Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
NAR Cancer ; 6(1): zcae009, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38444581

ABSTRACT

Translational regulation is an important step in the control of gene expression. In cancer cells, the orchestration of both global control of protein synthesis and selective translation of specific mRNAs promote tumor cell survival, angiogenesis, transformation, invasion and metastasis. N6-methyladenosine (m6A), the most prevalent mRNA modification in higher eukaryotes, impacts protein translation. Over the past decade, the development of m6A mapping tools has facilitated comprehensive functional investigations, revealing the involvement of this chemical mark, together with its writer METTL3, in promoting the translation of both oncogenes and tumor suppressor transcripts, with the impact being context-dependent. This review aims to consolidate our current understanding of how m6A and METTL3 shape translation regulation in the realm of cancer biology. In addition, it delves into the role of cytoplasmic METTL3 in protein synthesis, operating independently of its catalytic activity. Ultimately, our goal is to provide critical insights into the interplay between m6A, METTL3 and translational regulation in cancer, offering a deeper comprehension of the mechanisms sustaining tumorigenesis.

2.
Nat Commun ; 14(1): 1694, 2023 03 27.
Article in English | MEDLINE | ID: mdl-36973285

ABSTRACT

N6-methyladenosine (m6A), one of the most prevalent mRNA modifications in eukaryotes, plays a critical role in modulating both biological and pathological processes. However, it is unknown whether mutant p53 neomorphic oncogenic functions exploit dysregulation of m6A epitranscriptomic networks. Here, we investigate Li-Fraumeni syndrome (LFS)-associated neoplastic transformation driven by mutant p53 in iPSC-derived astrocytes, the cell-of-origin of gliomas. We find that mutant p53 but not wild-type (WT) p53 physically interacts with SVIL to recruit the H3K4me3 methyltransferase MLL1 to activate the expression of m6A reader YTHDF2, culminating in an oncogenic phenotype. Aberrant YTHDF2 upregulation markedly hampers expression of multiple m6A-marked tumor-suppressing transcripts, including CDKN2B and SPOCK2, and induces oncogenic reprogramming. Mutant p53 neoplastic behaviors are significantly impaired by genetic depletion of YTHDF2 or by pharmacological inhibition using MLL1 complex inhibitors. Our study reveals how mutant p53 hijacks epigenetic and epitranscriptomic machinery to initiate gliomagenesis and suggests potential treatment strategies for LFS gliomas.


Subject(s)
Glioma , Li-Fraumeni Syndrome , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Li-Fraumeni Syndrome/genetics , Cell Transformation, Neoplastic/genetics , Glioma/genetics , Proteoglycans/metabolism
3.
Oncogene ; 42(12): 911-925, 2023 03.
Article in English | MEDLINE | ID: mdl-36725888

ABSTRACT

Alternative splicing (AS) enables differential inclusion of exons from a given transcript, thereby contributing to the transcriptome and proteome diversity. Aberrant AS patterns play major roles in the development of different pathologies, including breast cancer. N6-methyladenosine (m6A), the most abundant internal modification of eukaryotic mRNA, influences tumor progression and metastasis of breast cancer, and it has been recently linked to AS regulation. Here, we identify a specific AS signature associated with breast tumorigenesis in vitro. We characterize for the first time the role of METTL3 in modulating breast cancer-associated AS programs, expanding the role of the m6A-methyltransferase in tumorigenesis. Specifically, we find that both m6A deposition in splice site boundaries and in splicing and transcription factor transcripts, such as MYC, direct AS switches of specific breast cancer-associated transcripts. Finally, we show that five of the AS events validated in vitro are associated with a poor overall survival rate for patients with breast cancer, suggesting the use of these AS events as a novel potential prognostic biomarker.


Subject(s)
Alternative Splicing , Breast Neoplasms , Humans , Female , Alternative Splicing/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Methyltransferases/genetics , Methyltransferases/metabolism , Transcriptome , Carcinogenesis
4.
EMBO Rep ; 23(3): e53191, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35037361

ABSTRACT

The pluripotent state is not solely governed by the action of the core transcription factors OCT4, SOX2, and NANOG, but also by a series of co-transcriptional and post-transcriptional events, including alternative splicing (AS) and the interaction of RNA-binding proteins (RBPs) with defined subpopulations of RNAs. Zinc Finger Protein 207 (ZFP207) is an essential transcription factor for mammalian embryonic development. Here, we employ multiple functional analyses to characterize its role in mouse embryonic stem cells (ESCs). We find that ZFP207 plays a pivotal role in ESC maintenance, and silencing of Zfp207 leads to severe neuroectodermal differentiation defects. In striking contrast to human ESCs, mouse ZFP207 does not transcriptionally regulate neuronal and stem cell-related genes but exerts its effects by controlling AS networks and by acting as an RBP. Our study expands the role of ZFP207 in maintaining ESC identity, and underscores the functional versatility of ZFP207 in regulating neural fate commitment.


Subject(s)
Alternative Splicing , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Octamer Transcription Factor-3/metabolism , RNA , Animals , Cell Differentiation/genetics , Mice , Mouse Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , RNA/metabolism
5.
Methods Mol Biol ; 2404: 355-362, 2022.
Article in English | MEDLINE | ID: mdl-34694619

ABSTRACT

N6-methyladenosine (m6A) is the most abundant internal modification on messenger RNAs (mRNAs) and long noncoding RNAs (lncRNAs) in eukaryotes. It influences gene expression by regulating RNA processing, nuclear export, mRNA decay, and translation. Hence, m6A controls fundamental cellular processes, and dysregulated deposition of m6A has been acknowledged to play a role in a broad range of human diseases, including cancer. m6A RNA immunoprecipitation followed by high-throughput sequencing (MeRIP-seq or m6A-seq) is a powerful technique to map m6A in a transcriptome-wide level. After immunoprecipitation of fragmented polyadenylated (poly(A)+) rich RNA by using specific anti-m6A antibodies, both the immunoprecipitated RNA fragments together with the input control are subjected to massively parallel sequencing. The generation of such comprehensive methylation profiles of signal enrichment relative to input control is necessary in order to better comprehend the pathogenesis behind aberrant m6A deposition.


Subject(s)
High-Throughput Nucleotide Sequencing , Adenosine/analogs & derivatives , Gene Expression Profiling , Humans , Immunoprecipitation , RNA/genetics
6.
Nucleic Acids Res ; 50(D1): D231-D235, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34893873

ABSTRACT

The MODOMICS database has been, since 2006, a manually curated and centralized resource, storing and distributing comprehensive information about modified ribonucleosides. Originally, it only contained data on the chemical structures of modified ribonucleosides, their biosynthetic pathways, the location of modified residues in RNA sequences, and RNA-modifying enzymes. Over the years, prompted by the accumulation of new knowledge and new types of data, it has been updated with new information and functionalities. In this new release, we have created a catalog of RNA modifications linked to human diseases, e.g., due to mutations in genes encoding modification enzymes. MODOMICS has been linked extensively to RCSB Protein Data Bank, and sequences of experimentally determined RNA structures with modified residues have been added. This expansion was accompanied by including nucleotide 5'-monophosphate residues. We redesigned the web interface and upgraded the database backend. In addition, a search engine for chemically similar modified residues has been included that can be queried by SMILES codes or by drawing chemical molecules. Finally, previously available datasets of modified residues, biosynthetic pathways, and RNA-modifying enzymes have been updated. Overall, we provide users with a new, enhanced, and restyled tool for research on RNA modification. MODOMICS is available at https://iimcb.genesilico.pl/modomics/.


Subject(s)
Databases, Nucleic Acid , Enzymes/genetics , RNA/genetics , Ribonucleosides/genetics , User-Computer Interface , Base Sequence , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Computer Graphics , Databases, Protein , Datasets as Topic , Enzymes/metabolism , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/pathology , Hematologic Diseases/genetics , Hematologic Diseases/metabolism , Hematologic Diseases/pathology , Humans , Internet , Mental Disorders/genetics , Mental Disorders/metabolism , Mental Disorders/pathology , Musculoskeletal Diseases/genetics , Musculoskeletal Diseases/metabolism , Musculoskeletal Diseases/pathology , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , RNA/metabolism , RNA Processing, Post-Transcriptional , Ribonucleosides/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
7.
Cells ; 10(11)2021 11 20.
Article in English | MEDLINE | ID: mdl-34831474

ABSTRACT

Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSC) provide a powerful model system to uncover fundamental mechanisms that control cellular identity during mammalian development. Histone methylation governs gene expression programs that play a key role in the regulation of the balance between self-renewal and differentiation of ESCs. Lysine-specific demethylase 1 (LSD1, also known as KDM1A), the first identified histone lysine demethylase, demethylates H3K4me1/2 and H3K9me1/2 at target loci in a context-dependent manner. Moreover, it has also been shown to demethylate non-histone substrates playing a central role in the regulation of numerous cellular processes. In this review, we summarize current knowledge about LSD1 and the molecular mechanism by which LSD1 influences the stem cells state, including the regulatory circuitry underlying self-renewal and pluripotency.


Subject(s)
Cell Differentiation , Histone Demethylases/metabolism , Stem Cells/cytology , Stem Cells/enzymology , Animals , Cell Self Renewal , Cellular Reprogramming , DNA Methylation/genetics , Histone Demethylases/chemistry , Humans
8.
NAR Cancer ; 3(3): zcab036, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34541538

ABSTRACT

Collectively referred to as the epitranscriptome, RNA modifications play important roles in gene expression control regulating relevant cellular processes. In the last few decades, growing numbers of RNA modifications have been identified not only in abundant ribosomal (rRNA) and transfer RNA (tRNA) but also in messenger RNA (mRNA). In addition, many writers, erasers and readers that dynamically regulate the chemical marks have also been characterized. Correct deposition of RNA modifications is prerequisite for cellular homeostasis, and its alteration results in aberrant transcriptional programs that dictate human disease, including breast cancer, the most frequent female malignancy, and the leading cause of cancer-related death in women. In this review, we emphasize the major RNA modifications that are present in tRNA, rRNA and mRNA. We have categorized breast cancer-associated chemical marks and summarize their contribution to breast tumorigenesis. In addition, we describe less abundant tRNA modifications with related pathways implicated in breast cancer. Finally, we discuss current limitations and perspectives on epitranscriptomics for use in therapeutic strategies against breast and other cancers.

9.
Nat Commun ; 12(1): 1716, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33741917

ABSTRACT

Cancer stem cells (CSCs) are a small but critical cell population for cancer biology since they display inherent resistance to standard therapies and give rise to metastases. Despite accruing evidence establishing a link between deregulation of epitranscriptome-related players and tumorigenic process, the role of messenger RNA (mRNA) modifications in the regulation of CSC properties remains poorly understood. Here, we show that the cytoplasmic pool of fat mass and obesity-associated protein (FTO) impedes CSC abilities in colorectal cancer through its N6,2'-O-dimethyladenosine (m6Am) demethylase activity. While m6Am is strategically located next to the m7G-mRNA cap, its biological function is not well understood and has not been addressed in cancer. Low FTO expression in patient-derived cell lines elevates m6Am level in mRNA which results in enhanced in vivo tumorigenicity and chemoresistance. Inhibition of the nuclear m6Am methyltransferase, PCIF1/CAPAM, fully reverses this phenotype, stressing the role of m6Am modification in stem-like properties acquisition. FTO-mediated regulation of m6Am marking constitutes a reversible pathway controlling CSC abilities. Altogether, our findings bring to light the first biological function of the m6Am modification and its potential adverse consequences for colorectal cancer management.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Colorectal Neoplasms/metabolism , Cytoplasm/metabolism , Demethylation , Adaptor Proteins, Signal Transducing/metabolism , Adenosine/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Methyltransferases/metabolism , Nuclear Proteins/metabolism , RNA, Messenger/metabolism
10.
RNA ; 27(4): 367-389, 2021 04.
Article in English | MEDLINE | ID: mdl-33376192

ABSTRACT

RNA modifications have recently emerged as a widespread and complex facet of gene expression regulation. Counting more than 170 distinct chemical modifications with far-reaching implications for RNA fate, they are collectively referred to as the epitranscriptome. These modifications can occur in all RNA species, including messenger RNAs (mRNAs) and noncoding RNAs (ncRNAs). In mRNAs the deposition, removal, and recognition of chemical marks by writers, erasers and readers influence their structure, localization, stability, and translation. In turn, this modulates key molecular and cellular processes such as RNA metabolism, cell cycle, apoptosis, and others. Unsurprisingly, given their relevance for cellular and organismal functions, alterations of epitranscriptomic marks have been observed in a broad range of human diseases, including cancer, neurological and metabolic disorders. Here, we will review the major types of mRNA modifications and editing processes in conjunction with the enzymes involved in their metabolism and describe their impact on human diseases. We present the current knowledge in an updated catalog. We will also discuss the emerging evidence on the crosstalk of epitranscriptomic marks and what this interplay could imply for the dynamics of mRNA modifications. Understanding how this complex regulatory layer can affect the course of human pathologies will ultimately lead to its exploitation toward novel epitranscriptomic therapeutic strategies.


Subject(s)
Metabolic Diseases/genetics , Neoplasms/genetics , Nervous System Diseases/genetics , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Untranslated/genetics , Apoptosis/genetics , Cell Cycle/genetics , Epigenesis, Genetic , Genetic Markers , Humans , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Neoplasms/metabolism , Neoplasms/pathology , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , RNA, Messenger/metabolism , RNA, Untranslated/metabolism
11.
Biochim Biophys Acta Gene Regul Mech ; 1862(3): 394-402, 2019 03.
Article in English | MEDLINE | ID: mdl-30412796

ABSTRACT

Chemical modifications of RNA provide a direct and rapid way to modulate the existing transcriptome, allowing the cells to adapt rapidly to the changing environment. Among these modifications, N6-methyladenosine (m6A) has recently emerged as a widely prevalent mark of messenger RNA in eukaryotes, linking external stimuli to an intricate network of transcriptional, post-transcriptional and translational processes. m6A modification modulates a broad spectrum of biochemical processes, including mRNA decay, translation and splicing. Both m6A modification and the enzymes that control m6A metabolism are essential for normal development. In this review, we summarized the most recent findings on the role of m6A modification in maintenance of the pluripotency of embryonic stem cells (ESCs), cell fate specification, the reprogramming of somatic cells into induced pluripotent stem cells (iPSCs), and differentiation of stem and progenitor cells. This article is part of a Special Issue entitled: mRNA modifications in gene expression control edited by Dr. Soller Matthias and Dr. Fray Rupert.


Subject(s)
Adenosine/analogs & derivatives , Cell Differentiation , Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , RNA Processing, Post-Transcriptional , Adenosine/metabolism , Animals , Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology
12.
Front Biosci (Landmark Ed) ; 23(11): 2106-2132, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29772549

ABSTRACT

High-throughput analyses have revealed that the vast majority of the transcriptome does not code for proteins. These non-translated transcripts, when larger than 200 nucleotides, are termed long non-coding RNAs (lncRNAs), and play fundamental roles in diverse cellular processes. LncRNAs are subject to dynamic chemical modification, adding another layer of complexity to our understanding of the potential roles that lncRNAs play in health and disease. Many lncRNAs regulate transcriptional programs by influencing the epigenetic state through direct interactions with chromatin-modifying proteins. Among these proteins, Polycomb repressive complexes 1 and 2 (PRC1 and PRC2) have been shown to be recruited by lncRNAs to silence target genes. Aberrant expression, deficiency or mutation of both lncRNA and Polycomb have been associated with numerous human diseases, including cancer. In this review, we have highlighted recent findings regarding the concerted mechanism of action of Polycomb group proteins (PcG), acting together with some classically defined lncRNAs including X-inactive specific transcript (XIST), antisense non-coding RNA in the INK4 locus (ANRIL), metastasis associated lung adenocarcinoma transcript 1 (MALAT1), and HOX transcript antisense RNA (HOTAIR).


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Polycomb-Group Proteins/genetics , RNA, Long Noncoding/genetics , Animals , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/metabolism , Polycomb-Group Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA, Long Noncoding/metabolism , Signal Transduction/genetics
13.
Nucleic Acids Res ; 45(19): 11106-11120, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-28977470

ABSTRACT

We observed overexpression and increased intra-nuclear accumulation of the PRMT5/WDR77 in breast cancer cell lines relative to immortalized breast epithelial cells. Utilizing mass spectrometry and biochemistry approaches we identified the Zn-finger protein ZNF326, as a novel interaction partner and substrate of the nuclear PRMT5/WDR77 complex. ZNF326 is symmetrically dimethylated at arginine 175 (R175) and this modification is lost in a PRMT5 and WDR77-dependent manner. Loss of PRMT5 or WDR77 in MDA-MB-231 cells leads to defects in alternative splicing, including inclusion of A-T rich exons in target genes, a phenomenon that has previously been observed upon loss of ZNF326. We observed that the alternatively spliced transcripts of a subset of these genes, involved in proliferation and tumor cell migration like REPIN1/AP4, ST3GAL6, TRNAU1AP and PFKM are degraded upon loss of PRMT5. In summary, we have identified a novel mechanism through which the PRMT5/WDR77 complex maintains the balance between splicing and mRNA stability through methylation of ZNF326.


Subject(s)
Alternative Splicing , Carrier Proteins/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Transcription Factors/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cell Line , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , MCF-7 Cells , Protein Binding , Protein-Arginine N-Methyltransferases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry , Transcription Factors/genetics
14.
Curr Opin Genet Dev ; 46: 77-82, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28683341

ABSTRACT

Chemical modifications of RNA provide a direct and rapid way to manipulate the existing transcriptome, allowing rapid responses to the changing environment further enriching the regulatory capacity of RNA. N6-Methyladenosine (m6A) has been identified as the most abundant internal modification of messenger RNA in eukaryotes, linking external stimuli to an intricate network of transcriptional, post-transcriptional and translational processes. M6A modification affects a broad spectrum of cellular functions, including maintenance of the pluripotency of embryonic stem cells (ESCs) and the reprogramming of somatic cells into induced pluripotent stem cells (iPSCs). In this review, we summarize the most recent findings on m6A modification with special focus on the different studies describing how m6A is implicated in ESC self-renewal, cell fate specification and iPSC generation.


Subject(s)
Adenosine/analogs & derivatives , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Pluripotent Stem Cells , Adenosine/genetics , Gene Expression Regulation, Developmental , RNA, Messenger/genetics , Transcriptome/genetics
15.
Stem Cell Reports ; 9(1): 92-107, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28579396

ABSTRACT

THAP1 (THAP [Thanatos-associated protein] domain-containing, apoptosis-associated protein 1) is a ubiquitously expressed member of a family of transcription factors with highly conserved DNA-binding and protein-interacting regions. Mutations in THAP1 cause dystonia, DYT6, a neurologic movement disorder. THAP1 downstream targets and the mechanism via which it causes dystonia are largely unknown. Here, we show that wild-type THAP1 regulates embryonic stem cell (ESC) potential, survival, and proliferation. Our findings identify THAP1 as an essential factor underlying mouse ESC survival and to some extent, differentiation, particularly neuroectodermal. Loss of THAP1 or replacement with a disease-causing mutation results in an enhanced rate of cell death, prolongs Nanog, Prdm14, and/or Rex1 expression upon differentiation, and results in failure to upregulate ectodermal genes. ChIP-Seq reveals that these activities are likely due in part to indirect regulation of gene expression.


Subject(s)
Cell Differentiation , Cell Survival , DNA-Binding Proteins/metabolism , Mouse Embryonic Stem Cells/cytology , Animals , Apoptosis , Cell Line , Cell Proliferation , DNA-Binding Proteins/genetics , Dystonia/genetics , Dystonia/metabolism , Gene Expression Regulation, Developmental , Mice , Mouse Embryonic Stem Cells/metabolism , Mutation
16.
Cancer Res ; 76(19): 5615-5627, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27520449

ABSTRACT

Cancer-initiating cells (CIC) undergo asymmetric growth patterns that increase phenotypic diversity and drive selection for chemotherapeutic resistance and tumor relapse. WNT signaling is a hallmark of colon CIC, often caused by APC mutations, which enable activation of ß-catenin and MYC Accumulating evidence indicates that long noncoding RNAs (lncRNA) contribute to the stem-like character of colon cancer cells. In this study, we report enrichment of the lncRNA RBM5-AS1/LUST during sphere formation of colon CIC. Its silencing impaired WNT signaling, whereas its overexpression enforced WNT signaling, cell growth, and survival in serum-free media. RBM5-AS1 has been little characterized previously, and we determined it to be a nuclear-retained transcript that selectively interacted with ß-catenin. Mechanistic investigations showed that silencing or overexpression of RBM5-AS1 caused a respective loss or retention of ß-catenin from TCF4 complexes bound to the WNT target genes SGK1, YAP1, and MYC Our work suggests that RBM5-AS1 activity is critical for the functional enablement of colon cancer stem-like cells. Furthermore, it defines the mechanism of action of RBM5-AS1 in the WNT pathway via physical interactions with ß-catenin, helping organize transcriptional complexes that sustain colon CIC function. Cancer Res; 76(19); 5615-27. ©2016 AACR.


Subject(s)
Cell Cycle Proteins/genetics , Colonic Neoplasms/pathology , DNA-Binding Proteins/genetics , Neoplastic Stem Cells/physiology , RNA, Antisense/physiology , RNA, Long Noncoding/physiology , RNA-Binding Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , CD24 Antigen/genetics , Cell Line, Tumor , Cyclin D1/genetics , Genes, myc , Humans , Hyaluronan Receptors/genetics , Immediate-Early Proteins/genetics , Mice , Protein Serine-Threonine Kinases/genetics , Transcription Factor 4 , Transcription Factors/physiology , Wnt Signaling Pathway , beta Catenin/physiology
17.
Trends Biochem Sci ; 41(12): 986-988, 2016 12.
Article in English | MEDLINE | ID: mdl-27519282
18.
ACS Med Chem Lett ; 7(6): 601-5, 2016 Jun 09.
Article in English | MEDLINE | ID: mdl-27326334

ABSTRACT

The chromobox 7 (CBX7) protein of the polycomb repressive complex 1 (PRC1) functions to repress transcription of tumor suppressor p16 (INK4a) through long noncoding RNA, ANRIL (antisense noncoding RNA in the INK4 locus) directed chromodomain (ChD) binding to trimethylated lysine 27 of histone H3 (H3K27me3), resulting in chromatin compaction at the INK4a/ARF locus. In this study, we report structure-guided discovery of two distinct classes of small-molecule antagonists for the CBX7ChD. Our Class A compounds, a series including analogues of the previously reported MS452, inhibit CBX7ChD/methyl-lysine binding by occupying the H3K27me3 peptide binding site, whereas our Class B compound, the newly discovered MS351, appears to inhibit H3K27me3 binding when CBX7ChD is bound to RNA. Our crystal structure of the CBX7ChD/MS351 complex reveals the molecular details of ligand recognition by the aromatic cage residues that typically engage in methyl-lysine binding. We further demonstrate that MS351 effectively induces transcriptional derepression of CBX7 target genes, including p16 (INK4a) in mouse embryonic stem cells and human prostate cancer PC3 cells. Thus, MS351 represents a new class of ChD antagonists that selectively targets the biologically active form of CBX7 of the PRC1 in long noncoding RNA- and H3K27me3-directed gene transcriptional repression.

19.
Cell Rep ; 14(3): 479-492, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26774474

ABSTRACT

The Peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) is a transcriptional co-activator that plays a central role in adapted metabolic responses. PGC-1α is dynamically methylated and unmethylated at the residue K779 by the methyltransferase SET7/9 and the Lysine Specific Demethylase 1A (LSD1), respectively. Interactions of methylated PGC-1α[K779me] with the Spt-Ada-Gcn5-acetyltransferase (SAGA) complex, the Mediator members MED1 and MED17, and the NOP2/Sun RNA methytransferase 7 (NSUN7) reinforce transcription, and are concomitant with the m(5)C mark on enhancer RNAs (eRNAs). Consistently, loss of Set7/9 and NSun7 in liver cell model systems resulted in depletion of the PGC-1α target genes Pfkl, Sirt5, Idh3b, and Hmox2, which was accompanied by a decrease in the eRNAs levels associated with these loci. Enrichment of m(5)C within eRNA species coincides with metabolic stress of fasting in vivo. Collectively, these findings illustrate the complex epigenetic circuitry imposed by PGC-1α at the eRNA level to fine-tune energy metabolism.


Subject(s)
5-Methylcytosine/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Cell Line , Enhancer Elements, Genetic , HEK293 Cells , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Methyltransferases/antagonists & inhibitors , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , NIH 3T3 Cells , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphofructokinase-1/genetics , Phosphofructokinase-1/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sirtuins/genetics , Sirtuins/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
20.
Curr Top Microbiol Immunol ; 394: 29-39, 2016.
Article in English | MEDLINE | ID: mdl-26220772

ABSTRACT

The long non-coding RNA CDKN2B-AS1, commonly referred to as the A ntisense N on-coding R NA in the I NK4 L ocus (ANRIL), is a 3.8-kb-long RNA transcribed from the short arm of human chromosome 9 on p21.3 that overlaps a critical region encompassing three major tumor suppressor loci juxtaposed to the INK4b-ARF-INK4a gene cluster and the methyl-thioadenosine phosphorylase (MTAP) gene. Genome-wide association studies have identified this region with a remarkable and growing number of disease-associated DNA alterations and single nucleotide polymorphisms, which corresponds to increased susceptibility to human disease. Recent attention has been devoted on whether these alterations in the ANRIL sequence affect its expression levels and/or its splicing transcript variation, and in consequence, global cellular homeostasis. Moreover, recent evidence postulates that ANRIL not only can regulate their immediate genomic neighbors in cis, but also has the capacity to regulate additional loci in trans. This action would further increase the complexity for mechanisms imposed through ANRIL and furthering the scope of this lncRNA in disease pathogenesis. In this chapter, we summarize the most recent findings on the investigation of ANRIL and provide a perspective on the biological and clinical significance of ANRIL as a putative biomarker, specifically, its potential role in directing cellular fates leading to cancer and cardiovascular disease.


Subject(s)
Cardiovascular Diseases/etiology , Neoplasms/etiology , Polycomb-Group Proteins/physiology , RNA, Long Noncoding/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...