Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Cell ; 186(9): 2002-2017.e21, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37080201

ABSTRACT

Paired mapping of single-cell gene expression and electrophysiology is essential to understand gene-to-function relationships in electrogenic tissues. Here, we developed in situ electro-sequencing (electro-seq) that combines flexible bioelectronics with in situ RNA sequencing to stably map millisecond-timescale electrical activity and profile single-cell gene expression from the same cells across intact biological networks, including cardiac and neural patches. When applied to human-induced pluripotent stem-cell-derived cardiomyocyte patches, in situ electro-seq enabled multimodal in situ analysis of cardiomyocyte electrophysiology and gene expression at the cellular level, jointly defining cell states and developmental trajectories. Using machine-learning-based cross-modal analysis, in situ electro-seq identified gene-to-electrophysiology relationships throughout cardiomyocyte development and accurately reconstructed the evolution of gene expression profiles based on long-term stable electrical measurements. In situ electro-seq could be applicable to create spatiotemporal multimodal maps in electrogenic tissues, potentiating the discovery of cell types and gene programs responsible for electrophysiological function and dysfunction.


Subject(s)
Electronics , Sequence Analysis, RNA , Humans , Cell Differentiation , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/metabolism , Single-Cell Analysis , Transcriptome , Electronics/methods
2.
Mol Cell ; 83(23): 4304-4317.e8, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37949069

ABSTRACT

RNA-binding proteins (RBPs) control messenger RNA fate in neurons. Here, we report a mechanism that the stimuli-induced neuronal translation is mediated by phosphorylation of a YTHDF1-binding protein FMRP. Mechanistically, YTHDF1 can condense with ribosomal proteins to promote the translation of its mRNA targets. FMRP regulates this process by sequestering YTHDF1 away from the ribosome; upon neuronal stimulation, FMRP becomes phosphorylated and releases YTHDF1 for translation upregulation. We show that a new small molecule inhibitor of YTHDF1 can reverse fragile X syndrome (FXS) developmental defects associated with FMRP deficiency in an organoid model. Our study thus reveals that FMRP and its phosphorylation are important regulators of activity-dependent translation during neuronal development and stimulation and identifies YTHDF1 as a potential therapeutic target for FXS in which developmental defects caused by FMRP depletion could be reversed through YTHDF1 inhibition.


Subject(s)
Fragile X Mental Retardation Protein , Fragile X Syndrome , Humans , Phosphorylation , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Neurons/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Ribosomal Proteins/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
3.
Cell ; 161(6): 1388-99, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26046440

ABSTRACT

N(6)-methyladenosine (m(6)A) is the most abundant internal modification in mammalian mRNA. This modification is reversible and non-stoichiometric and adds another layer to the dynamic control of mRNA metabolism. The stability of m(6)A-modified mRNA is regulated by an m(6)A reader protein, human YTHDF2, which recognizes m(6)A and reduces the stability of target transcripts. Looking at additional functional roles for the modification, we find that another m(6)A reader protein, human YTHDF1, actively promotes protein synthesis by interacting with translation machinery. In a unified mechanism of m(6)A-based regulation in the cytoplasm, YTHDF2-mediated degradation controls the lifetime of target transcripts, whereas YTHDF1-mediated translation promotion increases translation efficiency, ensuring effective protein production from dynamic transcripts that are marked by m(6)A. Therefore, the m(6)A modification in mRNA endows gene expression with fast responses and controllable protein production through these mechanisms.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/metabolism , Gene Expression Regulation , Protein Biosynthesis , Humans , Peptide Initiation Factors/metabolism , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribosomes/metabolism
4.
Nature ; 622(7983): 552-561, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37758947

ABSTRACT

Spatially charting molecular cell types at single-cell resolution across the 3D volume is critical for illustrating the molecular basis of brain anatomy and functions. Single-cell RNA sequencing has profiled molecular cell types in the mouse brain1,2, but cannot capture their spatial organization. Here we used an in situ sequencing method, STARmap PLUS3,4, to profile 1,022 genes in 3D at a voxel size of 194 × 194 × 345 nm3, mapping 1.09 million high-quality cells across the adult mouse brain and spinal cord. We developed computational pipelines to segment, cluster and annotate 230 molecular cell types by single-cell gene expression and 106 molecular tissue regions by spatial niche gene expression. Joint analysis of molecular cell types and molecular tissue regions enabled a systematic molecular spatial cell-type nomenclature and identification of tissue architectures that were undefined in established brain anatomy. To create a transcriptome-wide spatial atlas, we integrated STARmap PLUS measurements with a published single-cell RNA-sequencing atlas1, imputing single-cell expression profiles of 11,844 genes. Finally, we delineated viral tropisms of a brain-wide transgene delivery tool, AAV-PHP.eB5,6. Together, this annotated dataset provides a single-cell resource that integrates the molecular spatial atlas, brain anatomy and the accessibility to genetic manipulation of the mammalian central nervous system.


Subject(s)
Central Nervous System , Imaging, Three-Dimensional , Single-Cell Analysis , Transcriptome , Animals , Mice , Brain/anatomy & histology , Brain/cytology , Brain/metabolism , Central Nervous System/anatomy & histology , Central Nervous System/cytology , Central Nervous System/metabolism , Single-Cell Analysis/methods , Spinal Cord/anatomy & histology , Spinal Cord/cytology , Spinal Cord/metabolism , Transcriptome/genetics , Single-Cell Gene Expression Analysis , Viral Tropism , Datasets as Topic , Transgenes/genetics , Imaging, Three-Dimensional/methods
5.
Mol Cell ; 74(4): 640-650, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31100245

ABSTRACT

Cellular RNAs are naturally decorated with a variety of chemical modifications. The structural diversity of the modified nucleosides provides regulatory potential to sort groups of RNAs for organized metabolism and functions, thus affecting gene expression. Recent years have witnessed a burst of interest in and understanding of RNA modification biology, thanks to the emerging transcriptome-wide sequencing methods for mapping modified sites, highly sensitive mass spectrometry for precise modification detection and quantification, and extensive characterization of the modification "effectors," including enzymes ("writers" and "erasers") that alter the modification level and binding proteins ("readers") that recognize the chemical marks. However, challenges remain due to the vast heterogeneity in expression abundance of different RNA species, further complicated by divergent cell-type-specific and tissue-specific expression and localization of the effectors as well as modifications. In this review, we highlight recent progress in understanding the function of N6-methyladenosine (m6A), the most abundant internal mark on eukaryotic mRNA, in light of the specific biological contexts of m6A effectors. We emphasize the importance of context for RNA modification regulation and function.


Subject(s)
Adenosine/analogs & derivatives , Methylation , RNA, Messenger/genetics , RNA/genetics , Adenosine/genetics , Eukaryotic Cells/metabolism , Gene Expression Regulation/genetics , Organ Specificity/genetics , RNA Processing, Post-Transcriptional/genetics , Transcriptome
6.
Mol Cell ; 76(1): 70-81.e9, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31445886

ABSTRACT

N6-methyladenosine (m6A) modification occurs co-transcriptionally and impacts pre-mRNA processing; however, the mechanism of co-transcriptional m6A-dependent alternative splicing regulation is still poorly understood. Heterogeneous nuclear ribonucleoprotein G (hnRNPG) is an m6A reader protein that binds RNA through RRM and Arg-Gly-Gly (RGG) motifs. Here, we show that hnRNPG directly binds to the phosphorylated carboxy-terminal domain (CTD) of RNA polymerase II (RNAPII) using RGG motifs in its low-complexity region. Through interactions with the phosphorylated CTD and nascent RNA, hnRNPG associates co-transcriptionally with RNAPII and regulates alternative splicing transcriptome-wide. m6A near splice sites in nascent pre-mRNA modulates hnRNPG binding, which influences RNAPII occupancy patterns and promotes exon inclusion. Our results reveal an integrated mechanism of co-transcriptional m6A-mediated splicing regulation, in which an m6A reader protein uses RGG motifs to co-transcriptionally interact with both RNAPII and m6A-modified nascent pre-mRNA to modulate RNAPII occupancy and alternative splicing.


Subject(s)
Adenosine/analogs & derivatives , Alternative Splicing , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , RNA Precursors/biosynthesis , RNA, Messenger/biosynthesis , Transcription, Genetic , Adenosine/metabolism , Amino Acid Motifs , Binding Sites , Exons , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoproteins/chemistry , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Protein Binding , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA Precursors/genetics , RNA, Messenger/genetics , Structure-Activity Relationship
7.
Mol Cell ; 71(6): 973-985.e5, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30197295

ABSTRACT

FTO, the first RNA demethylase discovered, mediates the demethylation of internal N6-methyladenosine (m6A) and N6, 2-O-dimethyladenosine (m6Am) at the +1 position from the 5' cap in mRNA. Here we demonstrate that the cellular distribution of FTO is distinct among different cell lines, affecting the access of FTO to different RNA substrates. We find that FTO binds multiple RNA species, including mRNA, snRNA, and tRNA, and can demethylate internal m6A and cap m6Am in mRNA, internal m6A in U6 RNA, internal and cap m6Am in snRNAs, and N1-methyladenosine (m1A) in tRNA. FTO-mediated demethylation has a greater effect on the transcript levels of mRNAs possessing internal m6A than the ones with cap m6Am in the tested cells. We also show that FTO can directly repress translation by catalyzing m1A tRNA demethylation. Collectively, FTO-mediated RNA demethylation occurs to m6A and m6Am in mRNA and snRNA as well as m1A in tRNA.


Subject(s)
Adenosine/analogs & derivatives , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/physiology , 3T3-L1 Cells , Adenosine/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Animals , Cell Nucleus , Cytoplasm , Demethylation , Gene Expression/genetics , HEK293 Cells , HeLa Cells , Humans , Methylation , Mice , RNA Processing, Post-Transcriptional/physiology , RNA, Messenger/metabolism , RNA, Small Nuclear/metabolism , RNA, Transfer/metabolism
9.
Nature ; 563(7730): 249-253, 2018 11.
Article in English | MEDLINE | ID: mdl-30401835

ABSTRACT

N6-methyladenosine (m6A), the most prevalent internal RNA modification on mammalian messenger RNAs, regulates the fates and functions of modified transcripts through m6A-specific binding proteins1-5. In the nervous system, m6A is abundant and modulates various neural functions6-11. Whereas m6A marks groups of mRNAs for coordinated degradation in various physiological processes12-15, the relevance of m6A for mRNA translation in vivo remains largely unknown. Here we show that, through its binding protein YTHDF1, m6A promotes protein translation of target transcripts in response to neuronal stimuli in the adult mouse hippocampus, thereby facilitating learning and memory. Mice with genetic deletion of Ythdf1 show learning and memory defects as well as impaired hippocampal synaptic transmission and long-term potentiation. Re-expression of YTHDF1 in the hippocampus of adult Ythdf1-knockout mice rescues the behavioural and synaptic defects, whereas hippocampus-specific acute knockdown of Ythdf1 or Mettl3, which encodes the catalytic component of the m6A methyltransferase complex, recapitulates the hippocampal deficiency. Transcriptome-wide mapping of YTHDF1-binding sites and m6A sites on hippocampal mRNAs identified key neuronal genes. Nascent protein labelling and tether reporter assays in hippocampal neurons showed that YTHDF1 enhances protein synthesis in a neuronal-stimulus-dependent manner. In summary, YTHDF1 facilitates translation of m6A-methylated neuronal mRNAs in response to neuronal stimulation, and this process contributes to learning and memory.


Subject(s)
Adenine/analogs & derivatives , Hippocampus/cytology , Hippocampus/physiology , Memory/physiology , Neurons/metabolism , RNA-Binding Proteins/metabolism , Adenine/metabolism , Animals , Binding Sites , Female , Male , Methyltransferases/deficiency , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Knockout , Neuronal Plasticity , Protein Biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Spatial Learning/physiology , Synaptic Transmission
10.
Nature ; 542(7642): 475-478, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28192787

ABSTRACT

The maternal-to-zygotic transition (MZT) is one of the most profound and tightly orchestrated processes during the early life of embryos, yet factors that shape the temporal pattern of vertebrate MZT are largely unknown. Here we show that over one-third of zebrafish maternal messenger RNAs (mRNAs) can be N6-methyladenosine (m6A) modified, and the clearance of these maternal mRNAs is facilitated by an m6A-binding protein, Ythdf2. Removal of Ythdf2 in zebrafish embryos decelerates the decay of m6A-modified maternal mRNAs and impedes zygotic genome activation. These embryos fail to initiate timely MZT, undergo cell-cycle pause, and remain developmentally delayed throughout larval life. Our study reveals m6A-dependent RNA decay as a previously unidentified maternally driven mechanism that regulates maternal mRNA clearance during zebrafish MZT, highlighting the critical role of m6A mRNA methylation in transcriptome switching and animal development.


Subject(s)
Adenosine/analogs & derivatives , Embryonic Development/genetics , RNA Stability , RNA, Messenger, Stored/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zygote/metabolism , Adenosine/metabolism , Animals , Female , Male , RNA, Messenger, Stored/chemistry , RNA, Messenger, Stored/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Time Factors , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
11.
J Biol Chem ; 294(52): 19889-19895, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31753916

ABSTRACT

N6-Methyladenosine (m6A) is the most abundant post-transcriptional mRNA modification in eukaryotes and exerts many of its effects on gene expression through reader proteins that bind specifically to m6A-containing transcripts. Fragile X mental retardation protein (FMRP), an RNA-binding protein, has previously been shown to affect the translation of target mRNAs and trafficking of mRNA granules. Loss of function of FMRP causes fragile X syndrome, the most common form of inherited intellectual disability in humans. Using HEK293T cells, siRNA-mediated gene knockdown, cytoplasmic and nuclear fractions, RNA-Seq, and LC-MS/MS analyses, we demonstrate here that FMRP binds directly to a collection of m6A sites on mRNAs. FMRP depletion increased mRNA m6A levels in the nucleus. Moreover, the abundance of FMRP targets in the cytoplasm relative to the nucleus was decreased in Fmr1-KO mice, an effect also observed in highly methylated genes. We conclude that FMRP may affect the nuclear export of m6A-modified RNA targets.


Subject(s)
Adenosine/analogs & derivatives , Fragile X Mental Retardation Protein/metabolism , RNA, Messenger/metabolism , Active Transport, Cell Nucleus , Adenosine/metabolism , Animals , Binding Sites , Cell Nucleus/metabolism , Cerebral Cortex/metabolism , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA Interference , RNA Stability , RNA, Messenger/chemistry , RNA, Small Interfering/metabolism
12.
Methods ; 156: 85-90, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30471344

ABSTRACT

The post-transcriptional modification 2'-O-Methyl (2'OMe) could be present on the ribose of all four ribonucleosides, and is highly prevalent in a wide variety of RNA species, including the 5' RNA cap of viruses and higher eukaryotes, as well as internally in transfer RNA and ribosomal RNA. Recent studies have suggested that 2'OMe is also located internally in low-abundance RNA species such as viral RNA and mRNA. To profile 2'OMe on different RNA species, we have developed Nm-seq, which could identify 2'OMe sites at single base resolution. Nm-seq is particularly useful for identifying 2'OMe sites located at the 3' terminal ends of small RNAs. Here, we present an optimized protocol for Nm-seq and a protocol for applying Nm-seq to identify 2'OMe sites on small RNA 3' terminal ends.


Subject(s)
MicroRNAs/genetics , Molecular Sequence Annotation/methods , Poly A/genetics , RNA, Messenger/genetics , 3' Flanking Region , Arabidopsis/genetics , Arabidopsis/metabolism , Base Sequence , Gene Library , Humans , Hydrolysis , Methylation , MicroRNAs/metabolism , Oxidation-Reduction , Phosphorylation , Poly A/metabolism , RNA, Messenger/metabolism , Ribose/metabolism , Seedlings/genetics , Seedlings/metabolism
13.
Int J Obes (Lond) ; 42(11): 1912-1924, 2018 11.
Article in English | MEDLINE | ID: mdl-29487348

ABSTRACT

BACKGROUND/OBJECTIVE: N6-methyladenosine (m6A) modification of mRNA plays an important role in regulating adipogenesis. However, its underlying mechanism remains largely unknown. SUBJECTS/METHODS: Using Jinhua and Landrace pigs as fat and lean models, we presented a comprehensive transcriptome-wide m6A profiling in adipose tissues from these two pig breeds. Two differentially methylated genes were selected to explore the mechanisms of m6A-mediated regulation of gene function. RESULTS: The ratio of m6A/A in the layer of backfat (LB) was significantly higher in Landrace than that in Jinhua. Transcriptome-wide m6A profiling revealed that m6A modification on mRNA occurs in the conserved sequence motif of RRACH and that the pig transcriptome contains 0.53-0.91 peak per actively expressed transcript. The relative density of m6A peaks in the 3'UTR were higher than in 5'UTR. Genes with common m6A peaks from both Landrace (L-LB) and Jinhua (J-LB) were enriched in RNA splicing and cellular lipid metabolic process. The unique m6A peak genes (UMGs) from L-LB were mainly enriched in the extracellular matrix (ECM) and collagen catabolic process, whereas the UMGs from J-LB are mainly involved in RNA splicing, etc. Lipid metabolism processes were not significantly enriched in the UMGs from L-LB or J-LB. Uncoupling protein-2 (UCP2) and patatin-like phospholipase domain containing 2 (PNPLA2) were two of the UMGs in L-LB. Synonymous mutations (MUT) were conducted to reduce m6A level of UCP2 and PNPLA2 mRNAs. Adipogenesis test showed that UCP2-MUT further inhibited adipogenesis, while PNPLA2-MUT promoted lipid accumulation compared with UCP2-WT and PNPLA2-WT, respectively. Further study showed m6A negatively mediates UCP2 protein expression and positively mediates PNPLA2 protein expression. m6A modification affects the translation of PNPLA2 most likely through YTHDF1, whereas UCP2 is likely neither the target of YTHDF2 nor the target of YTHDF1. CONCLUSION: Our data demonstrated a conserved and yet dynamically regulated m6A methylome in pig transcriptomes and provided an important resource for studying the function of m6A epitranscriptomic modification in obesity development.


Subject(s)
Adipocytes/metabolism , Lipid Metabolism/physiology , Obesity/pathology , RNA, Messenger/metabolism , Thinness/pathology , Uncoupling Protein 2/metabolism , Animals , Disease Models, Animal , Gene Expression Profiling , Lipase/metabolism , Sequence Analysis, RNA , Swine , Up-Regulation/physiology
14.
16.
Science ; 380(6652): eadd3067, 2023 06 30.
Article in English | MEDLINE | ID: mdl-37384709

ABSTRACT

The precise control of messenger RNA (mRNA) translation is a crucial step in posttranscriptional gene regulation of cellular physiology. However, it remains a challenge to systematically study mRNA translation at the transcriptomic scale with spatial and single-cell resolution. Here, we report the development of ribosome-bound mRNA mapping (RIBOmap), a highly multiplexed three-dimensional in situ profiling method to detect cellular translatome. RIBOmap profiling of 981 genes in HeLa cells revealed cell cycle-dependent translational control and colocalized translation of functional gene modules. We mapped 5413 genes in mouse brain tissues, yielding spatially resolved single-cell translatomic profiles for 119,173 cells and revealing cell type-specific and brain region-specific translational regulation, including translation remodeling during oligodendrocyte maturation. Our method detected widespread patterns of localized translation in neuronal and glial cells in intact brain tissue networks.


Subject(s)
Brain , Chromosome Mapping , Neuroglia , Neurons , Protein Biosynthesis , RNA, Messenger , Single-Cell Gene Expression Analysis , Animals , Humans , Mice , Brain/metabolism , HeLa Cells , Neuroglia/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Gene Expression Regulation , Chromosome Mapping/methods , Neurons/metabolism , Single-Cell Gene Expression Analysis/methods
17.
Biomed Res Int ; 2022: 6098799, 2022.
Article in English | MEDLINE | ID: mdl-36246960

ABSTRACT

Diabetic retinopathy (DR) is the most common complication of diabetes and is often characterized by damage to retinal vascular microcirculation, resulting in retinal exudation, hemorrhage, fibrosis, and neovascularization. With the aging of my country's population, the incidence of DR is increasing year by year, and it has become one of the main blinding eye diseases in ophthalmic diseases also tends to be younger. So far, although the pathogenesis of DR is not completely clear, scholars generally believe that DR is based on the disorder of glucose metabolism, causing changes in the microcirculation of ocular tissues, nerves, and blood vessels, resulting in chronic damage to the nutrition and visual function of the eye disease. In order to explore the demand for cardiovascular disease treatment, make up for the lack of chronic diseases affecting people's physical harm, and improve the success rate of cardiovascular disease treatment, a method to observe the efficacy and myocardial remodeling of trimetazidine combined with metoprolol in elderly patients with coronary heart disease and heart failure based on integrated traditional Chinese and Western medicine was proposed. 54 elderly people over 60 years old are afraid of cardiovascular disease and take active protection. A method based on observation of integrated traditional Chinese and Western medicine was proposed, and at the same time, an intelligent medical monitoring system was constructed to better study, observe, and improve the efficacy of trimetazidine combined with metoprolol in elderly patients with coronary heart disease, heart failure, and myocardial impact of refactoring. The results of the study show that trimetazidine has a good clinical effect on ischemic cardiomyopathy heart failure based on the observation of integrated traditional Chinese and Western medicine.


Subject(s)
Cardiovascular Diseases , Coronary Disease , Heart Failure , Trimetazidine , Aged , Cardiovascular Diseases/complications , China , Coronary Disease/complications , Coronary Disease/drug therapy , Glucose , Heart Failure/complications , Heart Failure/drug therapy , Humans , Metoprolol , Middle Aged , Trimetazidine/therapeutic use
18.
ACS Chem Biol ; 17(12): 3352-3366, 2022 12 16.
Article in English | MEDLINE | ID: mdl-34995053

ABSTRACT

mRNA has recently been established as a new class of therapeutics, due to its programmability and ability to produce proteins of interest rapidly in vivo. Despite its demonstrated utility, mRNA as a protein expression platform remains limited by its translational capacity and RNA stability. Here, we introduce messenger-oligonucleotide conjugated RNAs (mocRNAs) to enable site-specific, robust, and modularized encoding of chemical modifications for highly efficient and stable protein expression. In mocRNA constructs, chemically synthesized oligonucleotides are ligated to the 3' terminus of mRNA substrates to protect poly(A) tails from degradation, without compromising their potency in stimulating translation. As a proof-of-concept, mocRNAs modified by deadenylase-resistant oligonucleotides result in augmented protein production by factors of 2-4 in human HeLa cells and by 10-fold in primary rat cortical neuronal cultures. By directly linking enzymatic and organic synthesis of mRNA, we envision that the mocRNA design will open new avenues to expand the chemical space and translational capacity of RNA-based vectors in basic research and therapeutic applications.


Subject(s)
Oligonucleotides , RNA Stability , Humans , Rats , Animals , HeLa Cells , Oligonucleotides/metabolism , RNA/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Protein Biosynthesis , Mammals/genetics , Mammals/metabolism
19.
Environ Pollut ; 283: 117037, 2021 Aug 15.
Article in English | MEDLINE | ID: mdl-33866220

ABSTRACT

N6-methyladenosine (m6A) mRNA methylation plays a role in various brain functions. Exposure to chronic constant light (CCL) has been reported to impair cognition, yet whether the underlying mechanism involves m6A remains unknown. In this study, mice exposed to CCL for 3 weeks show impaired cognitive behavior, which was associated with increased m6A level in hippocampus. Accordingly, the m6A demethylase FTO was inhibited while the methyltransferases METTL3, METTL14 and WTAP, as well as the reader protein YTHDF2, were elevated in the hippocampus of CCL-exposed mice. CCL exposure significantly activated hippocampal expression of circadian regulator cryptochrome 1 and 2 (CRY1 and 2). Meanwhile, hippocampal neurogenesis was impaired with suppression of BDNF/TrκB/ERK pathway. To further delineate the signaling pathway and the role of m6A, we altered the expression of CRY1/2 in hippocampus neuron cells. CRY1/2 overexpression inhibited FTO and increased m6A levels, while CRY1/2 knockdown led to opposite results. Luciferase reporter analysis further confirmed CRY1/2-induced FTO suppression. Furthermore, FTO knockdown increased m6A on 3'UTR of TrκB mRNA, and decreased TrκB mRNA stability and TrκB protein expression, in a YTHDF2-dependent manner. These results indicate that CCL-activated CRY1/2 causes transcriptional inhibition of FTO, which suppresses TrκB expression in hippocampus via m6A-dependent post-transcriptional regulation and contributes to impaired cognitive behavior in mice exposed to constant light.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO , RNA Stability , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Animals , Cognition , Hippocampus/metabolism , Mice , RNA, Messenger
20.
Nat Commun ; 12(1): 177, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420027

ABSTRACT

Glioblastoma (GBM) is the most common type of adult malignant brain tumor, but its molecular mechanisms are not well understood. In addition, the knowledge of the disease-associated expression and function of YTHDF2 remains very limited. Here, we show that YTHDF2 overexpression clinically correlates with poor glioma patient prognosis. EGFR that is constitutively activated in the majority of GBM causes YTHDF2 overexpression through the EGFR/SRC/ERK pathway. EGFR/SRC/ERK signaling phosphorylates YTHDF2 serine39 and threonine381, thereby stabilizes YTHDF2 protein. YTHDF2 is required for GBM cell proliferation, invasion, and tumorigenesis. YTHDF2 facilitates m6A-dependent mRNA decay of LXRA and HIVEP2, which impacts the glioma patient survival. YTHDF2 promotes tumorigenesis of GBM cells, largely through the downregulation of LXRα and HIVEP2. Furthermore, YTHDF2 inhibits LXRα-dependent cholesterol homeostasis in GBM cells. Together, our findings extend the landscape of EGFR downstream circuit, uncover the function of YTHDF2 in GBM tumorigenesis, and highlight an essential role of RNA m6A methylation in cholesterol homeostasis.


Subject(s)
Brain Neoplasms/metabolism , Cholesterol/metabolism , ErbB Receptors/metabolism , Glioblastoma/metabolism , RNA-Binding Proteins/metabolism , Adult , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/metabolism , ErbB Receptors/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioblastoma/genetics , Glioma , Humans , Liver X Receptors/metabolism , MAP Kinase Signaling System , Male , Mice , Phosphorylation , RNA Stability , RNA-Binding Proteins/genetics , Signal Transduction , Transcription Factors/metabolism , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL