Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cell ; 171(4): 877-889.e17, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-28965759

ABSTRACT

N6-methyladenosine (m6A), installed by the Mettl3/Mettl14 methyltransferase complex, is the most prevalent internal mRNA modification. Whether m6A regulates mammalian brain development is unknown. Here, we show 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. m6A depletion by Mettl3 knockdown also leads to a prolonged cell cycle and maintenance of radial glia cells. m6A sequencing of embryonic mouse cortex reveals enrichment of mRNAs related to transcription factors, neurogenesis, the cell cycle, and neuronal differentiation, and m6A tagging promotes their decay. Further analysis uncovers previously unappreciated transcriptional prepatterning in cortical neural stem cells. m6A signaling also regulates human cortical neurogenesis in forebrain organoids. Comparison of m6A-mRNA landscapes between mouse and human cortical neurogenesis reveals enrichment of human-specific m6A tagging of transcripts related to brain-disorder risk genes. Our study identifies an epitranscriptomic mechanism in heightened transcriptional coordination during mammalian cortical neurogenesis.


Subject(s)
Neurogenesis , Prosencephalon/embryology , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Animals , Cell Cycle , Gene Expression Regulation , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Humans , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Knockout , Neural Stem Cells/metabolism , Organoids/metabolism , Prosencephalon/cytology , Prosencephalon/metabolism , RNA Stability
2.
Cell ; 161(4): 879-892, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25936837

ABSTRACT

N(6)-methyldeoxyadenosine (6mA or m(6)A) is a DNA modification preserved in prokaryotes to eukaryotes. It is widespread in bacteria and functions in DNA mismatch repair, chromosome segregation, and virulence regulation. In contrast, the distribution and function of 6mA in eukaryotes have been unclear. Here, we present a comprehensive analysis of the 6mA landscape in the genome of Chlamydomonas using new sequencing approaches. We identified the 6mA modification in 84% of genes in Chlamydomonas. We found that 6mA mainly locates at ApT dinucleotides around transcription start sites (TSS) with a bimodal distribution and appears to mark active genes. A periodic pattern of 6mA deposition was also observed at base resolution, which is associated with nucleosome distribution near the TSS, suggesting a possible role in nucleosome positioning. The new genome-wide mapping of 6mA and its unique distribution in the Chlamydomonas genome suggest potential regulatory roles of 6mA in gene expression in eukaryotic organisms.


Subject(s)
Adenine/analogs & derivatives , Chlamydomonas reinhardtii/genetics , Transcription Initiation Site , 5-Methylcytosine/metabolism , Adenine/metabolism , Chlamydomonas reinhardtii/metabolism , DNA, Algal/metabolism , Gene Expression Regulation , Genome-Wide Association Study , Nucleosomes/metabolism , Transcription, Genetic
3.
Nature ; 530(7591): 441-6, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26863196

ABSTRACT

Gene expression can be regulated post-transcriptionally through dynamic and reversible RNA modifications. A recent noteworthy example is N(6)-methyladenosine (m(6)A), which affects messenger RNA (mRNA) localization, stability, translation and splicing. Here we report on a new mRNA modification, N(1)-methyladenosine (m(1)A), that occurs on thousands of different gene transcripts in eukaryotic cells, from yeast to mammals, at an estimated average transcript stoichiometry of 20% in humans. Employing newly developed sequencing approaches, we show that m(1)A is enriched around the start codon upstream of the first splice site: it preferentially decorates more structured regions around canonical and alternative translation initiation sites, is dynamic in response to physiological conditions, and correlates positively with protein production. These unique features are highly conserved in mouse and human cells, strongly indicating a functional role for m(1)A in promoting translation of methylated mRNA.


Subject(s)
Adenosine/analogs & derivatives , RNA, Messenger/metabolism , 5' Untranslated Regions/genetics , Adenosine/metabolism , Animals , Base Sequence , Cell Line , Cell Line, Tumor , Codon, Initiator/genetics , Conserved Sequence , Epigenesis, Genetic , Evolution, Molecular , GC Rich Sequence/genetics , Humans , Methylation , Mice , Organ Specificity , Peptide Chain Initiation, Translational/genetics , RNA Splice Sites/genetics , RNA, Messenger/genetics , Saccharomyces cerevisiae , Transcriptome/genetics
4.
Mol Cell ; 47(4): 608-21, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22771118

ABSTRACT

GATA-1 and its cofactor FOG-1 are required for the differentiation of erythrocytes and megakaryocytes. In contrast, mast cell development requires GATA-1 and the absence of FOG-1. Through genome-wide comparison of the chromatin occupancy of GATA-1 and a naturally occurring mutant that cannot bind FOG-1 (GATA-1(V205G)), we reveal that FOG-1 intricately regulates the chromatin occupancy of GATA-1. We identified GATA1-selective and GATA-1(V205G)-selective binding sites and show that GATA-1, in the absence of FOG-1, occupies GATA-1(V205G)-selective sites, but not GATA1-selective sites. By integrating ChIP-seq and gene expression data, we discovered that GATA-1(V205G) binds and activates mast cell-specific genes via GATA-1(V205G)-selective sites. We further show that exogenous expression of FOG-1 in mast cells leads to displacement of GATA-1 from mast cell-specific genes and causes their downregulation. Together these findings establish a mechanism of gene regulation whereby a non-DNA binding cofactor directly modulates the occupancy of a transcription factor to control lineage specification.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Expression Regulation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Amino Acid Motifs , Animals , Binding Sites , Cells, Cultured , Down-Regulation , Gene Expression Regulation/genetics , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mutation , Protein Binding , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
5.
Genes Dev ; 24(15): 1620-33, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20679398

ABSTRACT

The bicistronic microRNA (miRNA) locus miR-144/451 is highly expressed during erythrocyte development, although its physiological roles are poorly understood. We show that miR-144/451 ablation in mice causes mild erythrocyte instability and increased susceptibility to damage after exposure to oxidant drugs. This phenotype is deeply conserved, as miR-451 depletion synergizes with oxidant stress to cause profound anemia in zebrafish embryos. At least some protective activities of miR-451 stem from its ability to directly suppress production of 14-3-3zeta, a phospho-serine/threonine-binding protein that inhibits nuclear accumulation of transcription factor FoxO3, a positive regulator of erythroid anti-oxidant genes. Thus, in miR-144/451(-/-) erythroblasts, 14-3-3zeta accumulates, causing partial relocalization of FoxO3 from nucleus to cytoplasm with dampening of its transcriptional program, including anti-oxidant-encoding genes Cat and Gpx1. Supporting this mechanism, overexpression of 14-3-3zeta in erythroid cells and fibroblasts inhibits nuclear localization and activity of FoxO3. Moreover, shRNA suppression of 14-3-3zeta protects miR-144/451(-/-) erythrocytes against peroxide-induced destruction, and restores catalase activity. Our findings define a novel miRNA-regulated pathway that protects erythrocytes against oxidant stress, and, more generally, illustrate how a miRNA can influence gene expression by altering the activity of a key transcription factor.


Subject(s)
14-3-3 Proteins/metabolism , Erythroid Cells/metabolism , Gene Expression Regulation, Developmental , MicroRNAs/metabolism , Oxidative Stress , 14-3-3 Proteins/genetics , Active Transport, Cell Nucleus , Animals , Base Sequence , Catalase/metabolism , Erythroid Cells/enzymology , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Gene Knockdown Techniques , Mice , Mice, Knockout , MicroRNAs/genetics , Sequence Alignment , Sequence Deletion/genetics , Zebrafish/genetics , Zebrafish/metabolism
6.
Blood ; 122(14): e33-43, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23980066

ABSTRACT

Acute megakaryoblastic leukemia (AMKL) is more frequently observed in Down syndrome (DS) patients, in whom it is often preceded by a transient myeloproliferative disorder (TMD). The development of DS-TMD and DS-AMKL requires not only the presence of the trisomy 21 but also that of GATA1 mutations. Despite extensive studies into the genetics of DS-AMKL, the importance of epigenetic deregulation in this disease has been unexplored. We performed DNA methylation profiling at different stages of development of DS-AMKL and analyzed the dynamics of the epigenetic program. Early genome-wide DNA methylation changes can be detected in trisomy 21 fetal liver mononuclear cells, prior to the acquisition of GATA1 mutations. These early changes are characterized by marked loss of DNA methylation at genes associated with developmental disorders, including those affecting the cardiovascular, neurological, and endocrine systems. This is followed by a second wave of changes detected in DS-TMD and DS-AMKL, characterized by gains of methylation. This new wave of hypermethylation targets a distinct set of genes involved in hematopoiesis and regulation of cell growth and proliferation. These findings indicate that the final epigenetic landscape of DS-AMKL is the result of sequential and opposing changes in DNA methylation occurring at specific times in the disease development.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA Methylation/genetics , Down Syndrome/complications , Epigenesis, Genetic/genetics , Leukemia, Megakaryoblastic, Acute/genetics , Down Syndrome/genetics , Humans , Myeloproliferative Disorders/genetics
7.
Blood ; 121(13): 2440-51, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23335373

ABSTRACT

The transcription factor Ikaros regulates the development of hematopoietic cells. Ikaros-deficient animals fail to develop B cells and display a T-cell malignancy, which is correlated with altered Notch signaling. Recently, loss of Ikaros was associated with progression of myeloproliferative neoplasms to acute myeloid leukemia and increasing evidence shows that Ikaros is also critical for the regulation of myeloid development. Previous studies showed that Ikaros-deficient mice have increased megakaryopoiesis, but the molecular mechanism of this phenomenon remains unknown. Here, we show that Ikaros overexpression decreases NOTCH-induced megakaryocytic specification, and represses expression of several megakaryocytic genes including GATA-1 to block differentiation and terminal maturation. We also demonstrate that Ikaros expression is differentially regulated by GATA-2 and GATA-1 during megakaryocytic differentiation and reveal that the combined loss of Ikzf1 and Gata1 leads to synthetic lethality in vivo associated with prominent defects in erythroid cells and an expansion of megakaryocyte progenitors. Taken together, our observations demonstrate an important functional interplay between Ikaros, GATA factors, and the NOTCH signaling pathway in specification and homeostasis of the megakaryocyte lineage.


Subject(s)
GATA1 Transcription Factor/metabolism , Ikaros Transcription Factor/physiology , Receptors, Notch/metabolism , Thrombopoiesis/genetics , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Down-Regulation/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Megakaryocytes/metabolism , Megakaryocytes/physiology , Mice , Mice, Knockout , Models, Biological , Protein Binding/genetics , Protein Binding/physiology , Signal Transduction/genetics , Signal Transduction/physiology
8.
Blood ; 119(16): 3724-33, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22383799

ABSTRACT

There are many examples of transcription factor families whose members control gene expression profiles of diverse cell types. However, the mechanism by which closely related factors occupy distinct regulatory elements and impart lineage specificity is largely undefined. Here we demonstrate on a genome wide scale that the hematopoietic GATA factors GATA-1 and GATA-2 bind overlapping sets of genes, often at distinct sites, as a means to differentially regulate target gene expression and to regulate the balance between proliferation and differentiation. We also reveal that the GATA switch, which entails a chromatin occupancy exchange between GATA2 and GATA1 in the course of differentiation, operates on more than one-third of GATA1 bound genes. The switch is equally likely to lead to transcriptional activation or repression; and in general, GATA1 and GATA2 act oppositely on switch target genes. In addition, we show that genomic regions co-occupied by GATA2 and the ETS factor ETS1 are strongly enriched for regions marked by H3K4me3 and occupied by Pol II. Finally, by comparing GATA1 occupancy in erythroid cells and megakaryocytes, we find that the presence of ETS factor motifs is a major discriminator of megakaryocyte versus red cell specification.


Subject(s)
Chromatin/genetics , GATA1 Transcription Factor/genetics , GATA2 Transcription Factor/genetics , Genes, Switch/genetics , Genome-Wide Association Study , Hematopoiesis/genetics , Animals , Cell Lineage/physiology , Chromatin/metabolism , Erythroid Cells/cytology , GATA1 Transcription Factor/metabolism , GATA2 Transcription Factor/metabolism , Gene Expression Regulation, Developmental/physiology , Histones/metabolism , Megakaryocytes/cytology , Megakaryocytes/physiology , Methylation , Mice , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism
9.
Blood ; 118(2): 231-9, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21622645

ABSTRACT

Erythroid cells and megakaryocytes are derived from a common precursor, the megakaryocyte-erythroid progenitor. Although these 2 closely related hematopoietic cell types share many transcription factors, there are several key differences in their regulatory networks that lead to differential gene expression downstream of the megakaryocyte-erythroid progenitor. With the advent of next-generation sequencing and our ability to precisely define transcription factor chromatin occupancy in vivo on a global scale, we are much closer to understanding how these 2 lineages are specified and in general how transcription factor complexes govern hematopoiesis.


Subject(s)
Erythroid Cells , Gene Regulatory Networks/physiology , Hematopoiesis/genetics , Megakaryocyte-Erythroid Progenitor Cells/physiology , Megakaryocytes/physiology , Transcription Factors/genetics , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Lineage/genetics , Cell Lineage/physiology , Erythroid Cells/metabolism , Hematopoiesis/physiology , Humans , Megakaryocyte-Erythroid Progenitor Cells/metabolism , Megakaryocytes/metabolism , Models, Biological , Transcription Factors/metabolism , Transcription Factors/physiology
10.
Proc Natl Acad Sci U S A ; 105(9): 3333-8, 2008 Mar 04.
Article in English | MEDLINE | ID: mdl-18303114

ABSTRACT

MicroRNAs (miRNAs) control tissue development, but their mechanism of regulation is not well understood. We used a gene complementation strategy combined with microarray screening to identify miRNAs involved in the formation of erythroid (red blood) cells. Two conserved miRNAs, miR 144 and miR 451, emerged as direct targets of the critical hematopoietic transcription factor GATA-1. In vivo, GATA-1 binds a distal upstream regulatory element to activate RNA polymerase II-mediated transcription of a single common precursor RNA (pri-miRNA) encoding both mature miRNAs. Zebrafish embryos depleted of miR 451 by using antisense morpholinos form erythroid precursors, but their development into mature circulating red blood cells is strongly and specifically impaired. These results reveal a miRNA locus that is required for erythropoiesis and uncover a new regulatory axis through which GATA-1 controls this process.


Subject(s)
Erythroid Precursor Cells/cytology , Erythropoiesis/genetics , GATA1 Transcription Factor/physiology , MicroRNAs/physiology , Animals , Cell Line, Tumor , Erythroid-Specific DNA-Binding Factors , In Situ Hybridization , Mice , MicroRNAs/analysis , Microarray Analysis , Zebrafish
11.
J Clin Invest ; 117(7): 1856-65, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17607360

ABSTRACT

Erythrocyte precursors produce abundant alpha- and beta-globin proteins, which assemble with each other to form hemoglobin A (HbA), the major blood oxygen carrier. alphaHb-stabilizing protein (AHSP) binds free alpha subunits reversibly to maintain their structure and limit their ability to generate reactive oxygen species. Accordingly, loss of AHSP aggravates the toxicity of excessive free alpha-globin caused by beta-globin gene disruption in mice. Surprisingly, we found that AHSP also has important functions when free alpha-globin is limited. Thus, compound mutants lacking both Ahsp and 1 of 4 alpha-globin genes (genotype Ahsp(-/-)alpha-globin*(alpha/alphaalpha)) exhibited more severe anemia and Hb instability than mice with either mutation alone. In vitro, recombinant AHSP promoted folding of newly translated alpha-globin, enhanced its refolding after denaturation, and facilitated its incorporation into HbA. Moreover, in erythroid precursors, newly formed free alpha-globin was destabilized by loss of AHSP. Therefore, in addition to its previously defined role in detoxification of excess alpha-globin, AHSP also acts as a molecular chaperone to stabilize nascent alpha-globin for HbA assembly. Our findings illustrate what we believe to be a novel adaptive mechanism by which a specialized cell coordinates high-level production of a multisubunit protein and protects against various synthetic imbalances.


Subject(s)
Blood Proteins/metabolism , Erythroid Cells/metabolism , Hemoglobins/metabolism , Molecular Chaperones/metabolism , Protein Folding , Animals , Apoproteins/metabolism , Blood Proteins/deficiency , Blood Proteins/genetics , Cell Differentiation , Cell Shape , Erythroid Cells/cytology , Hemoglobins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Molecular Chaperones/genetics , Mutation/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , Reticulocytes/metabolism
12.
Mol Cell Biol ; 25(15): 6747-59, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16024808

ABSTRACT

Stem cell factor (SCF), erythropoietin (Epo), and GATA-1 play an essential role(s) in erythroid development. We examined how these proteins interact functionally in G1E cells, a GATA-1(-) erythroblast line that proliferates in an SCF-dependent fashion and, upon restoration of GATA-1 function, undergoes GATA-1 proliferation arrest and Epo-dependent terminal maturation. We show that SCF-induced cell cycle progression is mediated via activation of the Src kinase/c-Myc pathway. Restoration of GATA-1 activity induced G1 cell cycle arrest coincident with repression of c-Kit and its downstream effectors Vav1, Rac1, and Akt. Sustained expression of each of these individual signaling components inhibited GATA-1-induced cell cycle arrest to various degrees but had no effects on the expression of GATA-1-regulated erythroid maturation markers. Chromatin immunoprecipitation analysis revealed that GATA-1 occupies a defined Kit gene regulatory element in vivo, suggesting a direct mechanism for gene repression. Hence, in addition to its well-established function as an activator of erythroid genes, GATA-1 also participates in a distinct genetic program that inhibits cell proliferation by repressing the expression of multiple components of the c-Kit signaling axis. Our findings reveal a novel aspect of molecular cross talk between essential transcriptional and cytokine signaling components of hematopoietic development.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation , DNA-Binding Proteins/physiology , Erythroid Cells/enzymology , Growth Inhibitors/physiology , Proto-Oncogene Proteins c-kit/biosynthesis , Proto-Oncogene Proteins c-kit/metabolism , Repressor Proteins/physiology , Transcription Factors/physiology , Animals , Apoptosis/physiology , Cell Line, Transformed , Clone Cells , DNA-Binding Proteins/biosynthesis , Erythroid Cells/cytology , Erythroid Cells/metabolism , Erythroid-Specific DNA-Binding Factors , Erythropoietin/metabolism , GATA1 Transcription Factor , Gene Expression Regulation/physiology , Growth Inhibitors/biosynthesis , Mice , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/physiology , Repressor Proteins/biosynthesis , Signal Transduction/physiology , Stem Cell Factor/metabolism , Substrate Specificity , Transcription Factors/biosynthesis , src-Family Kinases/metabolism
13.
Nat Genet ; 50(5): 764, 2018 05.
Article in English | MEDLINE | ID: mdl-29255265

ABSTRACT

In the version of this article initially published, in the Methods, the Gene Expression Omnibus accession code for H3K36me3 ChIP-seq data was incorrectly given as GSM1003585 instead of GSM733725. The error has been corrected in the HTML, PDF and print versions of the article.

14.
Nat Genet ; 50(5): 764, 2018 05.
Article in English | MEDLINE | ID: mdl-29255266

ABSTRACT

The version of the Supplementary Text and Figures file initially posted was missing Supplementary Tables 1-6 and the Supplementary Note and used incorrect versions of the supplementary figures.

15.
Nat Genet ; 50(1): 83-95, 2018 01.
Article in English | MEDLINE | ID: mdl-29203910

ABSTRACT

TET enzymes oxidize 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), which can lead to DNA demethylation. However, direct connections between TET-mediated DNA demethylation and transcriptional output are difficult to establish owing to challenges in distinguishing global versus locus-specific effects. Here we show that TET1, TET2 and TET3 triple-knockout (TKO) human embryonic stem cells (hESCs) exhibit prominent bivalent promoter hypermethylation without an overall corresponding decrease in gene expression in the undifferentiated state. Focusing on the bivalent PAX6 locus, we find that increased DNMT3B binding is associated with promoter hypermethylation, which precipitates a neural differentiation defect and failure of PAX6 induction during differentiation. dCas9-mediated locus-specific demethylation and global inactivation of DNMT3B in TKO hESCs partially reverses the hypermethylation at the PAX6 promoter and improves differentiation to neuroectoderm. Taking these findings together with further genome-wide methylation and TET1 and DNMT3B ChIP-seq analyses, we conclude that TET proteins safeguard bivalent promoters from de novo methylation to ensure robust lineage-specific transcription upon differentiation.


Subject(s)
DNA Methylation , DNA-Binding Proteins/physiology , Embryonic Stem Cells/metabolism , Mixed Function Oxygenases/physiology , Promoter Regions, Genetic , Animals , Cell Differentiation/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Dioxygenases/physiology , Embryonic Stem Cells/cytology , Humans , Mice , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Mutation , Neural Plate/cytology , PAX6 Transcription Factor/biosynthesis , PAX6 Transcription Factor/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology
16.
Neuron ; 97(2): 313-325.e6, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29346752

ABSTRACT

N6-methyladenosine (m6A) affects multiple aspects of mRNA metabolism and regulates developmental transitions by promoting mRNA decay. Little is known about the role of m6A in the adult mammalian nervous system. Here we report that sciatic nerve lesion elevates levels of m6A-tagged transcripts encoding many regeneration-associated genes and protein translation machinery components in the adult mouse dorsal root ganglion (DRG). Single-base resolution m6A-CLIP mapping further reveals a dynamic m6A landscape in the adult DRG upon injury. Loss of either m6A methyltransferase complex component Mettl14 or m6A-binding protein Ythdf1 globally attenuates injury-induced protein translation in adult DRGs and reduces functional axon regeneration in the peripheral nervous system in vivo. Furthermore, Pten deletion-induced axon regeneration of retinal ganglion neurons in the adult central nervous system is attenuated upon Mettl14 knockdown. Our study reveals a critical epitranscriptomic mechanism in promoting injury-induced protein synthesis and axon regeneration in the adult mammalian nervous system.


Subject(s)
Adenosine/physiology , Axons/physiology , Epigenesis, Genetic/genetics , Methyltransferases/physiology , Nerve Regeneration/genetics , Nerve Tissue Proteins/physiology , RNA Processing, Post-Transcriptional , Transcription, Genetic , Adenosine/analogs & derivatives , Animals , Ganglia, Spinal/metabolism , Gene Ontology , Methyltransferases/deficiency , Mice, Knockout , Nerve Crush , PTEN Phosphohydrolase/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins/physiology , Sciatic Nerve/injuries , Sciatic Neuropathy/genetics , Sciatic Neuropathy/physiopathology , Sensory Receptor Cells/physiology , Sensory Receptor Cells/ultrastructure
17.
Cell Stem Cell ; 22(2): 191-205.e9, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29290617

ABSTRACT

N6-methyladenosine (m6A), the most prevalent internal modification in eukaryotic messenger RNAs (mRNAs), plays critical roles in many bioprocesses. However, its functions in normal and malignant hematopoiesis remain elusive. Here, we report that METTL14, a key component of the m6A methyltransferase complex, is highly expressed in normal hematopoietic stem/progenitor cells (HSPCs) and acute myeloid leukemia (AML) cells carrying t(11q23), t(15;17), or t(8;21) and is downregulated during myeloid differentiation. Silencing of METTL14 promotes terminal myeloid differentiation of normal HSPCs and AML cells and inhibits AML cell survival/proliferation. METTL14 is required for development and maintenance of AML and self-renewal of leukemia stem/initiation cells (LSCs/LICs). Mechanistically, METTL14 exerts its oncogenic role by regulating its mRNA targets (e.g., MYB and MYC) through m6A modification, while the protein itself is negatively regulated by SPI1. Collectively, our results reveal the SPI1-METTL14-MYB/MYC signaling axis in myelopoiesis and leukemogenesis and highlight the critical roles of METTL14 and m6A modification in normal and malignant hematopoiesis.


Subject(s)
Adenosine/analogs & derivatives , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Differentiation , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Methyltransferases/metabolism , Adenosine/metabolism , Animals , Carcinogenesis/metabolism , Cell Proliferation , Cell Survival , Down-Regulation/genetics , Gene Expression Regulation, Leukemic , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Methyltransferases/genetics , Mice, Inbred C57BL , Myeloid Cells/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Biosynthesis , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trans-Activators/metabolism , Transcriptome/genetics , Up-Regulation/genetics
18.
Cell Discov ; 3: 17013, 2017.
Article in English | MEDLINE | ID: mdl-28529766

ABSTRACT

Sertoli cells have dual roles during the cells' lifetime. In the juvenile mammal, Sertoli cells proliferate and create the structure of the testis, and during puberty they cease to proliferate and take on the adult role of supporting germ cells through spermatogenesis. Accordingly, many genes expressed in Sertoli cells during testis formation are repressed during spermatogenesis. 5-Hydroxymethylcytosine (5hmC) is a DNA modification enzymatically generated from 5mC and present in all investigated mammalian tissues at varying levels. Using mass spectrometry and immunofluorescence staining we identified a substantial Sertoli cell-specific global 5hmC increase during rat puberty. Chemical labeling, pull-down and sequencing of 5hmC-containing genomic DNA from juvenile and adult rat Sertoli cells revealed that genes that lose or gain 5hmC belong to different functional pathways and mirror the functions of the cells in the two different states. Loss of 5hmC is associated with genes involved in development and cell structure, whereas gain of 5hmC is associated with genes involved in cellular pathways pertaining to the function of the adult Sertoli cells. This redistribution during maturation shows that 5hmC is a dynamic nucleotide modification, correlated to gene expression.

19.
Nat Commun ; 5: 5630, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25430002

ABSTRACT

Recent discoveries of reversible N(6)-methyladenosine (m(6)A) methylation on messenger RNA (mRNA) and mapping of m(6)A methylomes in mammals and yeast have revealed potential regulatory functions of this RNA modification. In plants, defects in m(6)A methyltransferase cause an embryo-lethal phenotype, suggesting a critical role of m(6)A in plant development. Here, we profile m(6)A transcriptome-wide in two accessions of Arabidopsis thaliana and reveal that m(6)A is a highly conserved modification of mRNA in plants. Distinct from mammals, m(6)A in A. thaliana is enriched not only around the stop codon and within 3'-untranslated regions, but also around the start codon. Gene ontology analysis indicates that the unique distribution pattern of m(6)A in A. thaliana is associated with plant-specific pathways involving the chloroplast. We also discover a positive correlation between m(6)A deposition and mRNA abundance, suggesting a regulatory role of m(6)A in plant gene expression.


Subject(s)
Adenosine/analogs & derivatives , Arabidopsis , Gene Expression Regulation, Plant , RNA, Messenger/metabolism , 3' Untranslated Regions , Adenosine/metabolism , Codon, Initiator/metabolism , Codon, Terminator/metabolism , Gene Expression Profiling , Methyltransferases/genetics , Methyltransferases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL