Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
3.
Nat Commun ; 4: 2545, 2013.
Article in English | MEDLINE | ID: mdl-24089055

ABSTRACT

The mammalian circadian clock regulates the daily cycles of many important physiological processes, but its mechanism is not well understood. Here we provide genetic and biochemical evidence that metastasis-associated protein 1 (MTA1), a widely upregulated gene product in human cancers, is an integral component of the circadian molecular machinery. Knockout of MTA1 in mice disrupts the free-running period of circadian rhythms under constant light and normal entrainment of behaviour to 12-h-light/12-h-dark cycles. The CLOCK-BMAL1 heterodimer activates MTA1 transcription through a conserved E-box element at its promoter. MTA1, in turn, interacts with and recruits CLOCK-BMAL1 at its own and CRY1 promoters and promotes their transcription. Moreover, MTA1 deacetylates BMAL1 at lysine 538 through regulating deacetylase SIRT1 expression, thus disturbing the CRY1-mediated negative feedback loop. These findings uncover a previously unappreciated role for MTA1 in maintenance of circadian rhythmicity through acting on the positive limb of the clock machinery.


Subject(s)
Circadian Rhythm/genetics , Gene Expression Regulation , Histone Deacetylases/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Acetylation , Animals , Behavior, Animal , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cryptochromes/genetics , Cryptochromes/metabolism , Feedback, Physiological , Female , Histone Deacetylases/metabolism , Humans , Male , Mice , Mice, Knockout , Motor Activity/genetics , Photoperiod , Promoter Regions, Genetic , Protein Multimerization , Repressor Proteins/metabolism , Signal Transduction , Sirtuin 1/genetics , Sirtuin 1/metabolism , Trans-Activators , Transcription Factors/deficiency
4.
Sci Rep ; 3: 2256, 2013.
Article in English | MEDLINE | ID: mdl-23884293

ABSTRACT

Using RNA sequencing of triple-negative breast cancer (TNBC), non-TBNC and HER2-positive breast cancer sub-types, here we report novel expressed variants, allelic prevalence and abundance, and coexpression with other variation, and splicing signatures. To reveal the most prevalent variant alleles, we overlaid our findings with cancer- and population-based datasets and validated a subset of novel variants of cancer-related genes: ESRP2, GBP1, TPP1, MAD2L1BP, GLUD2 and SLC30A8. As a proof-of-principle, we demonstrated that a rare substitution in the splicing coordinator ESRP2 (R353Q) impairs its ability to bind to its substrate FGFR2 pre-mRNA. In addition, we describe novel SNPs and INDELs in cancer relevant genes with no prior reported association of point mutations with cancer, such as MTAP and MAGED1. For the first time, this study illustrates the power of RNA-sequencing in revealing the variation landscape of breast transcriptome and exemplifies analytical strategies to search regulatory interactions among cancer relevant molecules.


Subject(s)
Breast Neoplasms/genetics , Genetic Variation , RNA Precursors/genetics , Alleles , Computational Biology , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Gene Frequency , Genome-Wide Association Study , Genomics , Genotype , High-Throughput Nucleotide Sequencing , Humans , Mutation , Polymorphism, Single Nucleotide , RNA Splicing , Sequence Analysis, RNA , Transcriptome , Tripeptidyl-Peptidase 1
5.
Cancer Res ; 73(12): 3761-70, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23580571

ABSTRACT

Overexpression of the prometastatic chromatin modifier protein metastasis tumor antigen 1 (MTA1) in human cancer contributes to tumor aggressiveness, but the role of endogenous MTA1 in cancer has not been explored. Here, we report the effects of selective genetic depletion of MTA1 in a physiologically relevant spontaneous mouse model of breast cancer pulmonary metastasis. We found that MTA1 acts as a mandatory modifier of breast-to-lung metastasis without effects on primary tumor formation. The underlying mechanism involved MTA1-dependent stimulation of STAT3 transcription through action on the MTA1/STAT3/Pol II coactivator complex, and, in turn, on the expression and functions of STAT3 target genes including Twist1. Accordingly, we documented a positive correlation between levels of MTA1 and STAT3 in publicly available breast cancer data sets. Together, our findings reveal an essential modifying role of the physiologic level of MTA1 in supporting pulmonary metastasis of breast cancer.


Subject(s)
Lung Neoplasms/genetics , Mammary Neoplasms, Animal/genetics , STAT3 Transcription Factor/genetics , Transcription Factors/genetics , Animals , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement/genetics , Cells, Cultured , DNA Polymerase II/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mice , Mice, 129 Strain , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , RNA Interference , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Trans-Activators , Transcription Factors/metabolism , Transcription, Genetic , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
6.
Cell Rep ; 2(6): 1657-69, 2012 Dec 27.
Article in English | MEDLINE | ID: mdl-23260667

ABSTRACT

Chromatin dynamics play a central role in maintaining genome integrity, but how this is achieved remains largely unknown. Here, we report that microrchidia CW-type zinc finger 2 (MORC2), an uncharacterized protein with a derived PHD finger domain and a conserved GHKL-type ATPase module, is a physiological substrate of p21-activated kinase 1 (PAK1), an important integrator of extracellular signals and nuclear processes. Following DNA damage, MORC2 is phosphorylated on serine 739 in a PAK1-dependent manner, and phosphorylated MORC2 regulates its DNA-dependent ATPase activity to facilitate chromatin remodeling. Moreover, MORC2 associates with chromatin and promotes gamma-H2AX induction in a PAK1 phosphorylation-dependent manner. Consequently, cells expressing MORC2-S739A mutation displayed a reduction in DNA repair efficiency and were hypersensitive to DNA-damaging agent. These findings suggest that the PAK1-MORC2 axis is critical for orchestrating the interplay between chromatin dynamics and the maintenance of genomic integrity through sequentially integrating multiple essential enzymatic processes.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromatin Assembly and Disassembly , DNA Damage , Transcription Factors/metabolism , Adenosine Triphosphatases/genetics , Amino Acid Substitution , DNA Repair/genetics , HeLa Cells , Humans , Mutation, Missense , Phosphorylation/genetics , Protein Structure, Tertiary , Transcription Factors/genetics , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
7.
J Biol Chem ; 287(33): 27843-50, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22700976

ABSTRACT

Metastasis-associated protein 1 (MTA1) is widely overexpressed in human cancers and is associated with malignant phenotypic changes contributing to morbidity in the associated diseases. Here we discovered for the first time that MTA1, a master chromatin modifier, transcriptionally represses the expression of phosphatase and tensin homolog (PTEN), a tumor suppressor gene, by recruiting class II histone deacetylase 4 (HDAC4) along with the transcription factor Yin-Yang 1 (YY1) onto the PTEN promoter. We also found evidence of an inverse correlation between the expression levels of MTA1 and PTEN in physiologically relevant breast cancer microarray datasets. We found that MTA1 up-regulation leads to a decreased expression of PTEN protein and stimulation of PI3K as well as phosphorylation of its signaling targets. Accordingly, selective down-regulation of MTA1 in breast cancer cells increases PTEN expression and inhibits stimulation of the PI3K/AKT signaling. Collectively, these findings provide a mechanistic role for MTA1 in transcriptional repression of PTEN, leading to modulation of the resulting signaling pathways.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Histone Deacetylases/metabolism , Multienzyme Complexes/metabolism , PTEN Phosphohydrolase/biosynthesis , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , HeLa Cells , Histone Deacetylases/genetics , Humans , Mice , Multienzyme Complexes/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic/physiology , Repressor Proteins/genetics , Signal Transduction/physiology , Trans-Activators , Transcription Factors/genetics , Transcription, Genetic/physiology , Up-Regulation/physiology , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
8.
Sci Rep ; 2: 264, 2012.
Article in English | MEDLINE | ID: mdl-22355776

ABSTRACT

Breast cancer is a heterogeneous disease with a poorly defined genetic landscape, which poses a major challenge in diagnosis and treatment. By massively parallel mRNA sequencing, we obtained 1.2 billion reads from 17 individual human tissues belonging to TNBC, Non-TNBC, and HER2-positive breast cancers and defined their comprehensive digital transcriptome for the first time. Surprisingly, we identified a high number of novel and unannotated transcripts, revealing the global breast cancer transcriptomic adaptations. Comparative transcriptomic analyses elucidated differentially expressed transcripts between the three breast cancer groups, identifying several new modulators of breast cancer. Our study also identified common transcriptional regulatory elements, such as highly abundant primary transcripts, including osteonectin, RACK1, calnexin, calreticulin, FTL, and B2M, and "genomic hotspots" enriched in primary transcripts between the three groups. Thus, our study opens previously unexplored niches that could enable a better understanding of the disease and the development of potential intervention strategies.


Subject(s)
Breast Neoplasms/genetics , RNA, Messenger/genetics , Transcriptome , Breast Neoplasms/pathology , Female , Gene Expression Profiling , Humans , Polymerase Chain Reaction , Regulatory Sequences, Nucleic Acid
9.
J Biol Chem ; 287(8): 5615-26, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22184113

ABSTRACT

Metastasis-associated protein 1 (MTA1), a component of the nucleosome-remodeling and histone deacetylase complex, is widely up-regulated in human cancers and significantly correlated with tumor invasion and metastasis, but the mechanisms involved remain largely unknown. Here, we report that MTA1 transcriptionally represses the expression of RING finger protein 144A (RNF144A), an uncharacterized gene whose protein product possesses potential E3 ubiquitin ligase activity, by recruiting the histone deacetylase 2 (HDAC2) and CCAAT/enhancer-binding protein α (c/EBPα) co-repressor complex onto human RNF144A promoter. Furthermore, an inverse correlation between the expression levels of MTA1 and RNF144A was demonstrated in publicly available breast cancer microarray datasets and the MCF10 breast cancer progression model system. To address functional aspects of MTA1 regulation of RNF144A, we demonstrate that RNF144A is a novel suppressor of cancer migration and invasion, two requisite steps of metastasis in vivo, and knockdown of endogenous RNF144A by small interfering RNAs accelerates the migration and invasion of MTA1-overexpressing cells. These results suggest that RNF144A is partially responsible for MTA1-mediated migration and invasion and that MTA1 overexpression in highly metastatic cancer cells drives cell migration and invasion by, at least in part, interfering with the suppressive function of RNF144A through transcriptional repression of RNF144A expression. Together, these findings provide novel mechanistic insights into regulation of tumor progression and metastasis by MTA1 and highlight a previously unrecognized role of RNF144A in MTA1-driven cancer cell migration and invasion.


Subject(s)
Cell Movement/genetics , Gene Silencing , Histone Deacetylases/metabolism , Repressor Proteins/metabolism , Transcription, Genetic/genetics , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Animals , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Carrier Proteins , Cell Line, Tumor , Computational Biology , HeLa Cells , Histone Deacetylase 2/metabolism , Histone Deacetylases/genetics , Humans , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Trans-Activators
10.
Proc Natl Acad Sci U S A ; 108(21): 8791-6, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555589

ABSTRACT

Although metastasis-associated protein 1 (MTA1), a component of the nucleosome remodeling and histone deacetylation complex, is widely up-regulated in human cancers and correlates with tumor metastasis, its regulatory mechanism and related signaling pathways remain unknown. Here, we report a previously unrecognized bidirectional autoregulatory loop between MTA1 and tumor suppressor alternative reading frame (ARF). MTA1 transactivates ARF transcription by recruiting the transcription factor c-Jun onto the ARF promoter in a p53-independent manner. ARF, in turn, negatively regulates MTA1 expression independently of p53 and c-Myc. In this context, ARF interacts with transcription factor specificity protein 1 (SP1) and promotes its proteasomal degradation by enhancing its interaction with proteasome subunit regulatory particle ATPase 6, thereby abrogating the ability of SP1 to stimulate MTA1 transcription. ARF also physically associates with MTA1 and affects its protein stability. Thus, MTA1-mediated activation of ARF and ARF-mediated functional inhibition of MTA1 represent a p53-independent bidirectional autoregulatory mechanism in which these two opposites act in concert to regulate cell homeostasis and oncogenesis, depending on the cellular context and the environment.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Histone Deacetylases/genetics , Homeostasis/genetics , Neoplasms/etiology , Repressor Proteins/genetics , Cell Line , Cyclin-Dependent Kinase Inhibitor p16/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Expression Regulation , Histone Deacetylases/metabolism , Humans , Reading Frames , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Trans-Activators , Transcriptional Activation , Tumor Suppressor Protein p53
11.
Proc Natl Acad Sci U S A ; 108(10): 4200-5, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21368136

ABSTRACT

Despite ubiquitous expression and a high level of metastasis-associated protein 1 (MTA1) coregulator, the physiological role of the MTA1 coactivator remains unknown. We found that MTA1 is a bona fide coactivator and stimulator of tyrosine hydroxylase (TH) transcription in neuronal cells and that MTA1-null mice had lower TH expression in the striatum and substantial nigra. MTA1 physically achieves these functions by interacting directly with DJ1 (Parkinson disease 7) and in turn recruits the DJ1/MTA1/RNA polymerase II complex to the bicoid binding element (BBE) in the TH promoter. Furthermore, we found that the MTA1/DJ1 complex is required for optimum stimulation of the TH expression by paired like homeodomain transcription factor (Pitx3) homeodomain transcription factor and that the MTA1/DJ1 complex is recruited to the TH gene chromatin via the direct interaction of MTA1 with Pitx3. These findings reveal a role for MTA1 as an upstream coactivator of TH and advance the notion of polygenic regulation of a disease-causing gene by coordinated interactions of three regulatory proteins.


Subject(s)
Transcription, Genetic/genetics , Tyrosine 3-Monooxygenase/genetics , Animals , Corpus Striatum/enzymology , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Mice , Mice, Knockout , Repressor Proteins , Substantia Nigra/enzymology , Trans-Activators , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology
12.
J Biol Chem ; 286(9): 7132-8, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21156794

ABSTRACT

Although both metastatic tumor antigen 1 (MTA1), a master chromatin modifier, and transglutaminase 2 (TG2), a multifunctional enzyme, are known to be activated during inflammation, it remains unknown whether these molecules regulate inflammatory response in a coordinated manner. Here we investigated the role of MTA1 in the regulation of TG2 expression in bacterial lipopolysaccharide (LPS)-stimulated mammalian cells. While studying the impact of MTA1 status on global gene expression, we unexpectedly discovered that MTA1 depletion impairs the basal as well as the LPS-induced expression of TG2 in multiple experimental systems. We found that TG2 is a chromatin target of MTA1 and of NF-κB signaling in LPS-stimulated cells. In addition, LPS-mediated stimulation of TG2 expression is accompanied by the enhanced recruitment of MTA1, p65RelA, and RNA polymerase II to the NF-κB consensus sites in the TG2 promoter. Interestingly, both the recruitment of p65 and TG2 expression are effectively blocked by a pharmacological inhibitor of the NF-κB pathway. These findings reveal an obligatory coregulatory role of MTA1 in the regulation of TG2 expression and of the MTA1-TG2 pathway, at least in part, in LPS modulation of the NF-κB signaling in stimulated macrophages.


Subject(s)
GTP-Binding Proteins/metabolism , Inflammation/physiopathology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Transcription Factors/metabolism , Transglutaminases/metabolism , Animals , Breast Neoplasms , Cell Line, Tumor , Chromatin Immunoprecipitation , Female , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , GTP-Binding Proteins/genetics , Gene Expression Regulation, Enzymologic/immunology , Histone Deacetylases/metabolism , Humans , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/cytology , Mice , NF-kappa B/metabolism , Protein Glutamine gamma Glutamyltransferase 2 , Repressor Proteins/metabolism , Trans-Activators , Transglutaminases/genetics
13.
J Biol Chem ; 285(43): 32787-32792, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20702415

ABSTRACT

Although metastasis tumor antigen 1 (MTA1) contributes to the responsiveness of macrophages to LPS, the underlying mechanism remains unknown. Here, we investigated the role of MTA1 in the regulation of expression and function of MyD88, a proximal component of NF-κB signaling. We discovered that MTA1 targets MyD88 and that MyD88 is a NF-κB-responsive gene in LPS-stimulated macrophages. We found that MTA1 is required for MyD88-dependent stimulation of NF-κB signaling and expression of proinflammatory cytokines such as IL-1ß, MIP2, and TNF-α as MTA1 depletion leads to a substantial reduction in the expression of NF-κB target genes. In addition, LPS-mediated stimulation of MyD88 transcription was accompanied by an enhanced recruitment of MTA1, RNA polymerase II, and p65RelA complex to the NF-κB consensus sites in the MyD88 promoter. Interestingly, the recruitment of both MTA1 and MyD88 expression is effectively blocked by NF-κB inhibitor parthenolide. Selective knockdown of MyD88 by a dominant negative mutant of MyD88 or selective siRNA also impairs the ability of LPS to stimulate the NF-κB target genes. These findings reveal an inherent coregulatory role of MTA1 upon the expression of MyD88 and suggest that MTA1 regulation of MyD88 may constitute at least one of the mechanisms by which MTA1 stimulates LPS-induced NF-κB signaling in stimulated macrophages.


Subject(s)
Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Myeloid Differentiation Factor 88/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Line , Cytokines/biosynthesis , Cytokines/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Inflammation Mediators/metabolism , Macrophages, Peritoneal/cytology , Mice , Myeloid Differentiation Factor 88/genetics , Repressor Proteins , Response Elements/physiology , Sesquiterpenes/pharmacology , Signal Transduction/genetics , Trans-Activators , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transcription Factors/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
14.
J Biol Chem ; 285(31): 23590-7, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20519513

ABSTRACT

The MTA1 coregulator (metastatic tumor antigen 1), a component of the nucleosome remodeling and deacetylase (NuRD) complex, has been intimately linked with human cancer, but its role in inflammatory responses remains unknown. Here, we discovered that MTA1 is a target of inflammation, and stimulation of macrophages with Escherichia coli lipopolysaccharide (LPS) stimulates MTA1 transcription via the NF-kappaB pathway. Unexpectedly, we found that MTA1 depletion in LPS-stimulated macrophages impairs NF-kappaB signaling and expression of inflammatory molecules. MTA1 itself acts as a transcriptional coactivator of inflammatory cytokines in LPS-stimulated macrophages, and in contrast, it acts as a corepressor in resting primary macrophages as its depletion induced cytokine expression. LPS stimulates S-nitrosylation of histone deacetylase 2 (HDAC2) and interferes with its binding to MTA1, which, in turn, resulted in the loss of corepressor behavior of MTA1.HDAC complex in activated macrophages. Consequently, the net levels of inflammatory cytokines in LPS-stimulated macrophages from MTA1(-/-) mice were high compared with wild-type mice. Accordingly, MTA1(-/-) mice were much more susceptible than control mice to septic shock induced by LPS, revealing that MTA1 protects mice from deregulated host inflammatory response. These findings reveal a previously unrecognized, critical homeostatic role of MTA1, both as a target and as a component of the NF-kappaB circuitry, in the regulation of inflammatory responses.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation , NF-kappa B/metabolism , Nucleosomes/metabolism , Animals , Cloning, Molecular , Escherichia coli/metabolism , Histone Deacetylase 2/metabolism , Homeostasis , Inflammation , Lipopolysaccharides/metabolism , Mice , Mice, Transgenic , RNA, Small Interfering/metabolism , Repressor Proteins , Trans-Activators , Transcription Factors/metabolism
15.
Cancer Biol Ther ; 9(7): 479-82, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20190563

ABSTRACT

MicroRNAs are small, non-coding RNAs that regulate gene expression by degrading and/or suppressing the translation of target mRNA by Watson-Crick base pairing in the 3-'UTR of mRNA. The recent explosion of information about the biochemistry and action of microRNAs has implicated these regulatory molecules in many unexpected biologic processes, ranging from development and homeostasis to diseases such as cancer. In general, microRNAs are down regulated or deleted in cancer while a few are upregulated. However, some microRNAs suppress oncogenesis or metastasis, while others are involved in promoting tumorigenesis. All these developments make microRNAs attractive diagnostic markers as well as therapeutic targets. Here we will briefly review the opportunities and potential limitations of using microRNAs in cancer therapeutics.


Subject(s)
Genetic Therapy , MicroRNAs/genetics , Neoplasms/therapy , Animals , Gene Targeting , Humans , MicroRNAs/antagonists & inhibitors , Neoplasms/genetics
16.
J Biol Chem ; 285(13): 10044-10052, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20071335

ABSTRACT

Although metastasis-associated protein 1 (MTA1), a component of the nucleosome remodeling and deacetylase (NuRD) complex, is a DNA-damage response protein and regulates p53-dependent DNA repair, it remains unknown whether MTA1 also participates in p53-independent DNA damage response. Here, we provide evidence that MTA1 is a p53-independent transcriptional corepressor of p21(WAF1), and the underlying mechanism involves recruitment of MTA1-histone deacetylase 2 (HDAC2) complexes onto two selective regions of the p21(WAF1) promoter. Accordingly, MTA1 depletion, despite its effect on p53 down-regulation, superinduces p21(WAF1), increases p21(WAF1) binding to proliferating cell nuclear antigen (PCNA), and decreases the nuclear accumulation of PCNA in response to ionizing radiation. In support of a p53-independent role of MTA1 in DNA damage response, we further demonstrate that induced expression of MTA1 in p53-null cells inhibits p21(WAF1) promoter activity and p21(WAF1) binding to PCNA. Consequently, MTA1 expression in p53-null cells results in increased induction of gamma H2AX foci and DNA double strand break repair, and decreased DNA damage sensitivity following ionizing radiation treatment. These findings uncover a new target of MTA1 and the existence of an additional p53-independent role of MTA1 in DNA damage response, at least in part, by modulating the p21(WAF1)-PCNA pathway, and thus, linking two previously unconnected NuRD complex and DNA-damage response pathways.


Subject(s)
Antigens, Nuclear/metabolism , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Histone Deacetylases/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Trans-Activators
17.
Proc Natl Acad Sci U S A ; 106(41): 17493-8, 2009 Oct 13.
Article in English | MEDLINE | ID: mdl-19805145

ABSTRACT

Metastasis-associated protein 1 (MTA1), a component of the nucleosome remodeling and histone deacetylation (NuRD) complex, is widely upregulated in human cancers. However, the mechanism for regulating its protein stability remains unknown. Here we report that MTA1 is an ubiquitinated protein and targeted by the RING-finger E3 ubiquitin-protein ligase constitutive photomorphogenesis protein 1 (COP1) for degradation via the ubiquitin-proteasome pathway. Induced expression of wild-type COP1 but not its RING motif mutants promotes the ubiquitination and degradation of MTA1, indicating that the ligase activity is required for the COP1-mediated proteolysis of MTA1. Conversely, depletion of endogenous COP1 resulted in a marked decrease in MTA1 ubiquitination, accompanied by a pronounced accumulation of MTA1 protein. MTA1, in turn, destabilizes COP1 by promoting its autoubiquitination, thus creating a tight feedback loop that regulates both MTA1 and COP1 protein stability. Accordingly, disruption of the COP1-mediated proteolysis by ionizing radiation leads to MTA1 stabilization, accompanied by an increased coregulatory function of MTA1 on its target. Furthermore, we discovered that MTA1 is required for optimum DNA double-strand break repair after ionizing radiation. These findings provide novel insights into the regulation of MTA1 protein and reveal a novel function of MTA1 in DNA damage response.


Subject(s)
Histone Deacetylases/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line , DNA Damage , DNA Repair , Enzyme Stability , Fibroblasts/cytology , Fibroblasts/physiology , Histone Deacetylases/chemistry , Histone Deacetylases/radiation effects , Humans , Mice , Nuclear Proteins/genetics , Radiation, Ionizing , Repressor Proteins/chemistry , Repressor Proteins/radiation effects , Trans-Activators , Transcription Factors/deficiency , Transcription Factors/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics
18.
Cancer Res ; 69(14): 5639-42, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19584269

ABSTRACT

MicroRNAs (miR) have been identified as posttranscriptional modifiers of target gene regulation and control the expression of gene products important in cancer progression. Here, we show that miR-661 inhibits the expression of metastatic tumor antigen 1 (MTA1), a widely up-regulated gene product in human cancer, by targeting the 3' untranslated region (UTR) of MTA1 mRNA. We found that endogenous miR-661 expression was positively regulated by the c/EBPalpha transcription factor, which is down-regulated during cancer progression. c/EBPalpha directly interacted with the miR-661 chromatin and bound to miR-661 putative promoter that contains a c/EBPalpha-consensus motif. In addition, we found that the level of MTA1 protein was progressively up-regulated, whereas that of miR-661 and its activator, c/EBPalpha, were down-regulated in a breast cancer progression model consisting of MCF-10A cell lines whose phenotypes ranged from noninvasive to highly invasive. c/EBPalpha expression in breast cancer cells resulted in increased miR-661 expression and reduced MTA1 3'UTR-luciferase activity and MTA1 protein level. We also provide evidence that the introduction of miR-661 inhibited the motility, invasiveness, anchorage-independent growth, and tumorigenicity of invasive breast cancer cells. We believe our findings show for the first time that c/EBPalpha regulates the level of miR-661 and in turn modifies the functions of the miR661-MTA1 pathway in human cancer cells. Based on these findings, we suggest that miR-661 be further investigated for therapeutic use in down-regulating the expression of MTA1 in cancer cells.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/metabolism , Histone Deacetylases/genetics , MicroRNAs/genetics , Repressor Proteins/genetics , 3' Untranslated Regions/genetics , Animals , Blotting, Western , Cell Line, Tumor , Cell Movement , Chromatin Immunoprecipitation , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Histone Deacetylases/metabolism , Humans , Mice , Mice, Nude , MicroRNAs/metabolism , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators
19.
Cancer Res ; 68(20): 8195-200, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18922890

ABSTRACT

MicroRNAs are noncoding RNAs that inhibit the expression of their targets in a sequence-specific manner and play crucial roles during oncogenesis. Here we show that microRNA-7 (miR-7) inhibits p21-activated kinase 1 (Pak1) expression, a widely up-regulated signaling kinase in multiple human cancers, by targeting the 3'-untranslated region (UTR) of Pak1 mRNA. We noticed an inverse correlation between the levels of endogenous miR-7 and Pak1 expression in human cancer cells. We discovered that endogenous miR-7 expression is positively regulated by a homeodomain transcription factor, HoxD10, the loss of which leads to an increased invasiveness. HoxD10 directly interacts with the miR-7 chromatin. Accordingly, the levels of Pak1 protein are progressively up-regulated whereas those of miR-7 and its upstream activator HoxD10 are progressively down-regulated in a cellular model of breast cancer progression from low to highly invasive phenotypes. Furthermore, HoxD10 expression in highly invasive breast cancer cells resulted in an increased miR-7 expression but reduced Pak1 3'-UTR-luciferase activity and reduced Pak1 protein. Finally, we show that miR-7 introduction inhibits the motility, invasiveness, anchorage-independent growth, and tumorigenic potential of highly invasive breast cancer cells. Collectively, these findings establish for the first time that Pak1 is a target of miR-7 and that HoxD10 plays a regulatory role in modifying the expression of miR-7 and, consequently, the functions of the miR-7-Pak1 pathway in human cancer cells.


Subject(s)
Homeodomain Proteins/physiology , MicroRNAs/physiology , Neoplasm Invasiveness , Transcription Factors/physiology , p21-Activated Kinases/antagonists & inhibitors , 3' Untranslated Regions/physiology , Adaptor Proteins, Signal Transducing/analysis , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , ErbB Receptors/analysis , Female , Gene Expression Regulation , Humans , Insulin Receptor Substrate Proteins , MicroRNAs/genetics , Promoter Regions, Genetic , p21-Activated Kinases/analysis , p21-Activated Kinases/genetics , p21-Activated Kinases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...