scispace - formally typeset
Search or ask a question

Showing papers on "MRNA modification published in 2017"


Journal ArticleDOI
15 Jun 2017-Cell
TL;DR: Roles for mRNA modification in nearly every aspect of the mRNA life cycle, as well as in various cellular, developmental, and disease processes are revealed.

1,855 citations


Journal ArticleDOI
TL;DR: N6-adenosine methylation directs mRNAs to distinct fates by grouping them for differential processing, translation and decay in processes such as cell differentiation, embryonic development and stress responses.
Abstract: The recent discovery of reversible mRNA methylation has opened a new realm of post-transcriptional gene regulation in eukaryotes. The identification and functional characterization of proteins that specifically recognize RNA N6-methyladenosine (m6A) unveiled it as a modification that cells utilize to accelerate mRNA metabolism and translation. N6-adenosine methylation directs mRNAs to distinct fates by grouping them for differential processing, translation and decay in processes such as cell differentiation, embryonic development and stress responses. Other mRNA modifications, including N1-methyladenosine (m1A), 5-methylcytosine (m5C) and pseudouridine, together with m6A form the epitranscriptome and collectively code a new layer of information that controls protein synthesis.

1,369 citations



Journal ArticleDOI
TL;DR: M6A sequencing reveals that knockdown of METTL3 or METTL14 induced changes in mRNA m6A enrichment and altered mRNA expression of genes with critical biological functions in GSCs, identified as promising therapeutic targets for glioblastoma.

865 citations


Journal ArticleDOI
TL;DR: It is shown that shRNA-mediated depletion of the m6A-forming enzyme METTL3 in human hematopoietic stem/progenitor cells (HSPCs) promotes cell differentiation, coupled with reduced cell proliferation, and this results provide a rationale for the therapeutic targeting of MET TL3 in myeloid leukemia.
Abstract: N6-methyladenosine (m6A) is an abundant nucleotide modification in mRNA that is required for the differentiation of mouse embryonic stem cells. However, it remains unknown whether the m6A modification controls the differentiation of normal and/or malignant myeloid hematopoietic cells. Here we show that shRNA-mediated depletion of the m6A-forming enzyme METTL3 in human hematopoietic stem/progenitor cells (HSPCs) promotes cell differentiation, coupled with reduced cell proliferation. Conversely, overexpression of wild-type METTL3, but not of a catalytically inactive form of METTL3, inhibits cell differentiation and increases cell growth. METTL3 mRNA and protein are expressed more abundantly in acute myeloid leukemia (AML) cells than in healthy HSPCs or other types of tumor cells. Furthermore, METTL3 depletion in human myeloid leukemia cell lines induces cell differentiation and apoptosis and delays leukemia progression in recipient mice in vivo. Single-nucleotide-resolution mapping of m6A coupled with ribosome profiling reveals that m6A promotes the translation of c-MYC, BCL2 and PTEN mRNAs in the human acute myeloid leukemia MOLM-13 cell line. Moreover, loss of METTL3 leads to increased levels of phosphorylated AKT, which contributes to the differentiation-promoting effects of METTL3 depletion. Overall, these results provide a rationale for the therapeutic targeting of METTL3 in myeloid leukemia.

804 citations


Journal ArticleDOI
19 Jan 2017-Nature
TL;DR: Using a transcriptome-wide map of m6Am, it is found that m 6Am-initiated transcripts are markedly more stable than mRNAs that begin with other nucleotides and that m6 am is selectively demethylated by fat mass and obesity-associated protein (FTO).
Abstract: Internal bases in mRNA can be subjected to modifications that influence the fate of mRNA in cells. One of the most prevalent modified bases is found at the 5' end of mRNA, at the first encoded nucleotide adjacent to the 7-methylguanosine cap. Here we show that this nucleotide, N6,2'-O-dimethyladenosine (m6Am), is a reversible modification that influences cellular mRNA fate. Using a transcriptome-wide map of m6Am we find that m6Am-initiated transcripts are markedly more stable than mRNAs that begin with other nucleotides. We show that the enhanced stability of m6Am-initiated transcripts is due to resistance to the mRNA-decapping enzyme DCP2. Moreover, we find that m6Am is selectively demethylated by fat mass and obesity-associated protein (FTO). FTO preferentially demethylates m6Am rather than N6-methyladenosine (m6A), and reduces the stability of m6Am mRNAs. Together, these findings show that the methylation status of m6Am in the 5' cap is a dynamic and reversible epitranscriptomic modification that determines mRNA stability.

729 citations


Journal ArticleDOI
06 Oct 2017-eLife
TL;DR: It is shown that the m6A-binding protein YTHDC1 mediates export of methylated mRNA from the nucleus to the cytoplasm in HeLa cells, and supports an emerging paradigm of m 6A as a distinct biochemical entity for selective processing and metabolism of mammalian mRNAs.
Abstract: N6-methyladenosine (m6A) is the most abundant internal modification of eukaryotic messenger RNA (mRNA) and plays critical roles in RNA biology. The function of this modification is mediated by m6A-selective 'reader' proteins of the YTH family, which incorporate m6A-modified mRNAs into pathways of RNA metabolism. Here, we show that the m6A-binding protein YTHDC1 mediates export of methylated mRNA from the nucleus to the cytoplasm in HeLa cells. Knockdown of YTHDC1 results in an extended residence time for nuclear m6A-containing mRNA, with an accumulation of transcripts in the nucleus and accompanying depletion within the cytoplasm. YTHDC1 interacts with the splicing factor and nuclear export adaptor protein SRSF3, and facilitates RNA binding to both SRSF3 and NXF1. This role for YTHDC1 expands the potential utility of chemical modification of mRNA, and supports an emerging paradigm of m6A as a distinct biochemical entity for selective processing and metabolism of mammalian mRNAs.

726 citations


Journal ArticleDOI
07 Dec 2017-Nature
TL;DR: Together, these data define METTL3 as a regulator of a chromatin-based pathway that is necessary for maintenance of the leukaemic state and identify this enzyme as a potential therapeutic target for acute myeloid leukaemia.
Abstract: N6-methyladenosine (m6A) is an abundant internal RNA modification in both coding and non-coding RNAs that is catalysed by the METTL3-METTL14 methyltransferase complex. However, the specific role of these enzymes in cancer is still largely unknown. Here we define a pathway that is specific for METTL3 and is implicated in the maintenance of a leukaemic state. We identify METTL3 as an essential gene for growth of acute myeloid leukaemia cells in two distinct genetic screens. Downregulation of METTL3 results in cell cycle arrest, differentiation of leukaemic cells and failure to establish leukaemia in immunodeficient mice. We show that METTL3, independently of METTL14, associates with chromatin and localizes to the transcriptional start sites of active genes. The vast majority of these genes have the CAATT-box binding protein CEBPZ present at the transcriptional start site, and this is required for recruitment of METTL3 to chromatin. Promoter-bound METTL3 induces m6A modification within the coding region of the associated mRNA transcript, and enhances its translation by relieving ribosome stalling. We show that genes regulated by METTL3 in this way are necessary for acute myeloid leukaemia. Together, these data define METTL3 as a regulator of a chromatin-based pathway that is necessary for maintenance of the leukaemic state and identify this enzyme as a potential therapeutic target for acute myeloid leukaemia.

705 citations



Journal ArticleDOI
TL;DR: YTHDC2 is an m6A-binding protein that plays critical roles during spermatogenesis and enhances the translation efficiency of its targets and also decreases their mRNA abundance.
Abstract: N6-methyladenosine (m6A) is the most common internal modification in eukaryotic mRNA. It is dynamically installed and removed, and acts as a new layer of mRNA metabolism, regulating biological processes including stem cell pluripotency, cell differentiation, and energy homeostasis. m6A is recognized by selective binding proteins; YTHDF1 and YTHDF3 work in concert to affect the translation of m6A-containing mRNAs, YTHDF2 expedites mRNA decay, and YTHDC1 affects the nuclear processing of its targets. The biological function of YTHDC2, the final member of the YTH protein family, remains unknown. We report that YTHDC2 selectively binds m6A at its consensus motif. YTHDC2 enhances the translation efficiency of its targets and also decreases their mRNA abundance. Ythdc2 knockout mice are infertile; males have significantly smaller testes and females have significantly smaller ovaries compared to those of littermates. The germ cells of Ythdc2 knockout mice do not develop past the zygotene stage and accordingly, Ythdc2 is upregulated in the testes as meiosis begins. Thus, YTHDC2 is an m6A-binding protein that plays critical roles during spermatogenesis.

611 citations


Journal ArticleDOI
TL;DR: These studies reveal an important role of METTL14 in tumor metastasis and provide a fresh view on m6A modification in tumor progression and reveal an adverse prognosis factor for recurrence‐free survival of hepatocellular carcinoma.


Journal ArticleDOI
02 Nov 2017-Cell
TL;DR: It is shown that m6A depletion by Mettl14 knockout in embryonic mouse brains prolongs the cell cycle of radial glia cells and extends cortical neurogenesis into postnatal stages and uncovers previously unappreciated transcriptional prepatterning in cortical neural stem cells.

Journal ArticleDOI
TL;DR: Comprehensive and systematic mass-spectrometry-based screening of m6A interactors in various cell types and sequence contexts identifies G3BP1 as a protein that is repelled by m 6A and positively regulates mRNA stability in an m6a-regulated manner, thus revealing a connection between an mRNA modification and an autism spectrum disorder.
Abstract: RNA modifications are integral to the regulation of RNA metabolism. One abundant mRNA modification is N6-methyladenosine (m6A), which affects various aspects of RNA metabolism, including splicing, translation and degradation. Current knowledge about the proteins recruited to m6A to carry out these molecular processes is still limited. Here we describe comprehensive and systematic mass-spectrometry-based screening of m6A interactors in various cell types and sequence contexts. Among the main findings, we identified G3BP1 as a protein that is repelled by m6A and positively regulates mRNA stability in an m6A-regulated manner. Furthermore, we identified FMR1 as a sequence-context-dependent m6A reader, thus revealing a connection between an mRNA modification and an autism spectrum disorder. Collectively, our data represent a rich resource and shed further light on the complex interplay among m6A, m6A interactors and mRNA homeostasis.

Journal ArticleDOI
TL;DR: A base-resolution m1A profiling method is developed, based onm1A-induced misincorporation during reverse transcription, and distinct classes of m1 a methylome are revealed in the human transcriptome, providing a resource for functional studies of m 1A-mediated epitranscriptomic regulation.

Journal ArticleDOI
TL;DR: It is argued that m6A provides a fast mean to post-transcriptionally maximize gene expression, and appears to have a second function during developmental transitions by targeting m 6A-marked transcripts for degradation.

Journal ArticleDOI
TL;DR: The results suggest that m5C in mRNA is a new epitranscriptome marker in Arabidopsis, and that regulation of this modification is an integral part of gene regulatory networks underlying plant development.

Journal ArticleDOI
TL;DR: The results show the importance of FTO during memory formation and implicate mRNA modification and epi-transcriptomics as novel regulators of memory formation, and implicates CRISPR/Cas9 or shRNA targeted against Fto.

Journal ArticleDOI
TL;DR: This review summarizes the key methods and the recent discoveries in the field of epitranscriptomics through the prism of post-transcriptional mRNA methylation in eukaryotes.

Journal ArticleDOI
13 Jun 2017-RNA
TL;DR: The results suggest that an increase in m6A mRNA during hypoxic exposure leads to post-transcriptional stabilization of specific mRNAs and contributes to the recovery of translational efficiency after hypoxic stress.
Abstract: Post-transcriptional regulation of mRNA during oxygen deprivation, or hypoxia, can affect the survivability of cells. Hypoxia has been shown to increase stability of a subset of ischemia-related mRNAs, including VEGF. RNA binding proteins and miRNAs have been identified as important for post-transcriptional regulation of individual mRNAs, but corresponding mechanisms that regulate global stability are not well understood. Recently, mRNA modification by N6-methyladenosine (m6A) has been shown to be involved in post-transcriptional regulation processes including mRNA stability and promotion of translation, but the role of m6A in the hypoxia response is unknown. In this study, we investigate the effect of hypoxia on RNA modifications including m6A. Our results show hypoxia increases m6A content of poly(A)+ messenger RNA (mRNA), but not in total or ribosomal RNA in HEK293T cells. Using m6A mRNA immunoprecipitation, we identify specific hypoxia-modified mRNAs, including glucose transporter 1 (Glut1) and c-Myc, which show increased m6A levels under hypoxic conditions. Many of these mRNAs also exhibit increased stability, which was blocked by knockdown of m6A-specific methyltransferases METTL3/14. However, the increase in mRNA stability did not correlate with a change in translational efficiency or the steady-state amount of their proteins. Knockdown of METTL3/14 did reveal that m6A is involved in recovery of translational efficiency after hypoxic stress. Therefore, our results suggest that an increase in m6A mRNA during hypoxic exposure leads to post-transcriptional stabilization of specific mRNAs and contributes to the recovery of translational efficiency after hypoxic stress.

Journal ArticleDOI
TL;DR: Structural and functional analysis show that 2′-O methylation at N2, another abundant mRNA modification, is also detrimental for RNA binding, thus revealing a potentially synergistic role for it in self- versus nonself-mRNA discernment, and sheds new light on the molecular basis for IFIT1 translational inhibition of capped viral RNA.
Abstract: IFIT1 (IFN-induced protein with tetratricopeptide repeats-1) is an effector of the host innate immune antiviral response that prevents propagation of virus infection by selectively inhibiting translation of viral mRNA. It relies on its ability to compete with the translation initiation factor eIF4F to specifically recognize foreign capped mRNAs, while remaining inactive against host mRNAs marked by ribose 2′-O methylation at the first cap-proximal nucleotide (N1). We report here several crystal structures of RNA-bound human IFIT1, including a 1.6-A complex with capped RNA. IFIT1 forms a water-filled, positively charged RNA-binding tunnel with a separate hydrophobic extension that unexpectedly engages the cap in multiple conformations ( syn and anti ) giving rise to a relatively plastic and nonspecific mode of binding, in stark contrast to eIF4E. Cap-proximal nucleotides encircled by the tunnel provide affinity to compete with eIF4F while allowing IFIT1 to select against N1 methylated mRNA. Gel-shift binding assays confirm that N1 methylation interferes with IFIT1 binding, but in an RNA-dependent manner, whereas translation assays reveal that N1 methylation alone is not sufficient to prevent mRNA recognition at high IFIT1 concentrations. Structural and functional analysis show that 2′-O methylation at N2, another abundant mRNA modification, is also detrimental for RNA binding, thus revealing a potentially synergistic role for it in self- versus nonself-mRNA discernment. Finally, structure-guided mutational analysis confirms the importance of RNA binding for IFIT1 restriction of a human coronavirus mutant lacking viral N1 methylation. Our structural and biochemical analysis sheds new light on the molecular basis for IFIT1 translational inhibition of capped viral RNA.

Journal ArticleDOI
TL;DR: The functions and mechanisms of the main m6A players are reviewed with a particular focus on Drosophila melanogaster, which has shown to influence many physiological processes from yeast to mammals.
Abstract: RNA modifications are an emerging layer of posttranscriptional gene regulation in eukaryotes. N6-methyladenosine (m6A) is among the most abundant modifications in mRNAs (mRNAs) that was shown to influence many physiological processes from yeast to mammals. Like DNA methylation, m6A in mRNA is dynamically regulated. A conserved methyltransferase complex catalyzes the deposition of the methyl group on adenosine, which can be removed by specific classes of demethylases. Furthermore, YTH-domain containing proteins can recognize this modification to mediate m6A-dependent activities. Here we review the functions and mechanisms of the main m6A players with a particular focus on Drosophila melanogaster.

Journal ArticleDOI
TL;DR: AlKBH5, a demethylase of the mRNA modification N6-methyladenosine, regulates proliferation and self-renewal of glioblastoma stem-like cells by modulating pre-mRNA stability and expression of the FOXM1 gene.

Journal ArticleDOI
TL;DR: Crystallography reveals that N6‐methylation of adenine prevents the formation of trans Hoogsteen‐sugar A·G base pairs, explaining why the box C/D RNA cannot adopt its kinked conformation, and data indicate that sheared A·g base pairs are more susceptible to disruption by N6mA methylation and are therefore possible regulatory sites.
Abstract: N6-methyladenine is the most widespread mRNA modification. A subset of human box C/D snoRNA species have target GAC sequences that lead to formation of N6-methyladenine at a key trans Hoogsteen-sugar A·G base pair, of which half are methylated in vivo The GAC target is conserved only in those that are methylated. Methylation prevents binding of the 15.5-kDa protein and the induced folding of the RNA Thus, the assembly of the box C/D snoRNP could in principle be regulated by RNA methylation at its critical first stage. Crystallography reveals that N6-methylation of adenine prevents the formation of trans Hoogsteen-sugar A·G base pairs, explaining why the box C/D RNA cannot adopt its kinked conformation. More generally, our data indicate that sheared A·G base pairs (but not Watson-Crick base pairs) are more susceptible to disruption by N6mA methylation and are therefore possible regulatory sites. The human signal recognition particle RNA and many related Alu retrotransposon RNA species are also methylated at N6 of an adenine that forms a sheared base pair with guanine and mediates a key tertiary interaction.

Journal ArticleDOI
TL;DR: The unique topology of this mRNA modification is explored, its writer is identified and its involvement in nuclear-cytoplasmic shuttling mediated by a specific reader is demonstrated.
Abstract: 5-methylcytosine was shown before to be an epitranscriptomic mark Yang et al now explored the unique topology of this mRNA modification, identified its writer and demonstrated its involvement in nuclear-cytoplasmic shuttling mediated by a specific reader

Journal ArticleDOI
TL;DR: The review focuses on recent progress in mRNA modification and stem cell biology and an integrated landscape of m6A enrichments in both human and mouse embryonic stem cells (ESCs) is presented using publically available datasets.
Abstract: Epitranscriptome refers to any relevant changes in gene expression without changes in RNA sequences. Similar to epigenetic changes, the epitranscriptomic changes are in general mediated by post-transcriptional chemical modifications of RNA species. Recently, mRNA modifications, especially both N6-methyladenosine (m6A) and N1-methyladenosine (m1A), have received significant attention as proteins responsible for generating, removing or recognizing m6A modification have been identified. m6A in eukaryotic cells including human and mouse was initially identified in early 1970s. However, the function of the modification has not been intensively studied because of technical limitations. Recently, using next-generation sequencing (NGS) technology, several groups revealed transcriptome-wide distribution of m6A and its both in vitro and in vivo roles in biological processes such as metabolism and development. The review focuses on recent progress in mRNA modification and stem cell biology. In addition, an integrated landscape of m6A enrichments in both human and mouse embryonic stem cells (ESCs) is presented using publically available datasets.

Journal ArticleDOI
TL;DR: Resolving the dynamic function of ribonucleoprotein particles in splicing, APA, or RNA modification will enable us to understand their contribution to pathological conditions.

Posted ContentDOI
13 Oct 2017-bioRxiv
TL;DR: A base-resolution m1A profiling method is developed, based onm1A-induced misincorporation during reverse transcription, and distinct classes of m1 a methylome are revealed in the human transcriptome, providing a resource for functional studies of m 1A-mediated epitranscriptomic regulation.
Abstract: Gene expression can be post-transcriptionally regulated via dynamic and reversible RNA modifications. N1-methyladenosine (m1A) is a recently identified mRNA modification; however, little is known about its precise location, regulation and function. Here, we develop a base-resolution m1A profiling method, based on m1A-induced misincorporation during reverse transcription, and report distinct classes of m1A methylome in the human transcriptome. m1A in 59-UTR, particularly those at the first nucleotide of mRNA, associate with increased translation efficiency. A different subset of m1A exhibit a GUUCRA tRNA-like motif, are evenly distributed in the transcriptome and are dependent on the methyltransferase TRMT6/61A. Additionally, we show for the first time that m1A is prevalent in the mitochondrial-encoded transcripts. Manipulation of m1A level via TRMT61B, a mitochondria-localizing m1A methyltransferase, demonstrates that m1A in mitochondrial mRNA interferes with translation. Collectively, our approaches reveal distinct classes of m1A methylome and provide a resource for functional studies of m1A-mediated epitranscriptomic regulation.

29 Nov 2017
TL;DR: In this paper, the effect of hypoxia on RNA m6A content was investigated and it was shown that hypoxic exposure leads to striking changes in the m6a content of mRNA in HEK-293T cells as well as immortalized and oncogenically transformed human mammary epithelial cells (HMECs).
Abstract: Cancer is a prevalent disease that affects millions of people each year across the globe. In an effort to find therapies to the many types of cancers , regulation of protein expression through transcription and translation pathways have been extensively studied. However , one area which has been often overlooked is the importance of post-transcriptional regulation in protein output. The goal of this dissertation project has been to understand post-transcriptional regulation of mRNA during a cellular stress that all cancers must overcome in order to survive , oxygen deprivation. The lack of oxygen , termed hypoxia , is known to affect tumor growth and angiogenesis. Specifically , hypoxia affects the post-transcriptional regulation of mRNAs by increasing the stability of a subset of ischemia-related mRNAs , including VEGF. Multiple factors including RNA binding proteins and miRNAs have been identified to be important for the post-transcriptional regulation of individual mRNAs , but mechanisms regulating global stability have not been elucidated. Recently , the mRNA modification , N6 methyladenosine (m6A) , has been shown to be involved in the post-transcriptional regulation processes of mRNA stability and promotion of translation. Therefore , I set out to investigate the effect of hypoxia on RNA m6A content. My results show that hypoxic exposure leads to striking changes in the m6A content of mRNA in HEK-293T cells as well as immortalized and oncogenically transformed human mammary epithelial cells (HMECs). Using m6A mRNA immunoprecipitation , we identified a number of specific hypoxia related mRNAs , including Glut1 and c-Myc , which show increased m6A levels under hypoxic conditions. Many of these same mRNAs also exhibit increased mRNA stability revealed by metabolic labeling of RNA using 4sU. Furthermore , knockdown of the m6A-specific methyltransferases METTL3/14 blocked the hypoxic stabilization of these mRNA. The increase in mRNA stability through m6A led to greater translational efficiency after recovery from the hypoxic stress. Overexpressing m6A in oncogenically transformed HMEC in normal oxygen conditions led to an increase in would healing , proliferation , and invasion abilities. Ultimately , the mRNA modification , m6A , led to phenotypic changes in a cancer cell , and it may be possible to manipulate this mRNA modification in order to slow cancer growth.

Journal ArticleDOI
TL;DR: This week’s articles highlight the immune response to intestinal helminth parasites and an mRNA modification that determines cell fate.
Abstract: This week’s articles highlight the immune response to intestinal helminth parasites and an mRNA modification that determines cell fate.