Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Subcell Biochem ; 85: 263-80, 2014.
Article in English | MEDLINE | ID: mdl-25201200

ABSTRACT

Discovered in 1987 and 1997 respectively, Mdm2 and MdmX represent two critical cellular regulators of the p53 tumor suppressor. This chapter reviews each from initial discovery to our current understanding of their deregulation in human cancer with a focus on how each regulator impacts p53 function. While p53 independent activities of Mdm2 and MdmX are noted the reader is directed to other reviews on this topic. The chapter concludes with an examination of the various mechanisms of Mdm-deregulation and an assessment of the current therapeutic approaches to target Mdm2 and MdmX overexpression.


Subject(s)
Neoplasms/physiopathology , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-mdm2/physiology , Proto-Oncogene Proteins/physiology , Alternative Splicing , Animals , Cell Cycle Proteins , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Neoplasms/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/genetics
2.
PLoS One ; 7(8): e42034, 2012.
Article in English | MEDLINE | ID: mdl-22870278

ABSTRACT

BACKGROUND: MDM4, also called MDMX or HDMX in humans, is an important negative regulator of the p53 tumor suppressor. MDM4 is overexpressed in about 17% of all cancers and more frequently in some types, such as colon cancer or retinoblastoma. MDM4 is known to be post-translationally regulated by MDM2-mediated ubiquitination to decrease its protein levels in response to genotoxic stress, resulting in accumulation and activation of p53. At the transcriptional level, MDM4 gene regulation has been less clearly understood. We have reported that DNA damage triggers loss of MDM4 mRNA and a concurrent increase in p53 activity. These experiments attempt to determine a mechanism for down-regulation of MDM4 mRNA. METHODOLOGY/PRINCIPAL FINDINGS: Here we report that MDM4 mRNA is a target of hsa-mir-34a (miR-34a). MDM4 mRNA contains a lengthy 3' untranslated region; however, we find that it is a miR-34a site within the open reading frame (ORF) of exon 11 that is responsible for the repression. Overexpression of miR-34a, but not a mutant miR-34a, is sufficient to decrease MDM4 mRNA levels to an extent identical to those of known miR-34a target genes. Likewise, MDM4 protein levels are decreased by miR-34a overexpression. Inhibition of endogenous miR-34a increased expression of miR-34a target genes and MDM4. A portion of MDM4 exon 11 containing this 8mer-A1 miR-34a site fused to a luciferase reporter gene is sufficient to confer responsiveness, being inhibited by additional expression of exogenous mir-34a and activated by inhibition of miR-34a. CONCLUSIONS/SIGNIFICANCE: These data establish a mechanism for the observed DNA damage-induced negative regulation of MDM4 and potentially provide a novel means to manipulate MDM4 expression without introducing DNA damage.


Subject(s)
Gene Expression Regulation/physiology , MicroRNAs/metabolism , Nuclear Proteins/biosynthesis , Open Reading Frames , Proto-Oncogene Proteins/biosynthesis , RNA, Messenger/metabolism , Ubiquitination/physiology , Cell Cycle Proteins , Cell Line, Tumor , DNA Damage/physiology , Humans , MicroRNAs/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
3.
Int J Cancer ; 130(10): 2291-9, 2012 May 15.
Article in English | MEDLINE | ID: mdl-21671470

ABSTRACT

Estrogen signaling plays an important role in breast carcinogenesis. An increased understanding of estrogen gene targets and their effects will allow for more directed and effective therapies for individuals with breast cancer, particularly those with estrogen receptor positive tumors resistant to tamoxifen therapy. Here, we identify YPEL3 as a growth suppressive protein downregulated by estrogen in estrogen receptor positive breast cancer cell lines. Estrogen repression of YPEL3 expression was found to be independent of p53 but dependent on estrogen receptor alpha expression. Importantly, YPEL3 expression, which is induced by the removal of estrogen or treatment with tamoxifen triggers cellular senescence in MCF-7 cells while loss of YPEL3 increases the growth rate of MCF-7 cells. Taken together these findings suggest that YPEL3 may represent a potential target for directed hormonal therapy for estrogen receptor positive breast cancer patients.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Cellular Senescence/drug effects , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , Tumor Suppressor Proteins/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Estrogens , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms, Hormone-Dependent , Tumor Suppressor Proteins/metabolism
4.
Am J Cancer Res ; 1(1): 62-70, 2011.
Article in English | MEDLINE | ID: mdl-21743838

ABSTRACT

The Fragile Histidine Triad gene or FHIT functions as tumor suppressor in many epithelial cell types. Although its tumor suppressive mechanism is the subject of intense study, less is known about how FHIT gene expression itself is regulated. Here we show that PI3 kinase and its downstream target AKT suppress FHIT gene expression in response to growth factor stimulation in actively cycling cells. Upon removal of mitogens from the culture environment, FHIT mRNA and protein levels are observed to increase as a result of derepression from these protooncogenic kinases. AKT signaling through the FOXO transcription factors appears to be the basis for FHIT gene regulation. Increases in FHIT gene expression are directly dependent on endogenous FOXO3a in MCF7 breast carcinoma cells as evidenced by experiments with RNAi targeting FOXO transcription factor family members. Thus, this is the first report demonstrating that FHIT gene expression is normally repressed in actively cycling cells through the PI3K/AKT/FOXO3a axis.

5.
Ann Surg Oncol ; 18(6): 1791-6, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21267786

ABSTRACT

BACKGROUND: Previous work has demonstrated YPEL3 to be a growth-suppressive protein that acts through a pathway of cellular senescence. We set out to determine whether human colon tumors demonstrated downregulation of YPEL3. METHODS: We collected colon tumor samples with matched normal control samples and analyzed them for YPEL3 gene expression by reverse transcriptase-polymerase chain reaction and CpG hypermethylation of the YPEL3 promoter by base-specific polymerase chain reaction analysis. Colon cancer cell lines (Caco-2 and HCT116(-/-) p53) were used to assess YPEL3 gene expression after treatment with 5-azadeoxycytidine or trichostatin A. RESULTS: Reverse transcriptase-polymerase chain reaction analysis demonstrated a decrease in YPEL3 expression in tumor samples when compared to their patient-matched normal tissue. We determined that DNA methylation of the YPEL3 promoter CpG island does not play a role in YPEL3 regulation in human colon tumors or colon cancer cells lines, consistent with the inability of 5-azadeoxycytidine treatment to induce YPEL3 expression in colon cancer cell lines. In contrast, colon cell line results suggest that histone acetylation may play a role in YPEL3 regulation in colon cancer. CONCLUSIONS: YPEL3 is preferentially downregulated in human colon adenocarcinomas. DNA hypermethylation does not appear to be the mechanism of YPEL3 downregulation in this subset of collected patient samples or in colon cell lines. Histone acetylation may be a relevant epigenetic modulator of YPEL3 in colon adenocarcinomas. Future investigation of YPEL3 and its role in colon cancer signaling and development may lead to increased understanding and alternative treatment options for this disease.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Adenocarcinoma/genetics , Carcinoma, Signet Ring Cell/genetics , Cellular Senescence , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Tumor Suppressor Proteins/genetics , Adult , Aged , Aged, 80 and over , Case-Control Studies , Colon/metabolism , CpG Islands , DNA Methylation , Down-Regulation , Female , Humans , Male , Middle Aged , Signal Transduction , Tumor Cells, Cultured
6.
Front Biosci (Landmark Ed) ; 16(5): 1746-51, 2011 01 01.
Article in English | MEDLINE | ID: mdl-21196260

ABSTRACT

Yippee-like 3 (YPEL3) was reported in 2004 as one of five family members of the Yippee protein with conservation in species down to slime molds. While reports of other YPEL family members have surfaced our laboratory was the first to report that YPEL3 is induced by the p53 tumor suppressor. Furthermore we demonstrated that YPEL3 is growth suppressive, triggering cellular senescence in human cell lines and is down-regulated in several human tumors. Studies with mouse YPEL3, originally named small unstable apoptotic protein (SUAP), confirmed that the gene encodes a growth suppressive highly unstable protein. In this review we show that transcriptionally active forms of p73 and p63, family members of p53, can transactivate the human YPEL3 gene. While there are several reported YPEL3 transcripts and potentially 2 protein isoforms, no clear protein structure has been reported. As evidence mounts that YPEL3 is a tumor suppressor gene, studies aimed at understanding its biological function, regulation of gene expression and impact on tumorigenesis will help.


Subject(s)
Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/physiology , Animals , Apoptosis Regulatory Proteins , Cellular Senescence/genetics , Cellular Senescence/physiology , Genes, Tumor Suppressor , Genes, p53 , Humans , Mice , Proteins/physiology , Tumor Suppressor Proteins/genetics
8.
Cell Cycle ; 9(16): 3376-82, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20724842

ABSTRACT

Cellular senescence is an irreversible state of terminal growth arrest that requires functional p53. Acting to block tumor formation, induction of senescence has also been demonstrated to contribute to tumor clearance via the immune system following p53 reactivation. The Hdm2-antagonist, Nutlin-3a, has been shown to reactivate p53 and induce a quiescent state in various cancer cell lines, similar to the G(1) arrest observed upon RNAi targeting of Hdm2 in MCF7 breast cancer. In the present study we show that HdmX, a negative regulator of p53, impacts the senescence pathway. Specifically, overexpression of HdmX blocks Ras mediated senescence in primary human fibroblasts. The interaction of HdmX with p53 and the re-localization of HdmX to the nucleus through Hdm2 association appear to be required for this activity. Furthermore, inhibiting HdmX in prostate adenocarcinoma cells expressing wild-type p53, mutant Ras and high levels of HdmX induced cellular senescence as measured by an increase in irreversible b-galactosidase staining. Together these results suggest that HdmX overexpression may contribute to tumor formation by blocking senescence and that targeting HdmX may represent an attractive anti-cancer therapeutic approach.


Subject(s)
Cellular Senescence , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Cell Cycle Proteins , Cell Line , Fibroblasts/metabolism , Humans , Mutation , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , RNA Interference , Tumor Suppressor Protein p53/metabolism
9.
Cancer Res ; 70(9): 3566-75, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20388804

ABSTRACT

Cellular senescence, the limited ability of cultured normal cells to divide, can result from cellular damage triggered through oncogene activation (premature senescence) or the loss of telomeres following successive rounds of DNA replication (replicative senescence). Although both processes require a functional p53 signaling pathway, relevant downstream p53 targets have been difficult to identify. Discovery of senescence activators is important because induction of tumor cell senescence may represent a therapeutic approach for the treatment of cancer. In microarray studies in which p53 was reactivated in MCF7 cells, we discovered that Yippee-like-3 (YPEL3), a member of a recently discovered family of putative zinc finger motif coding genes consisting of YPEL1-5, is a p53-regulated gene. YPEL3 expression induced by DNA damage leads to p53 recruitment to a cis-acting DNA response element located near the human YPEL3 promoter. Physiologic induction of YPEL3 results in a substantial decrease in cell viability associated with an increase in cellular senescence. Through the use of RNAi and H-ras induction of cellular senescence, we show that YPEL3 activates cellular senescence downstream of p53. Consistent with its growth suppressive activity, YPEL3 gene expression is repressed in ovarian tumor samples. One mechanism of YPEL3 downregulation in ovarian tumor cell lines seems to be hypermethylation of a CpG island upstream of the YPEL3 promoter. We believe these findings point to YPEL3 being a novel tumor suppressor, which upon induction triggers a permanent growth arrest in human tumor and normal cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Cell Line, Tumor , Cellular Senescence/genetics , CpG Islands , DNA Methylation , HCT116 Cells , Hep G2 Cells , Humans , Neoplasms/metabolism , Neoplasms/pathology , Tumor Suppressor Proteins/biosynthesis
10.
Aging (Albany NY) ; 1(1): 89-108, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19946469

ABSTRACT

While half of all human tumors possess p53 mutations, inactivation of wild-type p53 can also occur through a variety of mechanisms that do not involve p53 gene mutation or deletion. Our laboratory has been interested in tumor cells possessing wild-type p53 protein and elevated levels of HdmX and/or Hdm2, two critical negative regulators of p53 function. In this study we utilized RNAi to knockdown HdmX or Hdm2 in MCF7 human breast cancer cells, which harbor wild-type p53 and elevated levels of HdmX and Hdm2 then examined gene expression changes and effects on cell growth.Cell cycle and growth assays confirmed that the loss of either HdmX or Hdm2 led to a significant growth inhibition and G1cell cycle arrest. Although the removal of overexpressed HdmX/2 appears limited to an anti-proliferative effect in MCF7cells, the loss of HdmX and/or Hdm2 enhanced cytotoxicity in these same cells exposed to DNA damage. Through the use of Affymetrix GeneChips and subsequent RT-qPCR validations, we uncovered a subset of anti-proliferative p53 target genes activated upon HdmX/2 knockdown. Interestingly, a second set of genes, normally transactivated by E2F1 as cells transverse the G1-S phase boundary, were found repressed in a p21-dependent manner following HdmX/2 knockdown.Taken together, these results provide novel insights into the reactivation of p53 in cells overexpressing HdmX and Hdm2.


Subject(s)
DNA Damage/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/physiology , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins/genetics , RNA Interference , Tumor Suppressor Protein p53/genetics , Actins/genetics , Breast Neoplasms/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Cell Survival/genetics , Cisplatin/pharmacology , Cyclin A2/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/drug effects , Down-Regulation/genetics , Doxorubicin/pharmacology , E2F1 Transcription Factor/genetics , Female , G1 Phase/genetics , Gene Expression/genetics , Humans , Immediate-Early Proteins/genetics , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Up-Regulation/genetics
11.
Toxicol Sci ; 95(2): 495-510, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17085751

ABSTRACT

The jet fuel jet propulsion fuel 8 (JP-8) has been shown to cause an inflammatory response in the skin, which is characterized histologically by erythema, edema, and hyperplasia. Studies in laboratory animal skin and cultured keratinocytes have identified a variety of changes in protein levels related to inflammation, oxidative damage, apoptosis, and cellular growth. Most of these studies have focused on prolonged exposures and subsequent effects. In an attempt to understand the earliest responses of the skin to JP-8, we have investigated changes in gene expression in the epidermis for up to 8 h after a 1-h cutaneous exposure in rats. After exposure, we separated the epidermis from the rest of the skin with a cryotome and isolated total mRNA. Gene expression was studied with microarray techniques, and changes from sham treatments were analyzed and characterized. We found consistent twofold increases in gene expression of 27 transcripts at 1, 4, and 8 h after the beginning of the 1-h exposure that were related primarily to structural proteins, cell signaling, inflammatory mediators, growth factors, and enzymes. Analysis of pathways changed showed that several signaling pathways were increased at 1 h and that the most significant changes at 8 h were in metabolic pathways, many of which were downregulated. These results confirm and expand many of the previous molecular studies with JP-8. Based on the 1-h changes in gene expression, we hypothesize that the trigger of the JP-8-induced, epidermal stress response is a physical disruption of osmotic, oxidative, and membrane stability which activates gene expression in the signaling pathways and results in the inflammatory, apoptotic, and growth responses that have been previously identified.


Subject(s)
Epidermis/drug effects , Gene Expression Profiling , Gene Expression/drug effects , Hydrocarbons/toxicity , Administration, Cutaneous , Animals , Epidermis/metabolism , Male , Oligonucleotide Array Sequence Analysis , Oxidative Stress/drug effects , Oxidative Stress/genetics , RNA/genetics , RNA/metabolism , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Time Factors
12.
Biomark Insights ; 2: 253-9, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-19662208

ABSTRACT

Gene expression profiles from DNA microarrays can provide molecular signatures that improve tumor classification, prognosis, and treatment options. While much of this work has focused on isolation of RNA from the resected tumor, fewer studies have utilized RNA from fine needle aspirates (FNA). In this pilot study we examined whether the gene signatures obtained from FNA samples would correlate with signatures taken from the resected tumor. Based on NSCLC gene expression profiles obtained from eleven sets of FNA and tumor samples we obtained a high concordance of FNA profiles matching their matched tumor sample. These results suggest that FNA samples may provide informative gene expression signatures regarding the potential aggressiveness of non-small-cell lung carcinomas.

14.
Cancer Biol Ther ; 5(7): 815-23, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16760658

ABSTRACT

The glycolytic inhibitor 2-deoxy-D-glucose (2-DG) has been shown to enhance the cell death induced by radiation and other DNA damaging agents selectively in cells with high rates of glycolysis, like cancer cells. While energy linked modification of DNA and cellular repair processes have been suggested as possible mechanisms of sensitization, other effects such as global stress response cannot be excluded. In this pilot study, we have investigated the effect of 2-DG and radiation on the transcriptome in an attempt to elucidate how 2-DG impacts gene expression in undamaged verses irradiation (IR) damaged cells using a human malignant glioma cell line, U-87. Exponentially growing U-87 cells were exposed to various combinations of 2-DG and X-rays and total RNA was isolated four hours after exposure. Gene expression changes were elucidated using Affymetrix GeneChips. As expected, U-87 cells treated with 2-DG showed activation of several endoplasmic reticulum stress response genes. Selective RT-PCR and Western blotting confirmed these gene alterations. Given that glucose deprivation leads to p53 activation and 2-DG led to activation of p53 response genes in our present study (e.g., PMAIP1 and GADD45A), we examined the impact of transient p53 knockdown and observed that induction of PMAIP1 and GADD45A appear to be via p53-independent mechanisms. The majority of gene alterations seen with IR-treatment alone were consistent with previous reports. While most gene alterations seen with 2-DG and IR dual treatment were confirmed in the gene profiles seen with individual (2-DG or IR) treatments, several genes appeared differentially regulated between IR and 2-DG (e.g., DUSP8, IL8, GADD45B). Additionally, gene expression patterns suggested alterations in cell cycle regulation, apoptosis, and cytokine signaling pathways. Taken together, this study provides new insights into how the transcriptome of tumor cells are likely to be affected by a combined stress caused by IR and 2-DG.


Subject(s)
Deoxyglucose/pharmacology , Gene Expression Profiling , Glioma/genetics , Glioma/radiotherapy , Radiation Tolerance/drug effects , Transcription, Genetic/drug effects , Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , Endoplasmic Reticulum/genetics , Gene Expression/drug effects , Gene Expression/radiation effects , Glioma/metabolism , Glycolysis/drug effects , Humans , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription, Genetic/radiation effects , Tumor Suppressor Protein p53/agonists , X-Rays
15.
Cell Cycle ; 4(4): 604-8, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15876864

ABSTRACT

Mdm2, by virtue of an intrinsic E3 ubiquitin ligase activity, is capable of autoubiquitination and the ubiquitination of the p53 tumor suppressor protein. Additionally, Hdm2 has been reported to undergo a p14ARF-dependent sumoylation with concurrent Hdm2 stabilization. In this present work, we report that MdmX can undergo ARF-mediated sumoylation similar to that reported for Mdm2. When coexpressed, MdmX overexpression results in a dose-dependent inhibition of Mdm2 sumoylation and a concurrent increase in Mdm2 ubiquitination. This switch from Mdm2 sumoylation to Mdm2 ubiquitination may explain the destablization of Mdm2 previously observed in cells overexpressing both ARF and MdmX. Given that MdmX can heterodimerize with Mdm2 and separately associate with ARF we employed a series of MdmX mutants to examine how MdmX blocks Mdm2 sumoylation. A MdmX miniprotein capable of binding to ARF, but not p53 or Mdm2 was able to competitively inhibit Mdm2 sumoylation and reverse ARF mediated activation of p53 transactivation. Taken together, these results demonstrate that MdmX can affect post-translational modification and stability of Mdm2 and p53 activity through interaction with ARF.


Subject(s)
Gene Expression Regulation , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/physiology , Tumor Suppressor Protein p14ARF/chemistry , Blotting, Western , Cell Cycle , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16 , Dimerization , Genes, Reporter , Genes, p53 , Humans , Luciferases/metabolism , Nuclear Proteins/metabolism , Plasmids/metabolism , Protein Binding , Protein Processing, Post-Translational , Proto-Oncogene Proteins , Smad Proteins/metabolism , Transcriptional Activation , Transfection , Tumor Suppressor Protein p53/metabolism , Ubiquitin/chemistry
16.
Cell Cycle ; 3(4): 472-8, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14739777

ABSTRACT

Based on knockout mouse studies, Mdm2 and MdmX have been identified as critical regulators of the p53 tumor suppressor protein, at least during early development. While many of the functions attributed to Mdm2 and MdmX involve p53 and overexpression of each gene appears to have oncogenic activities, a number of studies have suggested that each protein also possesses p53-independent functions. While examining the effect of Mdm2 overexpression on E2F1 transactivation we uncovered a novel MdmX function, the ability to inhibit E2F1 transactivation in a p53 and Mdm2 independent manner. Using a series of MdmX deletion mutants the central region of MdmX, amino acids 128-444 appears to possess the repressive domain. While an in vivo association of MdmX with either E2F1 or DP1 was not observed, a slight reduction in DP1 and an increased cytoplasmic localization of E2F1 were seen in cells overexpressing MdmX. These results suggest that elevated MdmX expression may repress E2F1-regulated genes like p14ARF and thus represent another regulatory mechanism in the Rb-p53 signaling pathway.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Blotting, Western , Cell Differentiation , Cell Division , Cell Line, Tumor , Dimerization , Dose-Response Relationship, Drug , E2F Transcription Factors , E2F1 Transcription Factor , Gene Deletion , Humans , Luciferases/metabolism , Microscopy, Fluorescence , Plasmids/metabolism , Precipitin Tests , Protein Structure, Tertiary , Proto-Oncogene Proteins c-mdm2 , Retinoblastoma Protein/metabolism , Signal Transduction , Transfection , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase/metabolism
17.
Biochemistry ; 42(8): 2291-9, 2003 Mar 04.
Article in English | MEDLINE | ID: mdl-12600196

ABSTRACT

Mdm2 and MdmX function as cellular regulators of the p53 tumor suppressor protein. Mdm2, a p53 inducible protein, negatively regulates p53 by inhibiting p53 transcriptional activity and promoting ubiquitin mediated proteasome degradation. The Mdm2 ring finger domain has been shown to possess E3 ligase activity and to be a necessary domain for targeting p53 degradation. MdmX, a p53 binding protein sharing a high degree of structural homology with Mdm2, has emerged as another negative regulator of the p53 tumor suppressor. MdmX has also been shown to block p53 transactivation but unlike Mdm2 cannot induce p53 degradation. Since MdmX also possesses a ring finger domain that allows MdmX to associate with Mdm2, this study focused on elucidating how the ring and zinc fingers of these two proteins affected p53 function. We have generated a series of fusion proteins between Mdm2 and MdmX by swapping the ring finger domains with or without the zinc finger domains and examined how these fusions regulated p53 induced transactivation, ubiquitination, and degradation. All fusions inhibited the transcriptional activity of p53. In the absence of Mdm2, none of the fusion proteins could trigger p53 ubiquitination or degradation. However, in a cell line with endogenous Hdm2, Mdm2:X fusions containing the ring finger domain with or without the zinc finger domain demonstrated p53 ubiquitination presumably through stabilization of Hdm2. Additionally, an Mdm2:XZFRF fusion also degraded p53 when endogenous Hdm2 was present. Results from immunofluorescence studies suggest that p53 is colocalized to the cytoplasm when coexpressed with a Mdm2:X fusion (Mdm2:XZFRF) and that this fusion is capable of stabilizing endogenous Hdm2. Since none of the fusions triggered p53 ubiquitination in cells lacking Mdm2, these results indicate that the E3 ligase domain within the ring finger of Mdm2 when part of MdmX and the MdmX ring finger fused to Mdm2 were not sufficient to trigger p53 ubiquitination, in vivo.


Subject(s)
Nuclear Proteins , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Transcriptional Activation , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Ubiquitin/antagonists & inhibitors , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , Mice , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-mdm2 , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Transcriptional Activation/genetics , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/physiology , Ubiquitin/metabolism
18.
Oncogene ; 21(57): 8776-85, 2002 Dec 12.
Article in English | MEDLINE | ID: mdl-12483531

ABSTRACT

Mdm2 overexpression confers a growth promoting activity upon cells primarily by downregulating the p53 tumor suppressor protein. Nevertheless, Mdm2 deregulation has also been implicated in inhibiting TGF-beta growth repression in a p53 independent manner. Our goal in this study was to examine whether overexpression of Mdm2 or MdmX, a Mdm2-related protein, could affect Smad-induced transactivation. As downstream signaling elements of the TGF-beta pathway, Smads represent one potential target for Mdm2 and MdmX. Here we show that MdmX but not Mdm2 is capable of inhibiting Smad induced transactivation. Based on deletion mutant analysis, MdmX inhibition of Smad transactivation was independent of the p53 and Mdm2 interaction domains, yet required amino acid residues 128-444. Using TGF-beta sensitive HepG2 cells, MdmX overexpression was shown to inhibit TGF-beta induced Smad transactivation. Additionally, mouse embryo fibroblasts (MEFs) lacking p53 and MdmX showed enhanced Smad transactivation when compared to MEFs lacking either p53 or p53 and Mdm2. Interestingly, the inhibition of Smad transactivation by MdmX could be reversed by p300, a functional co-activator of Smads and a necessary factor for Mdm2 nuclear export and did not result from altered Smad localization. In vitro studies demonstrate that MdmX binds to p300 as well as Smad3 and Smad4. Taken together, these results suggest that inhibition of Smad-induced transactivation by MdmX occurs by altering Smad interaction with its coactivator p300.


Subject(s)
DNA-Binding Proteins/genetics , Nuclear Proteins , Proto-Oncogene Proteins/physiology , Trans-Activators/genetics , Transcriptional Activation/physiology , Animals , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Humans , Mice , Mice, Knockout , Mutation , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Smad Proteins , Trans-Activators/metabolism , Transforming Growth Factor beta/physiology , Tumor Cells, Cultured
19.
Oncogene ; 21(28): 4414-21, 2002 Jun 27.
Article in English | MEDLINE | ID: mdl-12080472

ABSTRACT

The transcription factor E2F1 functions as a key regulator for both cell-cycle progression and apoptosis. Mdm2, a major cellular regulator of the p53 tumor suppressor protein, is also closely involved in cell cycle and apoptosis. In addition to regulation of p53, Mdm2 has been reported to stimulate E2F1 transactivation by a mechanism that remains unclear. Here we examined how overexpression of Mdm2 alters E2F1/DP1 transactivation. Using a set of cell lines with differing p53 and Rb status we determined that Mdm2 induction of E2F1 transactivation was p53-dependent, resulting from release of repression by p53. While Mdm2 association with p53 was required to increase E2F1 transactivation, Mdm2 mediated degradation of p53 was not. p53 repression of E2F1 transactivation required a functional DNA binding and transactivation domain. Consistent with Mdm2 activation of E2F1 via an inhibition of p53 transactivation we demonstrate a concomitant reduction in p21 protein levels with Mdm2 overexpression. Furthermore, E2F1 repression by an Rb-phosphorylation mutant could not be reversed by Mdm2 overexpression. Mdm2 was also unable to enhance E2F1 transactivation in Mouse embryo fibroblasts lacking p21. Taken together, these results suggest that Mdm2 activation of E2F1 occurs through the repression of p53-dependent transcription of p21, a p53-target gene and cyclin dependent kinase inhibitor.


Subject(s)
Cell Cycle Proteins , Cyclins/metabolism , DNA-Binding Proteins , Nuclear Proteins , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation/physiology , Tumor Suppressor Protein p53/metabolism , Apoptosis , Blotting, Western , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , E2F Transcription Factors , E2F1 Transcription Factor , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Humans , Luciferases/metabolism , Plasmids , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2 , Retinoblastoma Protein/physiology , Signal Transduction , Transcription Factors/genetics , Transfection , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL