Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
2.
Pediatr Res ; 95(3): 835-842, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37758866

ABSTRACT

BACKGROUND: Lower respiratory tract infection (LRTI) including pneumonia, bronchitis, and bronchiolitis is the sixth leading cause of mortality around the world and leading cause of death in children under 5 years. Systemic immune response to viral infection is well characterized. However, there is little data regarding the immune response at the upper respiratory tract mucosa. The upper respiratory mucosa is the site of viral entry, initial replication and the first barrier against respiratory infections. Lower respiratory tract samples can be challenging to obtain and require more invasive procedures. However, nasal wash (NW) samples from the upper respiratory tract can be obtained with minimal discomfort to the patient. METHOD: In a pilot study, we developed a protocol using NW samples obtained from hospitalized children with LRTI that enables single cell RNA sequencing (scRNA-seq) after the NW sample is methanol-fixed. RESULTS: We found no significant changes in scRNA-seq qualitative and quantitative parameters between methanol-fixed and fresh NW samples. CONCLUSIONS: We present a novel protocol to enable scRNA-seq in NW samples from children admitted with LRTI. With the inherent challenges associated with clinical samples, the protocol described allows for processing flexibility as well as multicenter collaboration. IMPACT: There are no significant differences in scRNA-seq qualitative and quantitative parameters between methanol fixed and fresh Pediatric Nasal wash samples. The study demonstrates the effectiveness of methanol fixation process on preserving respiratory samples for single cell sequencing. This enables Pediatric Nasal wash specimen for single cell RNA sequencing in pediatric patients with respiratory tract infection and allows processing flexibility and multicenter collaboration.


Subject(s)
Bronchiolitis , Pneumonia , Respiratory Tract Infections , Humans , Child , Infant , Child, Preschool , Methanol , Pilot Projects
3.
Mol Cancer Res ; 21(9): 892-907, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37256926

ABSTRACT

Small cell lung cancer (SCLC) is often a heterogeneous tumor, where dynamic regulation of key transcription factors can drive multiple populations of phenotypically different cells which contribute differentially to tumor dynamics. This tumor is characterized by a very low 2-year survival rate, high rates of metastasis, and rapid acquisition of chemoresistance. The heterogeneous nature of this tumor makes it difficult to study and to treat, as it is not clear how or when this heterogeneity arises. Here we describe temporal, single-cell analysis of SCLC to investigate tumor initiation and chemoresistance in both SCLC xenografts and an autochthonous SCLC model. We identify an early population of tumor cells with high expression of AP-1 network genes that are critical for tumor growth. Furthermore, we have identified and validated the cancer testis antigens (CTA) PAGE5 and GAGE2A as mediators of chemoresistance in human SCLC. CTAs have been successfully targeted in other tumor types and may be a promising avenue for targeted therapy in SCLC. IMPLICATIONS: Understanding the evolutionary dynamics of SCLC can shed light on key mechanisms such as cellular plasticity, heterogeneity, and chemoresistance.


Subject(s)
Drug Resistance, Neoplasm , Lung Neoplasms , Small Cell Lung Carcinoma , Single-Cell Analysis/methods , Humans , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/metabolism , Animals , Mice , Cell Line, Tumor , Transcription Factor AP-1/metabolism , Transcriptome , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism
4.
Neurooncol Adv ; 4(1): vdac072, 2022.
Article in English | MEDLINE | ID: mdl-35855490

ABSTRACT

Background: Neurofibromatosis type 2 (NF2) is an autosomal dominant genetic disease characterized by development of schwannomas on the VIIIth (vestibular) cranial nerves. Bromodomain and extra-terminal domain (BET) proteins regulate gene transcription and their activity is required in a variety of cancers including malignant peripheral nerve sheath tumors. The use of BET inhibitors as a therapeutic option to treat NF2 schwannomas has not been explored and is the focus of this study. Methods: A panel of normal and NF2-null Schwann and schwannoma cell lines were used to characterize the impact of the BET inhibitor JQ1 in vitro and in vivo. The mechanism of action was explored by chromatin immunoprecipitation of the BET BRD4, phospho-kinase arrays and immunohistochemistry (IHC) of BRD4 in vestibular schwannomas. Results: JQ1 inhibited proliferation of NF2-null schwannoma and Schwann cell lines in vitro and in vivo. Further, loss of NF2 by CRISPR deletion or siRNA knockdown increased sensitivity of cells to JQ1. Loss of function experiments identified BRD4, and to a lesser extent BRD2, as BET family members mediating the majority of JQ1 effects. IHC demonstrated elevated levels of BRD4 protein in human vestibular schwannomas. Analysis of signaling pathways effected by JQ1 treatment suggests that the effects of JQ1 treatment are mediated, at least in part, via inhibition of PI3K/Akt signaling. Conclusions: NF2-deficient Schwann and schwannoma cells are sensitive to BET inhibition, primarily mediated by BRD4, which is overexpressed in human vestibular schwannomas. Our results suggest BRD4 regulates PI3K signaling and likely impedes NF2 schwannoma growth via this inhibition. These findings implicate BET inhibition as a therapeutic option for NF2-deficient schwannomas.

5.
Mol Cancer Res ; 19(12): 2015-2025, 2021 12.
Article in English | MEDLINE | ID: mdl-34593608

ABSTRACT

Although many cancer prognoses have improved in the past 50 years due to advancements in treatments, there has been little improvement in therapies for small-cell lung cancer (SCLC). One promising avenue to improve treatment for SCLC is to understand its underlying genetic alterations that drive its formation, growth, and cellular heterogeneity. RB1 loss is one key driver of SCLC, and RB1 loss has been associated with an increase in pluripotency factors such as SOX2. SOX2 is highly expressed and amplified in SCLC and has been associated with SCLC growth. Using a genetically engineered mouse model, we have shown that Sox2 is required for efficient SCLC formation. Furthermore, genome-scale binding assays have indicated that SOX2 can regulate key SCLC pathways such as NEUROD1 and MYC. These data suggest that SOX2 can be associated with the switch of SCLC from an ASCL1 subtype to a NEUROD1 subtype. Understanding this genetic switch is key to understanding such processes as SCLC progression, cellular heterogeneity, and treatment resistance. IMPLICATIONS: Understanding the molecular mechanisms of SCLC initiation and development are key to opening new potential therapeutic options for this devastating disease.


Subject(s)
Lung Neoplasms/genetics , Oncogenes/genetics , SOXB1 Transcription Factors/genetics , Small Cell Lung Carcinoma/genetics , Animals , Cell Line, Tumor , Humans , Mice
6.
J Biol Chem ; 297(2): 100962, 2021 08.
Article in English | MEDLINE | ID: mdl-34265306

ABSTRACT

The Hippo pathway is a key regulatory pathway that is tightly regulated by mechanical cues such as tension, pressure, and contact with the extracellular matrix and other cells. At the distal end of the pathway is the yes-associated protein (YAP), a well-characterized transcriptional regulator. Through binding to transcription factors such as the TEA Domain TFs (TEADs) YAP regulates expression of several genes involved in cell fate, proliferation and death decisions. While the function of YAP as direct transcriptional regulator has been extensively characterized, only a small number of studies examined YAP function as a regulator of gene expression via microRNAs. We utilized bioinformatic approaches, including chromatin immunoprecipitation sequencing and RNA-Seq, to identify potential new targets of YAP regulation and identified miR-30a as a YAP target gene in Schwann cells. We find that YAP binds to the promoter and regulates the expression of miR-30a. Moreover, we identify several YAP-regulated genes that are putative miR-30a targets and focus on two of these, protein tyrosine pohosphatase non-receptor type 13 (PTPN13) and Kruppel like factor 9. We find that YAP regulation of Schwann cell proliferation and death is mediated, to a significant extent, through miR-30a regulation of PTPN13 in Schwann cells. These findings identify a new regulatory function by YAP, mediated by miR-30a, to downregulate expression of PTPN13 and Kruppel like factor 9. These studies expand our understanding of YAP function as a regulator of miRNAs and illustrate the complexity of YAP transcriptional functions.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , MicroRNAs , Protein Tyrosine Phosphatase, Non-Receptor Type 13/metabolism , Schwann Cells/pathology , Adaptor Proteins, Signal Transducing , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Humans , Signal Transduction , Transcription, Genetic , YAP-Signaling Proteins
7.
Methods Mol Biol ; 2352: 171-181, 2021.
Article in English | MEDLINE | ID: mdl-34324187

ABSTRACT

The processes that underlie neuronal conversion ultimately involve a reorganization of transcriptional networks to establish a neuronal cell fate. As such, transcriptional profiling is a key component toward understanding this process. In this chapter, we will discuss methods of elucidating transcriptional networks during neuronal reprogramming and considerations that should be incorporated in experimental design.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Gene Expression Profiling , Neurons/cytology , Neurons/metabolism , Transcriptome , Gene Expression Regulation, Developmental , High-Throughput Nucleotide Sequencing , Humans , Sequence Analysis, RNA , Single-Cell Analysis/methods
8.
Cancer Res ; 80(12): 2512-2522, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32409309

ABSTRACT

The Hippo pathway regulates cell proliferation and organ size through control of the transcriptional regulators YAP (yes-associated protein) and TAZ. Upon extracellular stimuli such as cell-cell contact, the pathway negatively regulates YAP through cytoplasmic sequestration. Under conditions of low cell density, YAP is nuclear and associates with enhancer regions and gene promoters. YAP is mainly described as a transcriptional activator of genes involved in cell proliferation and survival. Using a genome-wide approach, we show here that, in addition to its known function as a transcriptional activator, YAP functions as a transcriptional repressor by interacting with the multifunctional transcription factor Yin Yang 1 (YY1) and Polycomb repressive complex member enhancer of zeste homologue 2 (EZH2). YAP colocalized with YY1 and EZH2 on the genome to transcriptionally repress a broad network of genes mediating a host of cellular functions, including repression of the cell-cycle kinase inhibitor p27, whose role is to functionally promote contact inhibition. This work unveils a broad and underappreciated aspect of YAP nuclear function as a transcriptional repressor and highlights how loss of contact inhibition in cancer is mediated in part through YAP repressive function. SIGNIFICANCE: This study provides new insights into YAP as a broad transcriptional repressor of key regulators of the cell cycle, in turn influencing contact inhibition and tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Neoplasms/genetics , Transcription Factors/metabolism , Transcription, Genetic , YY1 Transcription Factor/metabolism , Animals , Carcinogenesis/genetics , Cell Fractionation , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks/genetics , Humans , Mice , Neoplasms/pathology , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Xenograft Model Antitumor Assays , YAP-Signaling Proteins
9.
Nat Cell Biol ; 22(4): 401-411, 2020 04.
Article in English | MEDLINE | ID: mdl-32231311

ABSTRACT

The on-target pioneer factors Ascl1 and Myod1 are sequence-related but induce two developmentally unrelated lineages-that is, neuronal and muscle identities, respectively. It is unclear how these two basic helix-loop-helix (bHLH) factors mediate such fundamentally different outcomes. The chromatin binding of Ascl1 and Myod1 was surprisingly similar in fibroblasts, yet their transcriptional outputs were drastically different. We found that quantitative binding differences explained differential chromatin remodelling and gene activation. Although strong Ascl1 binding was exclusively associated with bHLH motifs, strong Myod1-binding sites were co-enriched with non-bHLH motifs, possibly explaining why Ascl1 is less context dependent. Finally, we observed that promiscuous binding of Myod1 to neuronal targets results in neuronal reprogramming when the muscle program is inhibited by Myt1l. Our findings suggest that chromatin access of on-target pioneer factors is primarily driven by the protein-DNA interaction, unlike ordinary context-dependent transcription factors, and that promiscuous transcription factor binding requires specific silencing mechanisms to ensure lineage fidelity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , MyoD Protein/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , Transcription Factors/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites , Cell Lineage/genetics , Cellular Reprogramming , Chromatin/chemistry , Chromatin/metabolism , Embryo, Mammalian , Fibroblasts/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , MyoD Protein/metabolism , Nerve Tissue Proteins/metabolism , Neurons/cytology , Nucleotide Motifs , Protein Binding , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic
10.
Elife ; 82019 01 15.
Article in English | MEDLINE | ID: mdl-30644360

ABSTRACT

Direct reprogramming of fibroblasts to neurons induces widespread cellular and transcriptional reconfiguration. Here, we characterized global epigenomic changes during the direct reprogramming of mouse fibroblasts to neurons using whole-genome base-resolution DNA methylation (mC) sequencing. We found that the pioneer transcription factor Ascl1 alone is sufficient for inducing the uniquely neuronal feature of non-CG methylation (mCH), but co-expression of Brn2 and Mytl1 was required to establish a global mCH pattern reminiscent of mature cortical neurons. Ascl1 alone induced promoter CG methylation (mCG) of fibroblast specific genes, while BAM overexpression additionally targets a competing myogenic program and directs a more faithful conversion to neuronal cells. Ascl1 induces local demethylation at its binding sites. Surprisingly, co-expression with Brn2 and Mytl1 inhibited the ability of Ascl1 to induce demethylation, suggesting a contextual regulation of transcription factor - epigenome interaction. Finally, we found that de novo methylation by DNMT3A is required for efficient neuronal reprogramming.


Subject(s)
Cellular Reprogramming/genetics , DNA Methylation/genetics , Fibroblasts/cytology , Neurons/cytology , Animals , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Down-Regulation/genetics , Gene Expression Regulation, Developmental , Gene Silencing , Mice, Inbred C57BL , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcription, Genetic
11.
Science ; 362(6410): 30-31, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30287650

Subject(s)
Neoplasms , Epithelium , Humans
12.
SPG Biomed ; 1(2)2018.
Article in English | MEDLINE | ID: mdl-31172135

ABSTRACT

Embryonic stem cells possess the ability to differentiate into all cell types of the body. This pliable developmental state is achieved by the function of a series of pluripotency factors, classically identified as OCT4, SOX2, and NANOG. These pluripotency factors are responsible for activating the larger pluripotency networks and the self-renewal programs which give ES cells their unique characteristics. However, during differentiation pluripotency networks become downregulated as cells achieve greater lineage specification and exit the cell cycle. Typically the repression of pluripotency is viewed as a positive factor to ensure the fidelity of cellular identity by restricting cellular pliancy. Consistent with this view, the expression of pluripotency factors is greatly restricted in somatic cells. However, there are examples whereby cells either maintain or reactivate pluripotency factors to preserve the increased potential for the healing of wounds or tissue homeostasis. Additionally there are many examples where these pluripotency factors become reactivated in a variety of human pathologies, particularly cancer. In this review, we will summarize the somatic repression of pluripotency factors, their role in tissue homeostasis and wound repair, and the human diseases that are associated with pluripotency factor misregulation with an emphasis on their role in the etiology of multiple cancers.

13.
Nature ; 544(7649): 245-249, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28379941

ABSTRACT

Normal differentiation and induced reprogramming require the activation of target cell programs and silencing of donor cell programs. In reprogramming, the same factors are often used to reprogram many different donor cell types. As most developmental repressors, such as RE1-silencing transcription factor (REST) and Groucho (also known as TLE), are considered lineage-specific repressors, it remains unclear how identical combinations of transcription factors can silence so many different donor programs. Distinct lineage repressors would have to be induced in different donor cell types. Here, by studying the reprogramming of mouse fibroblasts to neurons, we found that the pan neuron-specific transcription factor Myt1-like (Myt1l) exerts its pro-neuronal function by direct repression of many different somatic lineage programs except the neuronal program. The repressive function of Myt1l is mediated via recruitment of a complex containing Sin3b by binding to a previously uncharacterized N-terminal domain. In agreement with its repressive function, the genomic binding sites of Myt1l are similar in neurons and fibroblasts and are preferentially in an open chromatin configuration. The Notch signalling pathway is repressed by Myt1l through silencing of several members, including Hes1. Acute knockdown of Myt1l in the developing mouse brain mimicked a Notch gain-of-function phenotype, suggesting that Myt1l allows newborn neurons to escape Notch activation during normal development. Depletion of Myt1l in primary postmitotic neurons de-repressed non-neuronal programs and impaired neuronal gene expression and function, indicating that many somatic lineage programs are actively and persistently repressed by Myt1l to maintain neuronal identity. It is now tempting to speculate that similar 'many-but-one' lineage repressors exist for other cell fates; such repressors, in combination with lineage-specific activators, would be prime candidates for use in reprogramming additional cell types.


Subject(s)
Cell Lineage/genetics , Cellular Reprogramming/genetics , Gene Silencing , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Neurons/cytology , Neurons/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Brain/cytology , Brain/embryology , Brain/metabolism , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mice , Nerve Tissue Proteins/deficiency , Organ Specificity/genetics , Protein Domains , Receptors, Notch/deficiency , Repressor Proteins/chemistry , Repressor Proteins/deficiency , Signal Transduction , Transcription Factor HES-1/deficiency , Transcription Factors/deficiency
14.
Cell Rep ; 16(3): 644-56, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27373157

ABSTRACT

Small cell lung cancer (SCLC) is a neuroendocrine lung cancer characterized by fast growth, early dissemination, and rapid resistance to chemotherapy. We identified a population of long-term tumor-propagating cells (TPCs) in a mouse model of SCLC. This population, marked by high levels of EpCAM and CD24, is also prevalent in human primary SCLC tumors. Murine SCLC TPCs are numerous and highly proliferative but not intrinsically chemoresistant, indicating that not all clinical features of SCLC are linked to TPCs. SCLC TPCs possess a distinct transcriptional profile compared to non-TPCs, including elevated MYC activity. Genetic and pharmacological inhibition of MYC in SCLC cells to non-TPC levels inhibits long-term propagation but not short-term growth. These studies identify a highly tumorigenic population of SCLC cells in mouse models, cell lines, and patient tumors and a means to target them in this most fatal form of lung cancer.


Subject(s)
Lung Neoplasms/pathology , Small Cell Lung Carcinoma/pathology , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/physiology , Disease Models, Animal , Humans , Lung Neoplasms/genetics , Mice , Small Cell Lung Carcinoma/genetics , Transcription, Genetic/physiology
15.
Curr Pharmacol Rep ; 2(3): 103-112, 2016 Jun.
Article in English | MEDLINE | ID: mdl-35663262

ABSTRACT

Cellular reprogramming, whereby cell fate can be changed by the expression of a few defined factors, is a remarkable process that harnesses the innate ability of a cell's own genome to rework its expressional networks and function. Since cell lineages are defined by global regulation of gene expression, transcriptional regulators, and coupled to the epigenetic markings of the chromatin, changing the cell fate necessitates broad changes to these central cellular features. To properly characterize these changes, and the mechanisms that drive them, computational and genomic approaches are perfectly suited to provide a holistic picture of the reprogramming mechanisms. In particular, the use of bioinformatic analysis has been a major driver in the study of cellular reprogramming, both as it relates to induced pluripotency or direct lineage conversion. This review will summarize many of the bioinformatic studies that have advanced our knowledge of reprogramming and address future directions for these investigations.

16.
Curr Opin Cell Biol ; 37: 68-74, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26520682

ABSTRACT

Pluripotent stem cells, defined by an unlimited self-renewal capacity and an undifferentiated state, are best typified by embryonic stem cells. These cells have a unique cell cycle compared to somatic cells as defined by a rapid progression through the cell cycle and a minimal time spent in G1. Recent reports indicate that pluripotency and cell cycle regulation are mechanistically linked. In this review, we discuss the reciprocal co-regulation of these processes, how this co-regulation may prevent differentiation, and how cellular reprogramming can re-establish the unique cell cycle regulation in induced pluripotent stem cells.


Subject(s)
Cell Cycle , Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation/physiology , Cellular Reprogramming , Humans
17.
Cell Stem Cell ; 16(1): 39-50, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25467916

ABSTRACT

Mutations in the retinoblastoma tumor suppressor gene Rb are involved in many forms of human cancer. In this study, we investigated the early consequences of inactivating Rb in the context of cellular reprogramming. We found that Rb inactivation promotes the reprogramming of differentiated cells to a pluripotent state. Unexpectedly, this effect is cell cycle independent, and instead reflects direct binding of Rb to pluripotency genes, including Sox2 and Oct4, which leads to a repressed chromatin state. More broadly, this regulation of pluripotency networks and Sox2 in particular is critical for the initiation of tumors upon loss of Rb in mice. These studies therefore identify Rb as a global transcriptional repressor of pluripotency networks, providing a molecular basis for previous reports about its involvement in cell fate pliability, and implicate misregulation of pluripotency factors such as Sox2 in tumorigenesis related to loss of Rb function.


Subject(s)
Carcinogenesis/pathology , Cellular Reprogramming , Induced Pluripotent Stem Cells/metabolism , Retinoblastoma Protein/metabolism , Animals , Carcinogenesis/metabolism , Cell Cycle , Chromatin/metabolism , Fibroblasts/metabolism , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Protein Binding , Repressor Proteins/metabolism , Retinoblastoma Protein/deficiency , SOXB1 Transcription Factors/genetics
18.
J Biol Chem ; 286(27): 24300-11, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21489993

ABSTRACT

Histone modifications and DNA methylation represent two layers of heritable epigenetic information that regulate eukaryotic chromatin structure and gene activity. UHRF1 is a unique factor that bridges these two layers; it is required for maintenance DNA methylation at hemimethylated CpG sites, which are specifically recognized through its SRA domain and also interacts with histone H3 trimethylated on lysine 9 (H3K9me3) in an unspecified manner. Here we show that UHRF1 contains a tandem Tudor domain (TTD) that recognizes H3 tail peptides with the heterochromatin-associated modification state of trimethylated lysine 9 and unmodified lysine 4 (H3K4me0/K9me3). Solution NMR and crystallographic data reveal the TTD simultaneously recognizes H3K9me3 through a conserved aromatic cage in the first Tudor subdomain and unmodified H3K4 within a groove between the tandem subdomains. The subdomains undergo a conformational adjustment upon peptide binding, distinct from previously reported mechanisms for dual histone mark recognition. Mutant UHRF1 protein deficient for H3K4me0/K9me3 binding shows altered localization to heterochromatic chromocenters and fails to reduce expression of a target gene, p16(INK4A), when overexpressed. Our results demonstrate a novel recognition mechanism for the combinatorial readout of histone modification states associated with gene silencing and add to the growing evidence for coordination of, and cross-talk between, the modification states of H3K4 and H3K9 in regulation of gene expression.


Subject(s)
CCAAT-Enhancer-Binding Proteins/chemistry , CCAAT-Enhancer-Binding Proteins/metabolism , Heterochromatin/chemistry , Heterochromatin/metabolism , Histones/chemistry , Histones/metabolism , Animals , Binding Sites , CCAAT-Enhancer-Binding Proteins/genetics , CpG Islands/physiology , Crystallography, X-Ray , Cyclin-Dependent Kinase Inhibitor p16/chemistry , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Methylation/physiology , Gene Expression Regulation/physiology , Heterochromatin/genetics , Histones/genetics , Humans , Mice , Mice, Knockout , Nuclear Magnetic Resonance, Biomolecular , Protein Processing, Post-Translational/physiology , Protein Structure, Tertiary , Ubiquitin-Protein Ligases
19.
PLoS Genet ; 6(9): e1001106, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20838592

ABSTRACT

The DNTM3A and DNMT3B de novo DNA methyltransferases (DNMTs) are responsible for setting genomic DNA methylation patterns, a key layer of epigenetic information. Here, using an in vivo episomal methylation assay and extensive bisulfite methylation sequencing, we show that human DNMT3A and DNMT3B possess significant and distinct flanking sequence preferences for target CpG sites. Selection for high or low efficiency sites is mediated by the base composition at the -2 and +2 positions flanking the CpG site for DNMT3A, and at the -1 and +1 positions for DNMT3B. This intrinsic preference reproducibly leads to the formation of specific de novo methylation patterns characterized by up to 34-fold variations in the efficiency of DNA methylation at individual sites. Furthermore, analysis of the distribution of signature methylation hotspot and coldspot motifs suggests that DNMT flanking sequence preference has contributed to shaping the composition of CpG islands in the human genome. Our results also show that the DNMT3L stimulatory factor modulates the formation of de novo methylation patterns in two ways. First, DNMT3L selectively focuses the DNA methylation machinery on properly chromatinized DNA templates. Second, DNMT3L attenuates the impact of the intrinsic DNMT flanking sequence preference by providing a much greater boost to the methylation of poorly methylated sites, thus promoting the formation of broader and more uniform methylation patterns. This study offers insights into the manner by which DNA methylation patterns are deposited and reveals a new level of interplay between members of the de novo DNMT family.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , DNA, Intergenic/metabolism , Animals , Base Sequence , Cell Cycle Proteins/metabolism , Cell Line , Chromatin/metabolism , CpG Islands/genetics , DNA Methyltransferase 3A , DNA Replication/genetics , DNA, Intergenic/genetics , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mammals/genetics , Molecular Sequence Data , Protein Binding , Reproducibility of Results , Templates, Genetic , DNA Methyltransferase 3B
20.
PLoS Genet ; 6(6): e1001003, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20585628

ABSTRACT

The retinoblastoma tumor suppressor (Rb) is a potent and ubiquitously expressed cell cycle regulator, but patients with a germline Rb mutation develop a very specific tumor spectrum. This surprising observation raises the possibility that mechanisms that compensate for loss of Rb function are present or activated in many cell types. In particular, p107, a protein related to Rb, has been shown to functionally overlap for loss of Rb in several cellular contexts. To investigate the mechanisms underlying this functional redundancy between Rb and p107 in vivo, we used gene targeting in embryonic stem cells to engineer point mutations in two consensus E2F binding sites in the endogenous p107 promoter. Analysis of normal and mutant cells by gene expression and chromatin immunoprecipitation assays showed that members of the Rb and E2F families directly bound these two sites. Furthermore, we found that these two E2F sites controlled both the repression of p107 in quiescent cells and also its activation in cycling cells, as well as in Rb mutant cells. Cell cycle assays further indicated that activation of p107 transcription during S phase through the two E2F binding sites was critical for controlled cell cycle progression, uncovering a specific role for p107 to slow proliferation in mammalian cells. Direct transcriptional repression of p107 by Rb and E2F family members provides a molecular mechanism for a critical negative feedback loop during cell cycle progression and tumorigenesis. These experiments also suggest novel therapeutic strategies to increase the p107 levels in tumor cells.


Subject(s)
Cell Cycle , E2F Transcription Factors/metabolism , Promoter Regions, Genetic , Retinoblastoma-Like Protein p107/metabolism , Animals , Base Sequence , Binding Sites , Cell Cycle Proteins , Cells, Cultured , Conserved Sequence , Humans , Mice , Molecular Sequence Data , Retinoblastoma-Like Protein p107/chemistry , Retinoblastoma-Like Protein p107/genetics , Sequence Alignment , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...