diverse function in differentiation. For discussion, differentiation will be divided into three general steps: cell cycle exit, apoptosis protection, and tissue-specific gene expression. These processes are coordinated to provide the final and unique tissue characteristics. The RB family and targets such as E2F and HBP1 have functions in each step. While there is much knowledge on each separate step of differentiation, the mechanisms that coordinate cell cycle and tissue-specific events are still not known. New evidence suggests that this coordination contains both positive and negative regulation of tissue-specific gene expression. RB. p130, HBP1, and other proteins appear to have unexpected functions in regulating tissue-specific gene expression. The ubiquitous expressions of these proteins suggest membership in a new and general pathway to coordinate cell cycle events with tissue-specific gene expression during differentiation. The collective observations hypothesize the existence of a differentiation checkpoint to insure fidelity.


Subject(s)
Apoptosis , Carrier Proteins , Cell Cycle Proteins , Cell Cycle , DNA-Binding Proteins , Gene Expression Regulation , Nuclear Proteins/physiology , Phosphoproteins/physiology , Proteins , Retinoblastoma Protein/physiology , Adipocytes/cytology , Animals , Cell Differentiation , Cell Line , E2F Transcription Factors , High Mobility Group Proteins/physiology , Mice , Mice, Knockout , Muscles/cytology , Nuclear Proteins/genetics , Organ Specificity , Phosphoproteins/genetics , Repressor Proteins/physiology , Retinoblastoma Protein/genetics , Retinoblastoma-Binding Protein 1 , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , Transcription Factor DP1 , Transcription Factors/physiology
10.
Oncogene ; 17(26): 3463-9, 1998 Dec 31.
Article in English | MEDLINE | ID: mdl-10030670

ABSTRACT

The Cdk inhibitor p21(WAF1/CIP1) is a negative regulator of the cell cycle, although its expression is induced by a number of mitogens that promote cell proliferation. We have found that E2F1 and E2F3, transcription factors that activate genes required for cell cycle progression, are strong activators of the p21 promoter. In contrast, HBP1 (HMG-box protein-1), a novel retinoblastoma protein-binding protein, can repress the p21 promoter and inhibit induction of p21 expression by E2F. Both E2Fs and HBP1 regulate p21 transcription through cis-acting elements located between nucleotides -119 to +16 of the p21 promoter and the DNA binding domains of each of these proteins are required for activity. Sequences between -119 and -60 basepairs containing four Sp1 consensus elements and two noncanonical E2F binding sites are of major importance for E2F activation, although E2F1 and E2F3 differ in the extent of their ability to activate expression when this segment is deleted. The opposing effects of E2Fs and HBP1 on p21 promoter activity suggest that interplay between these factors may determine the level of p21 transcription in vivo.


Subject(s)
Carrier Proteins , Cyclins/genetics , Cyclins/metabolism , DNA-Binding Proteins , Repressor Proteins/metabolism , Retinoblastoma Protein/metabolism , Transcription, Genetic , Base Sequence , Binding Sites , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , E2F Transcription Factors , E2F1 Transcription Factor , E2F2 Transcription Factor , E2F3 Transcription Factor , Gene Expression Regulation , Helix-Loop-Helix Motifs/genetics , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Repressor Proteins/genetics , Retinoblastoma-Binding Protein 1 , Sp1 Transcription Factor/metabolism , Transcription Factor DP1 , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured
11.
Genes Dev ; 11(3): 383-96, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9030690

ABSTRACT

A prominent feature of cell differentiation is the initiation and maintenance of an irreversible cell cycle arrest with the complex involvement of the retinoblastoma (RB) family (RB, p130, p107). We have isolated the HBP1 transcriptional repressor as a potential target of the RB family in differentiated cells. By homology, HBP1 is a sequence-specific HMG transcription factor, of which LEF-1 is the best-characterized family member. Several features of HBP1 suggest an intriguing role as a transcriptional and cell cycle regulator in differentiated cells. First, inspection of the HBP1 protein sequence revealed two consensus RB interaction motifs (LXCXE and IXCXE). Second, HBP1 interaction was selective for RB and p130, but not p107. HBP1, RB, and p130 levels are all up-regulated with differentiation; in contrast, p107 levels decline. Third, HBP1 can function as a transcriptional repressor of the promoter for N-MYC, which is a critical cell cycle and developmental gene. Fourth, because the activation of the N-MYC promoter in cycling cells required the E2F transcription factor, we show that E2F-1 and HBP1 represent opposite transcriptional signals that can be integrated within the N-MYC promoter. Fifth, the expression of HBP1 lead to efficient cell cycle arrest. The arrest phenotype was manifested in the presence of optimal proliferation signals, suggesting that HBP1 exerted a dominant regulatory role. Taken together, the results suggest that HBP1 may represent a unique transcriptional repressor with a role in initiation and establishment of cell cycle arrest during differentiation.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , High Mobility Group Proteins/metabolism , Proteins , Repressor Proteins/metabolism , Animals , Cell Cycle/physiology , Cell Differentiation , Cell Line , E2F Transcription Factors , E2F1 Transcription Factor , Mice , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma-Binding Protein 1 , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , Transcription Factor DP1 , Transcription Factors/metabolism
12.
J Biol Chem ; 271(21): 12261-8, 1996 May 24.
Article in English | MEDLINE | ID: mdl-8647824

ABSTRACT

Because of its expression in numerous cells, the herpes simplex virus thymidine kinase promoter (HSV-TK) is one of the best characterized promoters. Using the HSV-TK promoter as a model system, we have defined a new mode of E2F-1 transcriptional activation which utilizes the N-terminal region of E2F-1. We demonstrate that E2F-1 strongly activated HSV-TK, but in the absence of consensus E2F DNA elements. Nonetheless, E2F-1 could bind to GC-rich elements, which were conclusively identified in classic studies of HSV-TK as SP-1 sites. Second, the transcriptional activation of HSV-TK required the entire E2F-1 protein, including the N-terminal 89 amino acids. In contrast, the N-terminal 89 amino acids of E2F-1 were dispensable for transcriptional activation through consensus E2F sites. Third, we demonstrated that S phase entry is not sufficient for activation of HSV-TK by E2F-1, while the activation through consensus E2F sites is strictly linked to the cell cycle. Taken together, the activation of HSV-TK by E2F-1 proceeds by a different mechanism directed in part through the N-terminal region of E2F-1 and may be uncoupled from the known cell cycle regulatory role.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcriptional Activation , Base Sequence , Cell Cycle , Consensus Sequence , DNA , E2F Transcription Factors , E2F1 Transcription Factor , Enzyme Activation , Humans , Molecular Sequence Data , Retinoblastoma-Binding Protein 1 , Simplexvirus/enzymology , Simplexvirus/genetics , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Transcription Factor DP1 , Tumor Cells, Cultured
13.
Cell Growth Differ ; 7(1): 43-52, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8788032

ABSTRACT

Because of its critical role in the control of cell proliferation and differentiation, we postulated that E2F-1 could have a role in murine development. To this end, the organ and developmental expression of the E2F-1 transcription factor was analyzed from mid-gestation to late-stage embryogenesis. We demonstrate that the mRNA levels for E2F-1 and its heterodimeric partner DP-1 reach maximal levels in the late embryonic and early postnatal period but decline in the later postnatal and adult periods. Additionally, using high resolution in situ hybridization, high expression of E2F-1 was observed in specific cells of individual tissues, suggesting that the role of E2F-1 may be more complex than previously indicated from cell culture studies. Furthermore, the unusual pattern of E2F-1 and DP-1 developmental expression may have an essential role in certain cells and tissues in the late embryonic and early postnatal period.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins/genetics , Embryo, Mammalian/physiology , Embryonic and Fetal Development/genetics , Transcription Factors/genetics , Animals , E2F Transcription Factors , E2F1 Transcription Factor , Gene Expression Regulation, Developmental/genetics , In Situ Hybridization , Mice , RNA, Messenger/analysis , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1
14.
Mol Cell Biol ; 15(4): 2252-62, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7891719

ABSTRACT

We have examined regulation of the E2F transcription factor during differentiation of muscle cells. E2F regulates many genes involved in growth control and is also the target of regulation by diverse cellular signals, including the RB family of growth suppressors (e.g., the retinoblastoma protein [RB], p107, and p130). The following aspects of E2F function and regulation during muscle differentiation were investigated: (i) protein-protein interactions, (ii) protein levels, (iii) phosphorylation of the E2F protein, and (iv) transcriptional activity. A distinct E2F complex was present in differentiated cells but not in undifferentiated cells. The p130 protein was a prominent component of the E2F complex associated with differentiation. In contrast, in undifferentiated cells, the p107 protein was the prominent component in one of three E2F complexes. In addition, use of a differentiation-defective muscle line provided genetic and biochemical evidence that quiescence and differentiation are separable events. Exclusive formation of the E2F-p130 complex did not occur in this differentiation-defective line; however, E2F complexes diagnostic of quiescence were readily apparent. Thus, sole formation of the E2F-p130 complex is a necessary event in terminal differentiation. Other changes in E2F function and regulation upon differentiation include decreased phosphorylation and increased repression by E2F. These observations suggest that the regulation of E2F function during terminal differentiation may proceed through differential interaction within the RB family and/or phosphorylation.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Muscles/physiology , Phosphoproteins , Transcription Factors/metabolism , Cell Differentiation , Cells, Cultured , E2F Transcription Factors , Gene Expression Regulation , Macromolecular Substances , Models, Biological , Muscles/cytology , Nuclear Proteins/metabolism , Phosphorylation , Protein Binding , Proteins/metabolism , Recombinant Fusion Proteins/biosynthesis , Retinoblastoma Protein/metabolism , Retinoblastoma-Binding Protein 1 , Retinoblastoma-Like Protein p130 , Transcription Factor DP1 , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic
15.
Mol Cell Biol ; 14(12): 8420-31, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7969176

ABSTRACT

E2F-1, a member of the E2F transcription factor family, contributes to the regulation of the G1-to-S phase transition in higher eukaryotic cells. E2F-1 forms a heterodimer with DP-1 and binds to several cell cycle regulatory proteins, including the retinoblastoma family (RB, p107, p130) and cyclin A/CDK2 complexes. We have analyzed E2F-1 phosphorylation and its interaction with cyclin A/CDK2 complexes both in vivo and in vitro. In vitro, E2F-1 formed a stable complex with cyclin A/CDK2 but not with either subunit alone. DP-1 did not interact with cyclin A, CDK2, or the cyclin A/CDK2 complex. While the complex of cyclin A/CDK2 was required for stable complex formation with E2F-1, the kinase-active form of CDK2 was not required. However, E2F-1 was phosphorylated by cyclin A/CDK2 in vitro and was phosphorylated in vivo in HeLa cells. Two-dimensional tryptic phosphopeptide mapping studies demonstrated an overlap in the phosphopeptides derived from E2F-1 labeled in vitro and in vivo, indicating that cyclin A/CDK2 may be responsible for the majority of E2F-1 phosphorylation in vivo. Furthermore, an active DNA-binding complex could be reconstituted from purified E2F-1/DP-1 and cyclin A/CDK2. Binding studies conducted both in vitro and in vivo demonstrated that the cyclin A/CDK2-binding region resided within the N-terminal 124 amino acids of E2F-1. Because the stable association of E2F-1 with cyclin A/CDK2 in vitro and in vivo did not require a DP-1- or RB-binding domain and because the interactions could be reconstituted from purified components in vitro, we conclude that the interactions between cyclin A/CDK2 and E2F-1 are direct. Finally, we report that the DNA-binding activity of the E2F-1/DP-1 complex is inhibited following phosphorylation by cyclin A/CDK2.


Subject(s)
CDC2-CDC28 Kinases , Carrier Proteins , Cell Cycle Proteins , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Cyclin-Dependent Kinase 2 , E2F Transcription Factors , E2F1 Transcription Factor , HeLa Cells , Humans , In Vitro Techniques , Macromolecular Substances , Peptide Mapping , Phosphorylation , Protein Binding , Retinoblastoma-Binding Protein 1 , Structure-Activity Relationship , Transcription Factor DP1
16.
Fam Pract Res J ; 13(4): 365-72, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8285087

ABSTRACT

OBJECTIVE: The purpose of this study was to determine the prevalence of chlamydia in women attending a family planning clinic and to explore the feasibility of making chlamydia screening part of the routine procedure for all women. This study also investigated clinical and demographic parameters that may be associated with chlamydial infections. METHODS: Subjects were 239 female patients who attended the Planned Parenthood Clinic in Wichita, Kansas, during July 1990. Subjects included all patients receiving pelvic examinations regardless of indication. Each subject was screened for C. trachomatis using Testpack Chlamydia (Abbott Labs). RESULTS: Of the 239 women screened, 11 (4.6%) had positive Testpack Chlamydia tests. Young age (< 24 years), self-reported bleeding, and inflammation found on Papanicolaou exams were positively associated with chlamydial infection. CONCLUSION: These parameters provide additional information for the clinician deciding who should be screened for chlamydia infections.


Subject(s)
Chlamydia Infections/epidemiology , Chlamydia trachomatis , Adult , Ambulatory Care Facilities , Chlamydia Infections/diagnosis , Family Planning Services , Female , Humans , Prevalence
17.
DNA Cell Biol ; 11(7): 539-48, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1388726

ABSTRACT

Recent experiments in understanding the mechanism of the retinoblastoma protein (RB) function have revealed the existence of several cellular proteins that are complexed with RB. One of these cellular proteins is the E2F transcription factor, which was originally identified due to its inducibility by E1A during an adenovirus infection. The E2F recognition sequence is found in the promoters of several cellular genes involved in growth control, including several oncogenes. In this report, we provide evidence that the interaction of E2F and RB is mediated through a region on RB where viral oncogenes such as SV40 T antigen and adenovirus E1A bind and where tumorigenic mutations also cluster. Additional carboxy-terminal sequences are also required for the interaction with E2F. These observations provide evidence for a direct connection between tumor suppressor function and the gene expression program leading to cellular growth regulation.


Subject(s)
Adenovirus E1A Proteins/metabolism , Antigens, Polyomavirus Transforming/metabolism , Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Chromatography, Affinity , Chromosome Mapping , DNA/metabolism , E2F Transcription Factors , HeLa Cells , Humans , Molecular Sequence Data , Mutagenesis , Rabbits , Retinoblastoma Protein/genetics , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1
18.
Biochemistry ; 28(16): 6562-71, 1989 Aug 08.
Article in English | MEDLINE | ID: mdl-2790013

ABSTRACT

Alkylation of Torpedo californica purified nicotinic acetylcholine receptor (AChR) with N-phenylmaleimide (NPM) under nonreducing conditions led to ion flux inhibition without affecting ligand binding properties [Yee, A. S., Corey, D. E. & McNamee, M. G. (1986) Biochemistry 25, 2110-2119]. The gamma subunit was shown to be preferentially labeled by [3H]NPM with partial labeling of the alpha subunit at higher NPM concentrations. Alkylation occurs at cysteine residues as confirmed by amino acid analysis. Cyanogen bromide peptide mapping of the gamma subunit indicates that at least two residues corresponding to Cys-416, -420, or -451 are labeled. Residues 416 and 420 are part of the proposed amphipathic helix, and the functional role of these two cysteines is further investigated by site-directed mutagenesis of T. californica AChR cDNAs and expression of the mutants in Xenopus laevis oocytes following injection of SP6 transcripts. Several features of SP6 transcripts are shown to be important for efficient translation in vivo. Mutations Cys----Ser gamma 416,420 and Cys----Phe gamma 416 did not perturb either the receptor functional properties or its expression levels. The double mutant Cys----Phe gamma 416,420 displayed a 30% decrease of normalized AChR activity. The relatively small effect of large steric mutations in the amphipathic helix argues against its presence in the tightly packed transmembrane domain of the protein.


Subject(s)
Receptors, Cholinergic/metabolism , Alkylation , Animals , Binding Sites , Cloning, Molecular , Cysteine , In Vitro Techniques , Mutation , Protein Conformation , Receptors, Cholinergic/genetics , Sulfhydryl Compounds , Torpedo , Xenopus laevis
19.
Mol Cell Biol ; 9(2): 578-85, 1989 Feb.
Article in English | MEDLINE | ID: mdl-2523514

ABSTRACT

The promoter-specific factor E2F interacts with critical regulatory sequences within the adenovirus E2 promoter. In addition, the level of active factor increases markedly during a virus infection, dependent on E1A function and coincident with the trans activation of E2 transcription. We have purified the E2F factor through a combination of standard biochemical procedures and DNA affinity chromatography. The purified factor was a single polypeptide of 54,000 molecular weight, as determined by UV crosslinking and renaturation of gel-fractionated protein. Addition of affinity-purified factor to an in vitro transcription system resulted in stimulation of transcription from a promoter containing two E2F-binding sites but not promoters lacking binding sites. We thus conclude that E2F is indeed capable of stimulating transcription once it has bound to the promoter.


Subject(s)
DNA/metabolism , Oncogene Proteins, Viral/pharmacology , Transcription, Genetic/drug effects , Adenovirus Early Proteins , Binding Sites , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/pharmacology , HeLa Cells/drug effects , HeLa Cells/metabolism , Humans , Oncogene Proteins, Viral/isolation & purification , Oncogene Proteins, Viral/metabolism , Promoter Regions, Genetic/drug effects
20.
Biochem Cell Biol ; 66(6): 578-83, 1988 Jun.
Article in English | MEDLINE | ID: mdl-3048331

ABSTRACT

The 289aa product of the adenovirus E1A gene mediates the transcriptional activation of the set of early viral genes as well as several cellular genes. The E1A protein is not a DNA binding protein but, rather, acts indirectly to achieve the activation. The process of viral gene activation involves the use of cellular transcription factors, and in at least one case, in vivo assays have demonstrated a stimulation of stable promoter complex formation as a function of the E1A gene product. Analysis of transcription factors in nuclear extracts has identified a cellular factor, termed E2F, with specificity for the viral E2 promoter. The concentration of this factor increases as a result of the action of E1A. This increase in DNA binding activity does not require protein synthesis, thus indicating an E1A-mediated modification of a pre-existing factor. The E2F factor has been purified to homogeneity and is a polypeptide of 54,000 molecular weight. Analysis of an additional viral promoter, the E4 promoter, has identified a protein that interacts with sequences critical for transcription. This factor, termed E4F, is also increased as a function of the E1A product. The E4F factor has also been purified to homogeneity and has a molecular weight of 50,000. Therefore, the coordinate control of transcription by the E1A gene product involves the activation of multiple promoter specific factors.


Subject(s)
Adenoviridae/genetics , Gene Expression Regulation , Genes, Viral , Animals , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...