Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Mol Cell ; 82(1): 159-176.e12, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34847357

ABSTRACT

The MYCN oncoprotein drives the development of numerous neuroendocrine and pediatric tumors. Here we show that MYCN interacts with the nuclear RNA exosome, a 3'-5' exoribonuclease complex, and recruits the exosome to its target genes. In the absence of the exosome, MYCN-directed elongation by RNA polymerase II (RNAPII) is slow and non-productive on a large group of cell-cycle-regulated genes. During the S phase of MYCN-driven tumor cells, the exosome is required to prevent the accumulation of stalled replication forks and of double-strand breaks close to the transcription start sites. Upon depletion of the exosome, activation of ATM causes recruitment of BRCA1, which stabilizes nuclear mRNA decapping complexes, leading to MYCN-dependent transcription termination. Disruption of mRNA decapping in turn activates ATR, indicating transcription-replication conflicts. We propose that exosome recruitment by MYCN maintains productive transcription elongation during S phase and prevents transcription-replication conflicts to maintain the rapid proliferation of neuroendocrine tumor cells.


Subject(s)
Cell Nucleus/enzymology , Cell Proliferation , DNA Replication , Exosomes/enzymology , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/enzymology , RNA Polymerase II/metabolism , Transcription, Genetic , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Cell Line, Tumor , Cell Nucleus/genetics , DNA Breaks, Double-Stranded , Exoribonucleases/genetics , Exoribonucleases/metabolism , Exosomes/genetics , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Male , Mice , N-Myc Proto-Oncogene Protein/genetics , NIH 3T3 Cells , Neuroblastoma/genetics , Neuroblastoma/pathology , Promoter Regions, Genetic , RNA Caps/genetics , RNA Caps/metabolism , RNA Polymerase II/genetics , Transcription Termination, Genetic
2.
Nature ; 567(7749): 545-549, 2019 03.
Article in English | MEDLINE | ID: mdl-30894746

ABSTRACT

MYC is an oncogenic transcription factor that binds globally to active promoters and promotes transcriptional elongation by RNA polymerase II (RNAPII)1,2. Deregulated expression of the paralogous protein MYCN drives the development of neuronal and neuroendocrine tumours and is often associated with a particularly poor prognosis3. Here we show that, similar to MYC, activation of MYCN in human neuroblastoma cells induces escape of RNAPII from promoters. If the release of RNAPII from transcriptional pause sites (pause release) fails, MYCN recruits BRCA1 to promoter-proximal regions. Recruitment of BRCA1 prevents MYCN-dependent accumulation of stalled RNAPII and enhances transcriptional activation by MYCN. Mechanistically, BRCA1 stabilizes mRNA decapping complexes and enables MYCN to suppress R-loop formation in promoter-proximal regions. Recruitment of BRCA1 requires the ubiquitin-specific protease USP11, which binds specifically to MYCN when MYCN is dephosphorylated at Thr58. USP11, BRCA1 and MYCN stabilize each other on chromatin, preventing proteasomal turnover of MYCN. Because BRCA1 is highly expressed in neuronal progenitor cells during early development4 and MYC is less efficient than MYCN in recruiting BRCA1, our findings indicate that a cell-lineage-specific stress response enables MYCN-driven tumours to cope with deregulated RNAPII function.


Subject(s)
BRCA1 Protein/metabolism , N-Myc Proto-Oncogene Protein/metabolism , RNA Polymerase II/metabolism , Transcription Elongation, Genetic , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Gene Expression Regulation , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Protein Stability , Thiolester Hydrolases/metabolism
3.
EMBO J ; 36(13): 1854-1868, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28408437

ABSTRACT

Deregulated expression of MYC enhances glutamine utilization and renders cell survival dependent on glutamine, inducing "glutamine addiction". Surprisingly, colon cancer cells that express high levels of MYC due to WNT pathway mutations are not glutamine-addicted but undergo a reversible cell cycle arrest upon glutamine deprivation. We show here that glutamine deprivation suppresses translation of endogenous MYC via the 3'-UTR of the MYC mRNA, enabling escape from apoptosis. This regulation is mediated by glutamine-dependent changes in adenosine-nucleotide levels. Glutamine deprivation causes a global reduction in promoter association of RNA polymerase II (RNAPII) and slows transcriptional elongation. While activation of MYC restores binding of MYC and RNAPII function on most promoters, restoration of elongation is imperfect and activation of MYC in the absence of glutamine causes stalling of RNAPII on multiple genes, correlating with R-loop formation. Stalling of RNAPII and R-loop formation can cause DNA damage, arguing that the MYC 3'-UTR is critical for maintaining genome stability when ribonucleotide levels are low.


Subject(s)
3' Untranslated Regions , Gene Expression Regulation, Enzymologic , Glutamine/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , RNA Polymerase II/metabolism , RNA, Messenger/metabolism , Ribonucleotides/metabolism , Cell Line , Humans , Proto-Oncogene Proteins c-myc/genetics
4.
Nature ; 511(7510): 483-7, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25043018

ABSTRACT

In mammalian cells, the MYC oncoprotein binds to thousands of promoters. During mitogenic stimulation of primary lymphocytes, MYC promotes an increase in the expression of virtually all genes. In contrast, MYC-driven tumour cells differ from normal cells in the expression of specific sets of up- and downregulated genes that have considerable prognostic value. To understand this discrepancy, we studied the consequences of inducible expression and depletion of MYC in human cells and murine tumour models. Changes in MYC levels activate and repress specific sets of direct target genes that are characteristic of MYC-transformed tumour cells. Three factors account for this specificity. First, the magnitude of response parallels the change in occupancy by MYC at each promoter. Functionally distinct classes of target genes differ in the E-box sequence bound by MYC, suggesting that different cellular responses to physiological and oncogenic MYC levels are controlled by promoter affinity. Second, MYC both positively and negatively affects transcription initiation independent of its effect on transcriptional elongation. Third, complex formation with MIZ1 (also known as ZBTB17) mediates repression of multiple target genes by MYC and the ratio of MYC and MIZ1 bound to each promoter correlates with the direction of response.


Subject(s)
Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, myc/genetics , Neoplasms/genetics , Transcriptome , Up-Regulation/genetics , Animals , Binding Sites , Cell Line, Tumor , E-Box Elements/genetics , Humans , Kruppel-Like Transcription Factors/metabolism , Mice , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Inhibitors of Activated STAT/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Polymerase II/metabolism , Ubiquitin-Protein Ligases
5.
EMBO J ; 27(21): 2851-61, 2008 Nov 05.
Article in English | MEDLINE | ID: mdl-18923429

ABSTRACT

The Myc-associated zinc-finger protein, Miz1, activates transcription of the p21cip1 gene in response to UV irradiation. Miz1 associates with topoisomerase II binding protein1 (TopBP1), an essential activator of the Atr kinase. We show here that Miz1 is required for the recruitment of a fraction of TopBP1 to chromatin, for the protection of TopBP1 from proteasomal degradation and for Atr-dependent signal transduction. TopBP1 that is not bound to chromatin is degraded by the HectH9 (Mule, ARF-BP1 and HUWE1) ubiquitin ligase. Myc antagonizes the binding of TopBP1 to Miz1; as a result, expression of Myc leads to dissociation of TopBP1 from chromatin, reduces the amount of total TopBP1 and attenuates Atr-dependent signal transduction. Our data show that Miz1 and Myc affect the activity of the Atr checkpoint through their effect on TopBP1 chromatin association and stability.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Protein Inhibitors of Activated STAT/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Substitution , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Models, Biological , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Tumor Suppressor Proteins
6.
Nat Cell Biol ; 7(1): 30-41, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15580267

ABSTRACT

The transcription factor Miz1 is required for DNA-damage-induced cell-cycle arrest. We have now identified 14-3-3eta as a gene that inhibits Miz1 function through interaction with its DNA binding domain. Binding of 14-3-3eta to Miz1 depends on phosphorylation by Akt and regulates the recovery of cells from arrest after DNA damage. Miz1 has two functions in response to DNA damage: first, it is required for upregulation of a large group of genes, a function that is regulated by c-Myc, but not by 14-3-3eta; second, Miz1 represses the expression of many genes in response to DNA damage in an Akt- and 14-3-3eta-regulated manner.


Subject(s)
14-3-3 Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle/physiology , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , 14-3-3 Proteins/genetics , Animals , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation/physiology , Gene Library , HeLa Cells , Humans , Kruppel-Like Transcription Factors , Mice , Molecular Sequence Data , NIH 3T3 Cells , Phosphorylation , Protein Binding/physiology , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Rats , Transcription Factors/genetics , Up-Regulation/physiology
7.
J Cell Biol ; 172(1): 139-49, 2006 Jan 02.
Article in English | MEDLINE | ID: mdl-16391002

ABSTRACT

Myc plays a key role in homeostasis of the skin. We show that Miz1, which mediates Myc repression of gene expression, is expressed in the epidermal basal layer. A large percentage of genes regulated by the Myc-Miz1 complex in keratinocytes encode proteins involved in cell adhesion, and some, including the alpha6 and beta1 integrins, are directly bound by Myc and Miz1 in vivo. Using a Myc mutant deficient in Miz1 binding (MycV394D), we show that Miz1 is required for the effects of Myc on keratinocyte responsiveness to TGF-beta. Myc, but not MycV394D, decreases keratinocyte adhesion and spreading. In reconstituted epidermis, Myc induces differentiation and loss of cell polarization in a Miz1-dependent manner. In vivo, overexpression of beta1 integrins restores basal layer polarity and prevents Myc-induced premature differentiation. Our data show that regulation of cell adhesion is a major function of the Myc-Miz1 complex and suggest that it may contribute to Myc-induced exit from the epidermal stem cell compartment.


Subject(s)
Cell Differentiation/physiology , Genes, myc/physiology , Keratinocytes/chemistry , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Protein Inhibitors of Activated STAT/metabolism , Proto-Oncogene Proteins c-myc/physiology , Animals , Cell Adhesion/physiology , Cell Movement , Cell Polarity/genetics , Cell Polarity/physiology , Genes, myc/genetics , Humans , Integrin beta1/biosynthesis , Integrin beta1/genetics , Keratinocytes/cytology , Keratinocytes/drug effects , Mice , Multiprotein Complexes/genetics , Nuclear Proteins/genetics , Protein Inhibitors of Activated STAT/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transforming Growth Factor beta/pharmacology , Ubiquitin-Protein Ligases
8.
Trends Cell Biol ; 13(3): 146-50, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12628347

ABSTRACT

The Myc oncoprotein is a transcription factor that can both activate and repress genes. Transcriptional activation by Myc is well understood, but, by contrast, the mechanisms through which Myc represses transcription have remained elusive. Recent evidence suggests that complex formation by Myc with a zinc-finger transcription factor, Miz-1, plays an important role in mediating repression by Myc. The findings might explain how Myc interferes with cell-cycle arrest in response to TGF-beta, APC and DNA damage.


Subject(s)
Cell Cycle Proteins/genetics , Eukaryotic Cells/metabolism , Proto-Oncogene Proteins c-myc/genetics , Repressor Proteins/genetics , Silencer Elements, Transcriptional/genetics , Animals , Apoptosis/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genes, APC/physiology , Humans , Kruppel-Like Transcription Factors , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors , Tumor Suppressor Proteins/genetics
9.
Cell Rep ; 21(12): 3483-3497, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29262328

ABSTRACT

MYC proteins bind globally to active promoters and promote transcriptional elongation by RNA polymerase II (Pol II). To identify effector proteins that mediate this function, we performed mass spectrometry on N-MYC complexes in neuroblastoma cells. The analysis shows that N-MYC forms complexes with TFIIIC, TOP2A, and RAD21, a subunit of cohesin. N-MYC and TFIIIC bind to overlapping sites in thousands of Pol II promoters and intergenic regions. TFIIIC promotes association of RAD21 with N-MYC target sites and is required for N-MYC-dependent promoter escape and pause release of Pol II. Aurora-A competes with binding of TFIIIC and RAD21 to N-MYC in vitro and antagonizes association of TOP2A, TFIIIC, and RAD21 with N-MYC during S phase, blocking N-MYC-dependent release of Pol II from the promoter. Inhibition of Aurora-A in S phase restores RAD21 and TFIIIC binding to chromatin and partially restores N-MYC-dependent transcriptional elongation. We propose that complex formation with Aurora-A controls N-MYC function during the cell cycle.


Subject(s)
Aurora Kinase A/metabolism , N-Myc Proto-Oncogene Protein/metabolism , RNA Polymerase II/metabolism , S Phase , Cell Cycle Proteins , Cell Line, Tumor , DNA Topoisomerases, Type II/metabolism , DNA, Intergenic/metabolism , DNA-Binding Proteins , Humans , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Promoter Regions, Genetic , Protein Binding , RNA Polymerase II/genetics , Transcription Elongation, Genetic , Transcription Factors, TFIII/metabolism
10.
Cancer Discov ; 5(7): 768-781, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25934076

ABSTRACT

UNLABELLED: Deregulated expression of MYC is a driver of colorectal carcinogenesis, suggesting that inhibiting MYC may have significant therapeutic value. The PI3K and mTOR pathways control MYC turnover and translation, respectively, providing a rationale to target both pathways to inhibit MYC. Surprisingly, inhibition of PI3K does not promote MYC turnover in colon carcinoma cells, but enhances MYC expression because it promotes FOXO-dependent expression of growth factor receptors and MAPK-dependent transcription of MYC. Inhibition of mTOR fails to inhibit translation of MYC, because levels of 4EBPs are insufficient to fully sequester eIF4E and because an internal ribosomal entry site element in the 5'-untranslated region of the MYC mRNA permits translation independent of eIF4E. A small-molecule inhibitor of the translation factor eIF4A, silvestrol, bypasses the signaling feedbacks, reduces MYC translation, and inhibits tumor growth in a mouse model of colorectal tumorigenesis. We propose that targeting translation initiation is a promising strategy to limit MYC expression in colorectal tumors. SIGNIFICANCE: Inhibiting MYC function is likely to have a significant therapeutic impact in colorectal cancers. Here, we explore several strategies to target translation initiation in order to block MYC expression. We show that a small-molecule inhibitor of eIF4A inhibits MYC expression and suppresses tumor growth in vivo.


Subject(s)
Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Peptide Chain Initiation, Translational/drug effects , Proto-Oncogene Proteins c-myc/genetics , Triterpenes/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , HCT116 Cells , HeLa Cells , Humans , Mice , Signal Transduction/drug effects , Triterpenes/pharmacology , Up-Regulation , Xenograft Model Antitumor Assays
11.
Virology ; 422(2): 242-53, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22099967

ABSTRACT

We demonstrate that HPV-16 E7 forms a complex with Miz-1. UV-induced expression of the CDK-inhibitor p21(Cip1) and subsequent cell cycle arrest depends upon endogenous Miz-1 in HPV-negative C33A cervical cancer cells containing mutated p53. Transient expression of E7 in C33A inhibits UV-induced expression of p21(Cip1) and overcomes Miz-1-induced G1-phase arrest. The C-terminal E7Δ79LEDLL83-mutant with reduced Miz-1-binding capacity was impaired in its capability to repress p21(Cip1) expression; whereas the pRB-binding-deficient E7C24G-mutant inhibited p21(Cip1) expression similar to wild-type E7. Using ChIP, we demonstrate that endogenous E7 is bound to the endogenous p21(Cip1) core-promoter in CaSki cells and RNAi-mediated knock down of Miz-1 abrogates E7-binding to the p21(Cip1) promoter. Co-expression of E7 with Miz-1 inhibited Miz-1-induced p21(Cip1) expression from the minimal-promoter via Miz-1 DNA-binding sites. Co-expression of E7Δ79LEDLL83 did not inhibit Miz-1-induced p21(Cip1) expression. E7C24G retained E7-wild-type capability to inhibit Miz-1-dependent transactivation. These findings suggest that HPV-16 E7 can repress Miz-1-induced p21(Cip1) gene expression.


Subject(s)
Human papillomavirus 16/metabolism , Kruppel-Like Transcription Factors/metabolism , Papillomavirus E7 Proteins/metabolism , Cell Cycle , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation/physiology , Human papillomavirus 16/genetics , Humans , Kruppel-Like Transcription Factors/genetics , Papillomavirus E7 Proteins/genetics , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Two-Hybrid System Techniques
12.
Cancer Res ; 71(2): 404-12, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21123453

ABSTRACT

Neuroblastoma is the most common extracranial solid tumor of childhood. One important factor that predicts a favorable prognosis is the robust expression of the TRKA and p75NTR neurotrophin receptor genes. Interestingly, TRKA and p75NTR expression is often attenuated in aggressive MYCN-amplified tumors, suggesting a causal link between elevated MYCN activity and the transcriptional repression of TRKA and p75NTR, but the precise mechanisms involved are unclear. Here, we show that MYCN acts directly to repress TRKA and p75NTR gene transcription. Specifically, we found that MYCN levels were critical for repression and that MYCN targeted proximal/core promoter regions by forming a repression complex with transcription factors SP1 and MIZ1. When bound to the TRKA and p75NTR promoters, MYCN recruited the histone deacetylase HDAC1 to induce a repressed chromatin state. Forced re-expression of endogenous TRKA and p75NTR with exposure to the HDAC inhibitor TSA sensitized neuroblastoma cells to NGF-mediated apoptosis. By directly connecting MYCN to the repression of TRKA and p75NTR, our findings establish a key pathway of clinical pathogenicity and aggressiveness in neuroblastoma.


Subject(s)
Histone Deacetylase 1/metabolism , Kruppel-Like Transcription Factors/genetics , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/genetics , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Receptor, trkA/genetics , Receptors, Nerve Growth Factor/genetics , Sp1 Transcription Factor/genetics , HEK293 Cells , HeLa Cells , Humans , Kruppel-Like Transcription Factors/metabolism , N-Myc Proto-Oncogene Protein , Nerve Tissue Proteins/biosynthesis , Neuroblastoma/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , Receptor, trkA/biosynthesis , Receptors, Nerve Growth Factor/biosynthesis , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Transfection
13.
J Cell Biol ; 188(6): 905-18, 2010 Mar 22.
Article in English | MEDLINE | ID: mdl-20308430

ABSTRACT

Oncogenic stress induces expression of the alternate reading frame (Arf) tumor suppressor protein. Arf then stabilizes p53, which leads to cell cycle arrest or apoptosis. The mechanisms that distinguish both outcomes are incompletely understood. In this study, we show that Arf interacts with the Myc-associated zinc finger protein Miz1. Binding of Arf disrupts the interaction of Miz1 with its coactivator, nucleophosmin, induces the sumoylation of Miz1, and facilitates the assembly of a heterochromatic complex that contains Myc and trimethylated H3K9 in addition to Miz1. Arf-dependent assembly of this complex leads to the repression of multiple genes involved in cell adhesion and signal transduction and induces apoptosis. Our data point to a tumor-suppressive pathway that weakens cell-cell and cell-matrix interactions in response to expression of Arf and that may thereby facilitate the elimination of cells harboring an oncogenic mutation.


Subject(s)
Apoptosis , Kruppel-Like Transcription Factors/metabolism , Tumor Suppressor Protein p14ARF/metabolism , Cell Adhesion , Cells, Cultured , Humans
14.
Nat Rev Cancer ; 9(6): 441-4, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19461668

ABSTRACT

Deregulated expression of MYC contributes to the genesis of multiple human tumours. The encoded protein, MYC, functions through the transcriptional regulation of large numbers of target genes. Recent publications show that MYC is closely involved in DNA replication and the checkpoint processes that monitor progress through the S phase, and suggest that limiting replication stress is a key function of this protein. These findings could have considerable implications for our understanding of how MYC transforms cells and which mechanisms protect normal cells from transformation by activated oncogenes.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA Replication/physiology , Proto-Oncogene Proteins c-myc/physiology , Animals , Cell Cycle/physiology , Humans , Neoplasms/genetics
15.
Cell Cycle ; 6(19): 2327-31, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17873522

ABSTRACT

The cellular levels of the Myc oncoprotein are critical determinants of cell proliferation, cell growth and apoptosis and are tightly regulated by external growth factors. Levels of Myc oncoprotein also decline in response to intracellular stress signals such as DNA damage. We show here that this decline is in part due to proteasomal degradation and that it is mediated by the Fbw7 ubiquitin ligase. We have shown previously that the ubiquitin-specific protease Usp28, binds to the nucleoplasmic isoform of Fbw7, Fbw7alpha, and counteracts its function in mammalian cells. Usp28 dissociates from Fbw7alpha in response to UV irradiation, providing a mechanism how Fbw7-mediated degradation of Myc is enhanced upon DNA damage. Our data extend previous observations that link Myc function to the cellular response to DNA damage.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , F-Box Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis , Cell Cycle , Cell Line , DNA Damage/radiation effects , F-Box-WD Repeat-Containing Protein 7 , Humans , Ultraviolet Rays
16.
Proc Natl Acad Sci U S A ; 103(11): 4062-7, 2006 Mar 14.
Article in English | MEDLINE | ID: mdl-16537485

ABSTRACT

We used the Sleeping Beauty (SB) transposable element as a tool to probe transposon-host cell interactions in vertebrates. The Miz-1 transcription factor was identified as an interactor of the SB transposase in a yeast two-hybrid screen. Through its association with Miz-1, the SB transposase down-regulates cyclin D1 expression in human cells, as evidenced by differential gene expression analysis using microarray hybridization. Down-regulation of cyclin D1 results in a prolonged G(1) phase of the cell cycle and retarded growth of transposase-expressing cells. G(1) slowdown is associated with a decrease of cyclin D1/cdk4-specific phosphorylation of the retinoblastoma protein. Both cyclin D1 down-regulation and the G(1) slowdown induced by the transposase require Miz-1. A temporary G(1) arrest enhances transposition, suggesting that SB transposition is favored in the G(1) phase of the cell cycle, where the nonhomologous end-joining pathway of DNA repair is preferentially active. Because nonhomologous end-joining is required for efficient SB transposition, the transposase-induced G(1) slowdown is probably a selfish act on the transposon's part to maximize the chance for a successful transposition event.


Subject(s)
Cell Cycle/physiology , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Transposases/metabolism , Animals , CHO Cells , Cricetinae , Cyclin D1/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Down-Regulation , G1 Phase/physiology , HeLa Cells , Humans , In Vitro Techniques , Kruppel-Like Transcription Factors , Oligonucleotide Array Sequence Analysis , Phosphorylation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transposases/chemistry , Transposases/genetics , Two-Hybrid System Techniques , Zinc Fingers
17.
Mol Cell ; 10(3): 509-21, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12408820

ABSTRACT

The Myc oncoprotein represses initiator-dependent transcription through the POZ domain transcription factor Miz-1. We now show that transactivation by Miz-1 is negatively regulated by association with topoisomerase II binding protein (TopBP1); UV irradiation downregulates expression of TopBP1 and releases Miz-1. Miz-1 binds to the p21Cip1 core promoter in vivo and is required for upregulation of p21Cip1 upon UV irradiation. Using both c-myc(-/-) cells and a point mutant of Myc that is deficient in Miz-1 dependent repression, we show that Myc negatively regulates transcription of p21Cip1 upon UV irradiation and facilitates recovery from UV-induced cell cycle arrest through binding to Miz-1. Our data implicate Miz-1 in a pathway that regulates cell proliferation in response to UV irradiation.


Subject(s)
DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcription, Genetic/radiation effects , Transcriptional Activation , Tumor Suppressor Proteins , Ultraviolet Rays , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Division/physiology , Cell Division/radiation effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , Genes, Reporter , Humans , Keratinocytes/cytology , Keratinocytes/physiology , Keratinocytes/radiation effects , Kruppel-Like Transcription Factors , Models, Molecular , Molecular Sequence Data , Nuclear Proteins , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myc/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Transcription Factors , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL