Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cancer Med ; 13(2): e6949, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38334474

ABSTRACT

BACKGROUND: Acute myeloid leukemia (AML) with an internal tandem duplication in the fms-like tyrosine kinase receptor 3 gene (FLT3-ITD) is associated with poor survival, and few studies have examined the impact of modifiable behaviors, such as nutrient quality and timing, in this subset of acute leukemia. METHODS: The influence of diet composition (low-sucrose and/or low-fat diets) and timing of diet were tested in tandem with anthracycline treatment in orthotopic xenograft mouse models. A pilot clinical study to test receptivity of pediatric leukemia patients to macronutrient matched foods was conducted. A role for the circadian protein, BMAL1 (brain and muscle ARNT-like 1), in effects of diet timing was studied by overexpression in FLT3-ITD-bearing AML cells. RESULTS: Reduced tumor burden in FLT3-ITD AML-bearing mice was observed with interventions utilizing low-sucrose and/or low-fat diets, or time-restricted feeding (TRF) compared to mice fed normal chow ad libitum. In a tasting study, macronutrient matched low-sucrose and low-fat meals were offered to pediatric acute leukemia patients who largely reported liking the meals. Expression of the circadian protein, BMAL1, was heightened with TRF and the low-sucrose diet. BMAL1 overexpression and treatment with a pharmacological inducer of BMAL1 was cytotoxic to FLT3-ITD AML cells. CONCLUSIONS: Mouse models for FLT3-ITD AML show that diet composition and timing slows progression of FLT3-ITD AML growth in vivo, potentially mediated by BMAL1. These interventions to enhance therapy efficacy show preliminary feasibility, as pediatric leukemia patients responded favorable to preparation of macronutrient matched meals.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Humans , Child , Mice , Animals , ARNTL Transcription Factors/genetics , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/drug therapy , Antineoplastic Agents/therapeutic use , Disease Models, Animal , Diet , Sucrose/therapeutic use , fms-Like Tyrosine Kinase 3/genetics , Mutation
2.
iScience ; 26(7): 107126, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37426340

ABSTRACT

TRIM24 is an oncogenic chromatin reader that is frequently overexpressed in human tumors and associated with poor prognosis. However, TRIM24 is rarely mutated, duplicated, or rearranged in cancer. This raises questions about how TRIM24 is regulated and what changes in its regulation are responsible for its overexpression. Here, we perform a genome-wide CRISPR-Cas9 screen by fluorescence-activated cell sorting (FACS) that nominated 220 negative regulators and elucidated a regulatory network that includes the KAP1 corepressor, CNOT deadenylase, and GID/CTLH E3 ligase. Knocking out required components of these three complexes caused TRIM24 overexpression, confirming their negative regulation of TRIM24. Our findings identify regulators of TRIM24 that nominate previously unexplored contexts for this oncoprotein in biology and disease. These findings were enabled by SLIDER, a new scoring system designed and vetted in our study as a broadly applicable tool for analysis of CRISPR screens performed by FACS.

3.
Cell Rep ; 37(8): 110044, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34818540

ABSTRACT

ß-hydroxybutyrate (ß-OHB) is an essential metabolic energy source during fasting and functions as a chromatin regulator by lysine ß-hydroxybutyrylation (Kbhb) modification of the core histones H3 and H4. We report that Kbhb on histone H3 (H3K9bhb) is enriched at proximal promoters of critical gene subsets associated with lipolytic and ketogenic metabolic pathways in small intestine (SI) crypts during fasting. Similar Kbhb enrichment is observed in Lgr5+ stem cell-enriched epithelial spheroids treated with ß-OHB in vitro. Combinatorial chromatin state analysis reveals that H3K9bhb is associated with active chromatin states and that fasting enriches for an H3K9bhb-H3K27ac signature at active metabolic gene promoters and distal enhancer elements. Intestinal knockout of Hmgcs2 results in marked loss of H3K9bhb-associated loci, suggesting that local production of ß-OHB is responsible for chromatin reprogramming within the SI crypt. We conclude that modulation of H3K9bhb in SI crypts is a key gene regulatory event in response to fasting.


Subject(s)
3-Hydroxybutyric Acid/metabolism , Fasting/metabolism , Histones/metabolism , Acetylation , Animals , Chromatin/metabolism , Fasting/physiology , Female , Gene Expression/genetics , Gene Expression Regulation/genetics , Intestine, Small/metabolism , Ketone Bodies/metabolism , Lysine/analogs & derivatives , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Regulatory Sequences, Nucleic Acid/genetics
4.
Nat Commun ; 12(1): 5389, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34508101

ABSTRACT

Conditional overexpression of histone reader Tripartite motif containing protein 24 (TRIM24) in mouse mammary epithelia (Trim24COE) drives spontaneous development of mammary carcinosarcoma tumors, lacking ER, PR and HER2. Human carcinosarcomas or metaplastic breast cancers (MpBC) are a rare, chemorefractory subclass of triple-negative breast cancers (TNBC). Comparison of Trim24COE metaplastic carcinosarcoma morphology, TRIM24 protein levels and a derived Trim24COE gene signature reveals strong correlation with human MpBC tumors and MpBC patient-derived xenograft (PDX) models. Global and single-cell tumor profiling reveal Met as a direct oncogenic target of TRIM24, leading to aberrant PI3K/mTOR activation. Here, we find that pharmacological inhibition of these pathways in primary Trim24COE tumor cells and TRIM24-PROTAC treatment of MpBC TNBC PDX tumorspheres decreased cellular viability, suggesting potential in therapeutically targeting TRIM24 and its regulated pathways in TRIM24-expressing TNBC.


Subject(s)
Carcinosarcoma/genetics , Carrier Proteins/genetics , Mammary Neoplasms, Experimental/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Triple Negative Breast Neoplasms/genetics , Animals , Breast/pathology , Carcinosarcoma/pathology , Carrier Proteins/metabolism , Clinical Trials as Topic , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Nuclear Proteins/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-met/genetics , RNA-Seq , Single-Cell Analysis , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/pathology , Whole Genome Sequencing , Xenograft Model Antitumor Assays
5.
Br J Cancer ; 125(2): 176-189, 2021 07.
Article in English | MEDLINE | ID: mdl-33795809

ABSTRACT

BACKGROUND: The mechanism by which immune cells regulate metastasis is unclear. Understanding the role of immune cells in metastasis will guide the development of treatments improving patient survival. METHODS: We used syngeneic orthotopic mouse tumour models (wild-type, NOD/scid and Nude), employed knockout (CD8 and CD4) models and administered CXCL4. Tumours and lungs were analysed for cancer cells by bioluminescence, and circulating tumour cells were isolated from blood. Immunohistochemistry on the mouse tumours was performed to confirm cell type, and on a tissue microarray with 180 TNBCs for human relevance. TCGA data from over 10,000 patients were analysed as well. RESULTS: We reveal that intratumoral immune infiltration differs between metastatic and non-metastatic tumours. The non-metastatic tumours harbour high levels of CD8+ T cells and low levels of platelets, which is reverse in metastatic tumours. During tumour progression, platelets and CXCL4 induce differentiation of monocytes into myeloid-derived suppressor cells (MDSCs), which inhibit CD8+ T-cell function. TCGA pan-cancer data confirmed that CD8lowPlatelethigh patients have a significantly lower survival probability compared to CD8highPlateletlow. CONCLUSIONS: CD8+ T cells inhibit metastasis. When the balance between CD8+ T cells and platelets is disrupted, platelets produce CXCL4, which induces MDSCs thereby inhibiting the CD8+ T-cell function.


Subject(s)
Breast Neoplasms/immunology , CD4 Antigens/genetics , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/transplantation , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Platelet Factor 4/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Female , Gene Knockout Techniques , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, Nude , Myeloid-Derived Suppressor Cells/immunology , Neoplastic Cells, Circulating/immunology , Platelet Factor 4/administration & dosage , Platelet Factor 4/pharmacology , Survival Analysis , Transplantation, Isogeneic , Xenograft Model Antitumor Assays
6.
Cancer Discov ; 11(8): 2094-2111, 2021 08.
Article in English | MEDLINE | ID: mdl-33839689

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is almost uniformly fatal and characterized by early metastasis. Oncogenic KRAS mutations prevail in 95% of PDAC tumors and co-occur with genetic alterations in the TP53 tumor suppressor in nearly 70% of patients. Most TP53 alterations are missense mutations that exhibit gain-of-function phenotypes that include increased invasiveness and metastasis, yet the extent of direct cooperation between KRAS effectors and mutant p53 remains largely undefined. We show that oncogenic KRAS effectors activate CREB1 to allow physical interactions with mutant p53 that hyperactivate multiple prometastatic transcriptional networks. Specifically, mutant p53 and CREB1 upregulate the prometastatic, pioneer transcription factor FOXA1, activating its transcriptional network while promoting WNT/ß-catenin signaling, together driving PDAC metastasis. Pharmacologic CREB1 inhibition dramatically reduced FOXA1 and ß-catenin expression and dampened PDAC metastasis, identifying a new therapeutic strategy to disrupt cooperation between oncogenic KRAS and mutant p53 to mitigate metastasis. SIGNIFICANCE: Oncogenic KRAS and mutant p53 are the most commonly mutated oncogene and tumor suppressor gene in human cancers, yet direct interactions between these genetic drivers remain undefined. We identified a cooperative node between oncogenic KRAS effectors and mutant p53 that can be therapeutically targeted to undermine cooperation and mitigate metastasis.This article is highlighted in the In This Issue feature, p. 1861.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Genes, p53/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Carcinoma, Pancreatic Ductal/pathology , Female , Gene Regulatory Networks , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Metastasis , Pancreatic Neoplasms/pathology
8.
BMC Genomics ; 19(1): 150, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29458327

ABSTRACT

BACKGROUND: Epigenetic regulators are frequently mutated or aberrantly expressed in a variety of cancers, leading to altered transcription states that result in changes in cell identity, behavior, and response to therapy. RESULTS: To define alterations in epigenetic landscapes in breast cancers, we profiled the distributions of 8 key histone modifications by ChIP-Seq, as well as primary (GRO-seq) and steady state (RNA-Seq) transcriptomes, across 13 distinct cell lines that represent 5 molecular subtypes of breast cancer and immortalized human mammary epithelial cells. DISCUSSION: Using combinatorial patterns of distinct histone modification signals, we defined subtype-specific chromatin signatures to nominate potential biomarkers. This approach identified AFAP1-AS1 as a triple negative breast cancer-specific gene associated with cell proliferation and epithelial-mesenchymal-transition. In addition, our chromatin mapping data in basal TNBC cell lines are consistent with gene expression patterns in TCGA that indicate decreased activity of the androgen receptor pathway but increased activity of the vitamin D biosynthesis pathway. CONCLUSIONS: Together, these datasets provide a comprehensive resource for histone modification profiles that define epigenetic landscapes and reveal key chromatin signatures in breast cancer cell line subtypes with potential to identify novel and actionable targets for treatment.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Histones/metabolism , Biomarkers, Tumor , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Female , Gene Expression Profiling , Humans , Transcriptome
9.
Genome Res ; 28(2): 159-170, 2018 02.
Article in English | MEDLINE | ID: mdl-29273624

ABSTRACT

Noncoding transcription is a defining feature of active enhancers, linking transcription factor (TF) binding to the molecular mechanisms controlling gene expression. To determine the relationship between enhancer activity and biological outcomes in breast cancers, we profiled the transcriptomes (using GRO-seq and RNA-seq) and epigenomes (using ChIP-seq) of 11 different human breast cancer cell lines representing five major molecular subtypes of breast cancer, as well as two immortalized ("normal") human breast cell lines. In addition, we developed a robust and unbiased computational pipeline that simultaneously identifies putative subtype-specific enhancers and their cognate TFs by integrating the magnitude of enhancer transcription, TF mRNA expression levels, TF motif P-values, and enrichment of H3K4me1 and H3K27ac. When applied across the 13 different cell lines noted above, the Total Functional Score of Enhancer Elements (TFSEE) identified key breast cancer subtype-specific TFs that act at transcribed enhancers to dictate gene expression patterns determining growth outcomes, including Forkhead TFs, FOSL1, and PLAG1. FOSL1, a Fos family TF, (1) is highly enriched at the enhancers of triple negative breast cancer (TNBC) cells, (2) acts as a key regulator of the proliferation and viability of TNBC cells, but not Luminal A cells, and (3) is associated with a poor prognosis in TNBC breast cancer patients. Taken together, our results validate our enhancer identification pipeline and reveal that enhancers transcribed in breast cancer cells direct critical gene regulatory networks that promote pathogenesis.


Subject(s)
Carcinogenesis/genetics , Enhancer Elements, Genetic/genetics , Transcriptome/genetics , Triple Negative Breast Neoplasms/genetics , Adult , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks/genetics , Histones/genetics , Humans , Middle Aged , RNA, Messenger/genetics , Transcription Factors/genetics , Triple Negative Breast Neoplasms/classification , Triple Negative Breast Neoplasms/pathology
10.
Genes Dev ; 31(10): 959-972, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28607180

ABSTRACT

DNA methylation is a key regulator of embryonic stem cell (ESC) biology, dynamically changing between naïve, primed, and differentiated states. The p53 tumor suppressor is a pivotal guardian of genomic stability, but its contributions to epigenetic regulation and stem cell biology are less explored. We report that, in naïve mouse ESCs (mESCs), p53 restricts the expression of the de novo DNA methyltransferases Dnmt3a and Dnmt3b while up-regulating Tet1 and Tet2, which promote DNA demethylation. The DNA methylation imbalance in p53-deficient (p53-/-) mESCs is the result of augmented overall DNA methylation as well as increased methylation landscape heterogeneity. In differentiating p53-/- mESCs, elevated methylation persists, albeit more mildly. Importantly, concomitant with DNA methylation heterogeneity, p53-/- mESCs display increased cellular heterogeneity both in the "naïve" state and upon induced differentiation. This impact of p53 loss on 5-methylcytosine (5mC) heterogeneity was also evident in human ESCs and mouse embryos in vivo. Hence, p53 helps maintain DNA methylation homeostasis and clonal homogeneity, a function that may contribute to its tumor suppressor activity.


Subject(s)
DNA Methylation/genetics , Gene Expression Regulation/genetics , Genetic Heterogeneity , Homeostasis/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cell Differentiation/genetics , Clone Cells , DNA (Cytosine-5-)-Methyltransferases/genetics , Embryonic Stem Cells , Gene Deletion , Humans , Mice , Proto-Oncogene Proteins/genetics
11.
Mol Cell ; 64(5): 967-981, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27912097

ABSTRACT

Recent evidence suggests that lncRNAs play an integral regulatory role in numerous functions, including determination of cellular identity. We determined global expression (RNA-seq) and genome-wide profiles (ChIP-seq) of histone post-translational modifications and p53 binding in human embryonic stem cells (hESCs) undergoing differentiation to define a high-confidence set of 40 lncRNAs, which are p53 transcriptional targets. We focused on lncRNAs highly expressed in pluripotent hESCs and repressed by p53 during differentiation to identify lncPRESS1 as a p53-regulated transcript that maintains hESC pluripotency in concert with core pluripotency factors. RNA-seq of hESCs depleted of lncPRESS1 revealed that lncPRESS1 controls a gene network that promotes pluripotency. Further, we found that lncPRESS1 physically interacts with SIRT6 and prevents SIRT6 chromatin localization, which maintains high levels of histone H3K56 and H3K9 acetylation at promoters of pluripotency genes. In summary, we describe a p53-regulated, pluripotency-specific lncRNA that safeguards the hESC state by disrupting SIRT6 activity.


Subject(s)
Cell Differentiation/genetics , Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Histones/metabolism , Pluripotent Stem Cells/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Tumor Suppressor Protein p53/metabolism , Chromatin/metabolism , Embryonic Stem Cells/cytology , Histone Deacetylases , Histones/genetics , Humans , Pluripotent Stem Cells/cytology , Protein Processing, Post-Translational/genetics , Sirtuins/genetics , Sirtuins/metabolism , Tumor Suppressor Protein p53/genetics
12.
Nucleic Acids Res ; 44(8): 3659-74, 2016 05 05.
Article in English | MEDLINE | ID: mdl-26762983

ABSTRACT

Trimethylated histone H3 lysine 27 (H3K27me3) is linked to gene silencing, whereas H3K4me3 is associated with gene activation. These two marks frequently co-occupy gene promoters, forming bivalent domains. Bivalency signifies repressed but activatable states of gene expression and can be resolved to active, H3K4me3-prevalent states during multiple cellular processes, including differentiation, development and epithelial mesenchymal transition. However, the molecular mechanism underlying bivalency resolution remains largely unknown. Here, we show that the H3K27 demethylase UTX (also called KDM6A) is required for the resolution and activation of numerous retinoic acid (RA)-inducible bivalent genes during the RA-driven differentiation of mouse embryonic stem cells (ESCs). Notably, UTX loss in mouse ESCs inhibited the RA-driven bivalency resolution and activation of most developmentally critical homeobox (Hox) a-d genes. The UTX-mediated resolution and activation of many bivalent Hox genes during mouse ESC differentiation were recapitulated during RA-driven differentiation of human NT2/D1 embryonal carcinoma cells. In support of the importance of UTX in bivalency resolution, Utx-null mouse ESCs and UTX-depleted NT2/D1 cells displayed defects in RA-driven cellular differentiation. Our results define UTX as a bivalency-resolving histone modifier necessary for stem cell differentiation.


Subject(s)
Cell Differentiation/genetics , Histone Demethylases/physiology , Nuclear Proteins/physiology , Promoter Regions, Genetic , Transcriptional Activation , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Genes, Homeobox , Histone Demethylases/metabolism , Humans , Mice , Nuclear Proteins/metabolism , Tretinoin/pharmacology
13.
Mol Cell Biol ; 34(14): 2695-709, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24820418

ABSTRACT

Tumor suppressor p53 protects cells from genomic insults and is a target of mutation in more than 50% of human cancers. Stress-mediated modification and increased stability of p53 promote p53 interaction with chromatin, which results in transcription of target genes that are critical for the maintenance of genomic integrity. We recently discovered that TRIM24, an E3-ubiquitin ligase, ubiquitinates and promotes proteasome-mediated degradation of p53. Here, we show that TRIM24 is destabilized by ATM-mediated phosphorylation of TRIM24S768 in response to DNA damage, which disrupts TRIM24-p53 interactions and promotes the degradation of TRIM24. Transcription of TRIM24 is directly induced by damage-activated p53, which binds p53 response elements and activates expression of TRIM24. Newly synthesized TRIM24 interacts with phosphorylated p53 to target it for degradation and termination of the DNA damage response. These studies indicate that TRIM24, like MDM2, controls p53 levels in an autoregulatory feedback loop. However, unlike MDM2, TRIM24 also targets activated p53 to terminate p53-regulated response to DNA damage.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA Damage , Tumor Suppressor Protein p53/metabolism , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , DNA Damage/drug effects , Dactinomycin/pharmacology , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , MCF-7 Cells , Phosphorylation , Proto-Oncogene Proteins c-mdm2/metabolism , Serine/metabolism , Ubiquitination
14.
Nucleic Acids Res ; 42(1): 205-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24078252

ABSTRACT

How tumor suppressor p53 selectively responds to specific signals, especially in normal cells, is poorly understood. We performed genome-wide profiling of p53 chromatin interactions and target gene expression in human embryonic stem cells (hESCs) in response to early differentiation, induced by retinoic acid, versus DNA damage, caused by adriamycin. Most p53-binding sites are unique to each state and define stimulus-specific p53 responses in hESCs. Differentiation-activated p53 targets include many developmental transcription factors and, in pluripotent hESCs, are bound by OCT4 and NANOG at chromatin enriched in both H3K27me3 and H3K4me3. Activation of these genes occurs with recruitment of p53 and H3K27me3-specific demethylases, UTX and JMJD3, to chromatin. In contrast, genes associated with cell migration and motility are bound by p53 specifically after DNA damage. Surveillance functions of p53 in cell death and cell cycle regulation are conserved in both DNA damage and differentiation. Comparative genomic analysis of p53-targets in mouse and human ESCs supports an inter-species divergence in p53 regulatory functions during evolution. Our findings expand the registry of p53-regulated genes to define p53-regulated opposition to pluripotency during early differentiation, a process highly distinct from stress-induced p53 response in hESCs.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Animals , Binding Sites , Cell Line , DNA Damage , Embryonic Stem Cells/cytology , Genome, Human , Histone Demethylases/metabolism , Histones/metabolism , Humans , Mice , Transcription Factors/metabolism
15.
PLoS Biol ; 10(2): e1001268, 2012.
Article in English | MEDLINE | ID: mdl-22389628

ABSTRACT

Multiple studies show that tumor suppressor p53 is a barrier to dedifferentiation; whether this is strictly due to repression of proliferation remains a subject of debate. Here, we show that p53 plays an active role in promoting differentiation of human embryonic stem cells (hESCs) and opposing self-renewal by regulation of specific target genes and microRNAs. In contrast to mouse embryonic stem cells, p53 in hESCs is maintained at low levels in the nucleus, albeit in a deacetylated, inactive state. In response to retinoic acid, CBP/p300 acetylates p53 at lysine 373, which leads to dissociation from E3-ubiquitin ligases HDM2 and TRIM24. Stabilized p53 binds CDKN1A to establish a G(1) phase of cell cycle without activation of cell death pathways. In parallel, p53 activates expression of miR-34a and miR-145, which in turn repress stem cell factors OCT4, KLF4, LIN28A, and SOX2 and prevent backsliding to pluripotency. Induction of p53 levels is a key step: RNA-interference-mediated knockdown of p53 delays differentiation, whereas depletion of negative regulators of p53 or ectopic expression of p53 yields spontaneous differentiation of hESCs, independently of retinoic acid. Ectopic expression of p53R175H, a mutated form of p53 that does not bind DNA or regulate transcription, failed to induce differentiation. These studies underscore the importance of a p53-regulated network in determining the human stem cell state.


Subject(s)
Cell Cycle , Cell Differentiation , Embryonic Stem Cells/physiology , MicroRNAs/metabolism , Tumor Suppressor Protein p53/physiology , Acetylation , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Apoptosis , Base Sequence , Binding Sites , Cells, Cultured , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Humans , Kruppel-Like Factor 4 , MicroRNAs/genetics , Promoter Regions, Genetic , Protein Binding , Protein Processing, Post-Translational , Protein Stability , Transcription, Genetic , Tretinoin/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
16.
J Biol Chem ; 285(21): 16135-44, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20348100

ABSTRACT

Epigenetic control of genes that are silent in embryonic stem cells, but destined for expression during differentiation, includes distinctive hallmarks, such as simultaneous activating/repressing (bivalent) modifications of chromatin and DNA hypomethylation at enhancers of gene expression. Although alpha-fetoprotein (Afp) falls into this class of genes, as it is silent in pluripotent stem cells and activated during differentiation of endoderm, we find that Afp chromatin lacks bivalent histone modifications. However, critical regulatory sites for Afp activation, overlapping Foxa1/p53/Smad-binding elements, are located within a 300-bp region lacking DNA methylation, due to transposed elements underrepresented in CpG sequences: a short interspersed transposable element and a medium reiterated sequence 1 element. Forkhead family member Foxa1 is activated by retinoic acid treatment of embryonic stem cells, binds its DNA consensus site within the short interspersed transposable/medium reiterated sequence 1 elements, and displaces linker histone H1 from silent Afp chromatin. Small interfering RNA depletion of Foxa1 showed that Foxa1 is essential in providing chromatin access to transforming growth factor beta-activated Smad2 and Smad4 and their subsequent DNA binding. Together these transcription factors establish highly acetylated chromatin and promote expression of Afp. Foxa1 acts as a pioneer transcription factor in de novo activation of Afp, by exploiting a lack of methylation at juxtaposed transposed elements, to bind and poise chromatin for intersection with transforming growth factor beta signaling during differentiation of embryonic stem cells.


Subject(s)
Cell Differentiation/physiology , DNA Transposable Elements/physiology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/physiology , Hepatocyte Nuclear Factor 3-alpha/metabolism , alpha-Fetoproteins/biosynthesis , Animals , Antineoplastic Agents/pharmacology , Cell Line , Chromatin Assembly and Disassembly/drug effects , Chromatin Assembly and Disassembly/physiology , DNA Methylation/drug effects , DNA Methylation/physiology , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental/drug effects , Histones/metabolism , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/physiology , Response Elements/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Smad2 Protein/metabolism , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Tretinoin/pharmacology
17.
Proc Natl Acad Sci U S A ; 106(28): 11612-6, 2009 Jul 14.
Article in English | MEDLINE | ID: mdl-19556538

ABSTRACT

Numerous studies focus on the tumor suppressor p53 as a protector of genomic stability, mediator of cell cycle arrest and apoptosis, and target of mutation in 50% of all human cancers. The vast majority of information on p53, its protein-interaction partners and regulation, comes from studies of tumor-derived, cultured cells where p53 and its regulatory controls may be mutated or dysfunctional. To address regulation of endogenous p53 in normal cells, we created a mouse and stem cell model by knock-in (KI) of a tandem-affinity-purification (TAP) epitope at the endogenous Trp-53 locus. Mass spectrometry of TAP-purified p53-complexes from embryonic stem cells revealed Tripartite-motif protein 24 (Trim24), a previously unknown partner of p53. Mutation of TRIM24 homolog, bonus, in Drosophila led to apoptosis, which could be rescued by p53-depletion. These in vivo analyses establish TRIM24/bonus as a pathway that negatively regulates p53 in Drosophila. The Trim24-p53 link is evolutionarily conserved, as TRIM24 depletion in human breast cancer cells caused p53-dependent, spontaneous apoptosis. We found that Trim24 ubiquitylates and negatively regulates p53 levels, suggesting Trim24 as a therapeutic target to restore tumor suppression by p53.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Chromatography, Gel , Drosophila , Gene Knock-In Techniques , Humans , Immunoblotting , Mass Spectrometry , Mice , Mutation/genetics , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...