Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
J Clin Invest ; 134(15)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38875287

ABSTRACT

It is unknown which posttranscriptional regulatory mechanisms are required for oncogenic competence. Here, we show that the LIN28 family of RNA-binding proteins (RBPs), which facilitate posttranscriptional RNA metabolism within ribonucleoprotein networks, is essential for the initiation of diverse oncotypes of hepatocellular carcinoma (HCC). In HCC models driven by NRASG12V/Tp53, CTNNB1/YAP/Tp53, or AKT/Tp53, mice without Lin28a and Lin28b were markedly impaired in cancer initiation. We biochemically defined an oncofetal regulon of 15 factors connected to LIN28 through direct mRNA and protein interactions. Interestingly, all were RBPs and only 1 of 15 was a Let-7 target. Polysome profiling and reporter assays showed that LIN28B directly increased the translation of 8 of these 15 RBPs. As expected, overexpression of LIN28B and IGFBP1-3 was able to genetically rescue cancer initiation. Using this platform to probe components downstream of LIN28, we found that 8 target RBPs were able to restore NRASG12V/Tp53 cancer formation in Lin28a/Lin28b-deficient mice. Furthermore, these LIN28B targets promote cancer initiation through an increase in protein synthesis. LIN28B, central to an RNP regulon that increases translation of RBPs, is important for tumor initiation in the liver.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , RNA-Binding Proteins , Animals , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Mice , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Humans , Protein Biosynthesis , Regulon , MicroRNAs/genetics , MicroRNAs/metabolism , Gene Expression Regulation, Neoplastic , Mice, Knockout
2.
Cancers (Basel) ; 16(8)2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38672672

ABSTRACT

MYCN amplification (MNA) and disruption of tumor suppressor microRNA (TSmiR) function are key drivers of poor outcomes in neuroblastoma (NB). While MYCN and TSmiRs regulate glucose metabolism, their role in de novo fatty acid synthesis (FAS) and unsaturated FAS (UFAS) remains poorly understood. Here, we show that FAS and UFAS (U/FAS) genes FASN, ELOVL6, SCD, FADS2, and FADS1 are upregulated in high-risk (HR) NB and that their expression is associated with lower overall survival. RNA-Seq analysis of human NB cell lines revealed parallel U/FAS gene expression patterns. Consistent with this, we found that NB-related TSmiRs were predicted to target these genes extensively. We further observed that both MYC and MYCN upregulated U/FAS pathway genes while suppressing TSmiR host gene expression, suggesting a possible U/FAS regulatory network between MYCN and TSmiRs in NB. NB cells are high in de novo synthesized omega 9 (ω9) unsaturated fatty acids and low in both ω6 and ω3, suggesting a means for NB to limit cell-autonomous immune stimulation and reactive oxygen species (ROS)-driven apoptosis from ω6 and ω3 unsaturated fatty acid derivatives, respectively. We propose a model in which MYCN and TSmiRs regulate U/FAS and play an important role in NB pathology, with implications for other MYC family-driven cancers.

3.
J Clin Invest ; 131(22)2021 11 15.
Article in English | MEDLINE | ID: mdl-34779407

ABSTRACT

High expression of LIN28B is associated with aggressive malignancy and poor survival. Here, probing MYCN-amplified neuroblastoma as a model system, we showed that LIN28B expression was associated with enhanced cell migration in vitro and invasive and metastatic behavior in murine xenografts. Sequence analysis of the polyribosome fraction of LIN28B-expressing neuroblastoma cells revealed let-7-independent enrichment of transcripts encoding components of the translational and ribosomal apparatus and depletion of transcripts of neuronal developmental programs. We further observed that LIN28B utilizes both its cold shock and zinc finger RNA binding domains to preferentially interact with MYCN-induced transcripts of the ribosomal complex, enhancing their translation. These data demonstrated that LIN28B couples the MYCN-driven transcriptional program to enhanced ribosomal translation, thereby implicating LIN28B as a posttranscriptional driver of the metastatic phenotype.


Subject(s)
N-Myc Proto-Oncogene Protein/physiology , Neoplasm Metastasis , Neuroblastoma/pathology , RNA-Binding Proteins/physiology , Ribosomes/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Neuroblastoma/etiology
4.
Proc Natl Acad Sci U S A ; 117(28): 16516-16526, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32601179

ABSTRACT

LIN28B is highly expressed in neuroblastoma and promotes tumorigenesis, at least, in part, through inhibition of let-7 microRNA biogenesis. Here, we report that overexpression of either wild-type (WT) LIN28B or a LIN28B mutant that is unable to inhibit let-7 processing increases the penetrance of MYCN-induced neuroblastoma, potentiates the invasion and migration of transformed sympathetic neuroblasts, and drives distant metastases in vivo. Genome-wide chromatin immunoprecipitation coupled with massively parallel DNA sequencing (ChIP-seq) and coimmunoprecipitation experiments show that LIN28B binds active gene promoters in neuroblastoma cells through protein-protein interaction with the sequence-specific zinc-finger transcription factor ZNF143 and activates the expression of downstream targets, including transcription factors forming the adrenergic core regulatory circuitry that controls the malignant cell state in neuroblastoma as well as GSK3B and L1CAM that are involved in neuronal cell adhesion and migration. These findings reveal an unexpected let-7-independent function of LIN28B in transcriptional regulation during neuroblastoma pathogenesis.


Subject(s)
N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Animals , Animals, Genetically Modified , Cell Movement , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Neuroblastoma/physiopathology , Protein Binding , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Zebrafish
5.
Nat Cell Biol ; 19(1): 60-67, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27992407

ABSTRACT

Signalling and post-transcriptional gene control are both critical for the regulation of pluripotency, yet how they are integrated to influence cell identity remains poorly understood. LIN28 (also known as LIN28A), a highly conserved RNA-binding protein, has emerged as a central post-transcriptional regulator of cell fate through blockade of let-7 microRNA biogenesis and direct modulation of mRNA translation. Here we show that LIN28 is phosphorylated by MAPK/ERK in pluripotent stem cells, which increases its levels via post-translational stabilization. LIN28 phosphorylation had little impact on let-7 but enhanced the effect of LIN28 on its direct mRNA targets, revealing a mechanism that uncouples LIN28's let-7-dependent and -independent activities. We have linked this mechanism to the induction of pluripotency by somatic cell reprogramming and the transition from naive to primed pluripotency. Collectively, our findings indicate that MAPK/ERK directly impacts LIN28, defining an axis that connects signalling, post-transcriptional gene control, and cell fate regulation.


Subject(s)
MAP Kinase Signaling System , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA-Binding Proteins/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Blotting, Western , HeLa Cells , Humans , Immunoprecipitation , Mass Spectrometry , Mice , Mice, Transgenic , MicroRNAs/metabolism , Mouse Embryonic Stem Cells/metabolism , Phosphorylation , Protein Domains , Protein Stability
6.
Nature ; 535(7611): 246-51, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27383785

ABSTRACT

Poor prognosis in neuroblastoma is associated with genetic amplification of MYCN. MYCN is itself a target of let-7, a tumour suppressor family of microRNAs implicated in numerous cancers. LIN28B, an inhibitor of let-7 biogenesis, is overexpressed in neuroblastoma and has been reported to regulate MYCN. Here we show, however, that LIN28B is dispensable in MYCN-amplified neuroblastoma cell lines, despite de-repression of let-7. We further demonstrate that MYCN messenger RNA levels in amplified disease are exceptionally high and sufficient to sponge let-7, which reconciles the dispensability of LIN28B. We found that genetic loss of let-7 is common in neuroblastoma, inversely associated with MYCN amplification, and independently associated with poor outcomes, providing a rationale for chromosomal loss patterns in neuroblastoma. We propose that let-7 disruption by LIN28B, MYCN sponging, or genetic loss is a unifying mechanism of neuroblastoma development with broad implications for cancer pathogenesis.


Subject(s)
Gene Amplification/genetics , MicroRNAs/genetics , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , RNA-Binding Proteins/genetics , 3' Untranslated Regions/genetics , Animals , Chromosome Deletion , Female , Gene Deletion , Genes, Neoplasm/genetics , Humans , Mice , MicroRNAs/metabolism , Models, Genetic , N-Myc Proto-Oncogene Protein , Neuroblastoma/pathology , Xenograft Model Antitumor Assays
7.
Stem Cell Reports ; 3(5): 691-8, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25316190

ABSTRACT

Reprogramming of fibroblasts to induced pluripotent stem cells (iPSCs) entails a mesenchymal to epithelial transition (MET). While attempting to dissect the mechanism of MET during reprogramming, we observed that knockdown (KD) of the epithelial-to-mesenchymal transition (EMT) factor SNAI1 (SNAIL) paradoxically reduced, while overexpression enhanced, reprogramming efficiency in human cells and in mouse cells, depending on strain. We observed nuclear localization of SNAI1 at an early stage of fibroblast reprogramming and using mouse fibroblasts expressing a knockin SNAI1-YFP reporter found cells expressing SNAI1 reprogrammed at higher efficiency. We further demonstrated that SNAI1 binds the let-7 promoter, which may play a role in reduced expression of let-7 microRNAs, enforced expression of which, early in the reprogramming process, compromises efficiency. Our data reveal an unexpected role for the EMT factor SNAI1 in reprogramming somatic cells to pluripotency.


Subject(s)
Cellular Reprogramming/genetics , Epithelial-Mesenchymal Transition/genetics , Induced Pluripotent Stem Cells/metabolism , Transcription Factors/genetics , Animals , Animals, Newborn , Cell Differentiation/genetics , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression , Humans , Induced Pluripotent Stem Cells/cytology , Keratinocytes/cytology , Keratinocytes/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Microscopy, Confocal , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Snail Family Transcription Factors , Transcription Factors/metabolism
8.
Cancer Cell ; 26(2): 248-61, 2014 Aug 11.
Article in English | MEDLINE | ID: mdl-25117712

ABSTRACT

Lin28a/b are RNA-binding proteins that influence stem cell maintenance, metabolism, and oncogenesis. Poorly differentiated, aggressive cancers often overexpress Lin28, but its role in tumor initiation or maintenance has not been definitively addressed. We report that LIN28B overexpression is sufficient to initiate hepatoblastoma and hepatocellular carcinoma in murine models. We also detected Lin28b overexpression in MYC-driven hepatoblastomas, and liver-specific deletion of Lin28a/b reduced tumor burden, extended latency, and prolonged survival. Both intravenous siRNA against Lin28b and conditional Lin28b deletion reduced tumor burden and prolonged survival. Igf2bp proteins are upregulated, and Igf2bp3 is required in the context of LIN28B overexpression to promote growth. Therefore, multiple murine models demonstrate that Lin28b is both sufficient to initiate liver cancer and necessary for its maintenance.


Subject(s)
Carcinogenesis/metabolism , Hepatoblastoma/metabolism , Liver Neoplasms, Experimental/metabolism , RNA-Binding Proteins/physiology , Animals , Hepatoblastoma/pathology , Humans , Liver Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Oncogenes , Proto-Oncogene Proteins c-myc/metabolism , RNA-Binding Proteins/metabolism , Tumor Burden
9.
PLoS Genet ; 8(11): e1003105, 2012.
Article in English | MEDLINE | ID: mdl-23209448

ABSTRACT

The Zcchc11 enzyme is implicated in microRNA (miRNA) regulation. It can uridylate let-7 precursors to decrease quantities of the mature miRNA in embryonic stem cell lines, suggested to mediate stem cell maintenance. It can uridylate mature miR-26 to relieve silencing activity without impacting miRNA content in cancer cell lines, suggested to mediate cytokine and growth factor expression. Broader roles of Zcchc11 in shaping or remodeling the miRNome or in directing biological or physiological processes remain entirely speculative. We generated Zcchc11-deficient mice to address these knowledge gaps. Zcchc11 deficiency had no impact on embryogenesis or fetal development, but it significantly decreased survival and growth immediately following birth, indicating a role for this enzyme in early postnatal fitness. Deep sequencing of small RNAs from neonatal livers revealed roles of this enzyme in miRNA sequence diversity. Zcchc11 deficiency diminished the lengths and terminal uridine frequencies for diverse mature miRNAs, but it had no influence on the quantities of any miRNAs. The expression of IGF-1, a liver-derived protein essential to early growth and survival, was enhanced by Zcchc11 expression in vitro, and miRNA silencing of IGF-1 was alleviated by uridylation events observed to be Zcchc11-dependent in the neonatal liver. In neonatal mice, Zcchc11 deficiency significantly decreased IGF-1 mRNA in the liver and IGF-1 protein in the blood. We conclude that the Zcchc11-mediated terminal uridylation of mature miRNAs is pervasive and physiologically significant, especially important in the neonatal period for fostering IGF-1 expression and enhancing postnatal growth and survival. We propose that the miRNA 3' terminus is a regulatory node upon which multiple enzymes converge to direct silencing activity and tune gene expression.


Subject(s)
DNA-Binding Proteins , Insulin-Like Growth Factor I , MicroRNAs , Uridine , Animals , Cell Differentiation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Development/genetics , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Liver/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/metabolism , Uridine/genetics , Uridine/metabolism
10.
Chem Biol Drug Des ; 75(2): 223-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20028401

ABSTRACT

Mutation in the ABL kinase domain is the principal mechanism of imatinib resistance in patients with chronic myelogenous leukaemia. The second generation BCR/ABL inhibitors nilotinib and dasatinib effectively inhibit most imatinib resistance variants, but are ineffective against the gatekeeper mutant, T315I. Gatekeeper mutation activates the kinase by stabilizing the hydrophobic spine. Here, we describe that the rationally designed compound AP24163 can inhibit native and gatekeeper mutants of the BCR/ABL kinase. Structural modelling suggests that AP24163 affects the flexibility of the P-loop and destabilizes the active conformation by disrupting the hydrophobic spine. In vitro screening for drug resistance identified clones with compound mutations involving both the P-loop and T315I. Our studies provide structural insights for the design of inhibitors against the gatekeeper mutant and suggest that up-front combination therapy may be required to prevent the emergence of compound-resistant mutations.


Subject(s)
Adenine/analogs & derivatives , Benzamides/chemistry , Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/antagonists & inhibitors , Adenine/chemistry , Adenine/pharmacology , Animals , Benzamides/pharmacology , Binding Sites , Cell Line , Computer Simulation , Dasatinib , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Mice , Mutation , Piperazines/chemistry , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Pyrimidines/chemistry , Pyrimidines/pharmacology , Thiazoles/chemistry , Thiazoles/pharmacology
11.
Nat Genet ; 41(7): 843-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19483683

ABSTRACT

Multiple members of the let-7 family of miRNAs are often repressed in human cancers, thereby promoting oncogenesis by derepressing targets such as HMGA2, K-Ras and c-Myc. However, the mechanism by which let-7 miRNAs are coordinately repressed is unclear. The RNA-binding proteins LIN28 and LIN28B block let-7 precursors from being processed to mature miRNAs, suggesting that their overexpression might promote malignancy through repression of let-7. Here we show that LIN28 and LIN28B are overexpressed in primary human tumors and human cancer cell lines (overall frequency approximately 15%), and that overexpression is linked to repression of let-7 family miRNAs and derepression of let-7 targets. LIN28 and LIN28b facilitate cellular transformation in vitro, and overexpression is associated with advanced disease across multiple tumor types. Our work provides a mechanism for the coordinate repression of let-7 miRNAs observed in a subset of human cancers, and associates activation of LIN28 and LIN28B with poor clinical prognosis.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , RNA-Binding Proteins/genetics , Animals , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Humans , Liver Neoplasms/genetics , Mice , MicroRNAs/genetics
12.
Cell Cycle ; 5(8): 801-3, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16582589

ABSTRACT

Activation of the ATM DNA damage response pathway is commonly observed in a variety of early-stage neoplasias. It has been proposed that this checkpoint response functions to suppress the development of cancer. A recent report from our laboratory demonstrates that ATM does indeed function to suppress tumorigenesis by responding to at least some oncogenic stresses. Transgenic expression of Myc is found to cause DNA damage in vivo and ATM is shown to respond to this damage by inducing the accumulation and phosphorylation of p53. In the absence of ATM, p53-dependent apoptosis is reduced and epithelial tumorigenesis is accelerated in Myc transgenic mice. Deregulated expression of the E2F1 transcription factor also elicits an ATM-dependent checkpoint response that activates p53 and promotes apoptosis, although the mechanism by which E2F1 and Myc stimulate ATM may differ. These findings have relevance for understanding why the ATM pathway is activated in many human cancers, what generates the selective pressure for p53 inactivation during tumorigenesis, and why AT patients and carriers are predisposed to developing cancer.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , E2F1 Transcription Factor/physiology , Oncogenes , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/physiology , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-myc/metabolism
13.
Proc Natl Acad Sci U S A ; 103(5): 1446-51, 2006 Jan 31.
Article in English | MEDLINE | ID: mdl-16432227

ABSTRACT

Overexpression of the c-myc oncogene contributes to the development of a significant number of human cancers. In response to deregulated Myc activity, the p53 tumor suppressor is activated to promote apoptosis and inhibit tumor formation. Here we demonstrate that p53 induction in response to Myc overexpression requires the ataxia-telangiectasia mutated (ATM) kinase, a major regulator of the cellular response to DNA double-strand breaks. In a transgenic mouse model overexpressing Myc in squamous epithelial tissues, inactivation of Atm suppresses apoptosis and accelerates tumorigenesis. Deregulated Myc expression induces DNA damage in primary transgenic keratinocytes and the formation of gammaH2AX and phospho-SMC1 foci in transgenic tissue. These findings suggest that Myc overexpression causes DNA damage in vivo and that the ATM-dependent response to this damage is critical for p53 activation, apoptosis, and the suppression of tumor development.


Subject(s)
Apoptosis , Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Neoplasms/pathology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Proteins/physiology , Animals , Ataxia Telangiectasia/metabolism , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Caspase 3 , Caspases/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Cells, Cultured , Chromosomal Proteins, Non-Histone/metabolism , Comet Assay , DNA Damage , DNA-Binding Proteins/metabolism , Female , Fibroblasts/metabolism , Genotype , Histones/chemistry , Humans , Immunoblotting , Immunohistochemistry , Keratinocytes/cytology , Keratinocytes/metabolism , Lymphoma/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Neoplasms/metabolism , Oncogenes , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Thymus Gland/pathology , Time Factors , Tumor Suppressor Proteins/metabolism
14.
Mol Cancer Res ; 2(4): 203-14, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15140942

ABSTRACT

The p53 tumor suppressor protein is phosphorylated and activated by several DNA damage-inducible kinases, such as ATM, and is a key effector of the DNA damage response by promoting cell cycle arrest or apoptosis. Deregulation of the Rb-E2F1 pathway also results in the activation of p53 and the promotion of apoptosis, and this contributes to the suppression of tumor development. Here, we describe a novel connection between E2F1 and the ATM DNA damage response pathway. In primary human fibroblasts lacking functional ATM, the ability of E2F1 to induce the phosphorylation of p53 and apoptosis is impaired. In contrast, ATM status has no effect on transcriptional activation of target genes or the stimulation of DNA synthesis by E2F1. Cells containing mutant Nijmegen breakage syndrome protein (NBS1), a component of the Mre11-Rad50 DNA repair complex, also have attenuated p53 phosphorylation and apoptosis in response to E2F1 expression. Moreover, E2F1 induces ATM- and NBS1-dependent phosphorylation of the checkpoint kinase Chk2 at Thr68, a phosphorylation site that stimulates Chk2 activity. Delayed gammaH2AX phosphorylation and absence of ATM autophosphorylation at Ser1981 suggest that E2F1 stimulates ATM through a unique mechanism that is distinct from agents that cause DNA double-strand breaks. These findings identify new roles for several DNA damage response factors by demonstrating that they also participate in the oncogenic stress signaling pathway between E2F1 and p53.


Subject(s)
Apoptosis , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins , Caffeine/pharmacology , Cell Cycle Proteins/genetics , Cells, Cultured , Checkpoint Kinase 2 , E2F Transcription Factors , E2F1 Transcription Factor , Histones/metabolism , Humans , Mice , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins
15.
Mol Cell Biol ; 22(5): 1360-8, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11839803

ABSTRACT

The ARF tumor suppressor participates in a p53-dependent apoptotic pathway that is stimulated in response to some oncogenic stimuli. The E2F1 transcription factor is a critical downstream target of the Rb tumor suppressor and, when active, can promote proliferation as well as apoptosis. The finding that E2F1 transcriptionally regulates the ARF gene has led to the suggestion that ARF contributes to E2F1-induced apoptosis. Counter to this hypothesis, this study demonstrates not only that ARF is unnecessary for E2F1 to induce apoptosis but also that inactivation of ARF actually enhances the ability of E2F1 to promote apoptosis. Inactivation of ARF also cooperates with E2F1 activity to promote entry into the S phase of the cell cycle. This relationship between ARF and E2F1 is demonstrated in transgenic epidermis in vivo and in mouse embryo fibroblast cultures in vitro. In contrast, the ability of Myc to induce apoptosis is diminished in the absence of ARF. E2F1 induces the accumulation of p53 in the absence of ARF, and this is associated with the phosphorylation of p53 on several residues. These findings demonstrate that ARF is a negative regulator of E2F1 activity and is not required for E2F1-induced apoptosis.


Subject(s)
Apoptosis/physiology , Cell Cycle Proteins , DNA-Binding Proteins , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , E2F Transcription Factors , E2F1 Transcription Factor , Epidermis/metabolism , Fibroblasts/cytology , Humans , Mice , Mice, Transgenic , S Phase , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL