Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Cell ; 167(3): 816-828.e16, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27745969

ABSTRACT

tRNA is a central component of protein synthesis and the cell signaling network. One salient feature of tRNA is its heavily modified status, which can critically impact its function. Here, we show that mammalian ALKBH1 is a tRNA demethylase. It mediates the demethylation of N1-methyladenosine (m1A) in tRNAs. The ALKBH1-catalyzed demethylation of the target tRNAs results in attenuated translation initiation and decreased usage of tRNAs in protein synthesis. This process is dynamic and responds to glucose availability to affect translation. Our results uncover reversible methylation of tRNA as a new mechanism of post-transcriptional gene expression regulation.


Subject(s)
AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Gene Expression Regulation , Protein Biosynthesis/genetics , RNA, Transfer/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , Glucose/deficiency , HeLa Cells , Humans , Methylation , Polyribosomes/metabolism
2.
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
3.
Nature ; 591(7849): 317-321, 2021 03.
Article in English | MEDLINE | ID: mdl-33505026

ABSTRACT

METTL3 (methyltransferase-like 3) mediates the N6-methyladenosine (m6A) methylation of mRNA, which affects the stability of mRNA and its translation into protein1. METTL3 also binds chromatin2-4, but the role of METTL3 and m6A methylation in chromatin is not fully understood. Here we show that METTL3 regulates mouse embryonic stem-cell heterochromatin, the integrity of which is critical for silencing retroviral elements and for mammalian development5. METTL3 predominantly localizes to the intracisternal A particle (IAP)-type family of endogenous retroviruses. Knockout of Mettl3 impairs the deposition of multiple heterochromatin marks onto METTL3-targeted IAPs, and upregulates IAP transcription, suggesting that METTL3 is important for the integrity of IAP heterochromatin. We provide further evidence that RNA transcripts derived from METTL3-bound IAPs are associated with chromatin and are m6A-methylated. These m6A-marked transcripts are bound by the m6A reader YTHDC1, which interacts with METTL3 and in turn promotes the association of METTL3 with chromatin. METTL3 also interacts physically with the histone 3 lysine 9 (H3K9) tri-methyltransferase SETDB1 and its cofactor TRIM28, and is important for their localization to IAPs. Our findings demonstrate that METTL3-catalysed m6A modification of RNA is important for the integrity of IAP heterochromatin in mouse embryonic stem cells, revealing a mechanism of heterochromatin regulation in mammals.


Subject(s)
Chromatin Assembly and Disassembly , Heterochromatin/genetics , Heterochromatin/metabolism , Methyltransferases/metabolism , Mouse Embryonic Stem Cells/metabolism , Animals , Endogenous Retroviruses/genetics , Gene Expression Regulation , Genes, Intracisternal A-Particle/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/chemistry , Histones/metabolism , Mice , Tripartite Motif-Containing Protein 28/metabolism
4.
Mol Cell ; 74(6): 1304-1316.e8, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31031084

ABSTRACT

N7-methylguanosine (m7G) is a positively charged, essential modification at the 5' cap of eukaryotic mRNA, regulating mRNA export, translation, and splicing. m7G also occurs internally within tRNA and rRNA, but its existence and distribution within eukaryotic mRNA remain to be investigated. Here, we show the presence of internal m7G sites within mammalian mRNA. We then performed transcriptome-wide profiling of internal m7G methylome using m7G-MeRIP sequencing (MeRIP-seq). To map this modification at base resolution, we developed a chemical-assisted sequencing approach that selectively converts internal m7G sites into abasic sites, inducing misincorporation at these sites during reverse transcription. This base-resolution m7G-seq enabled transcriptome-wide mapping of m7G in human tRNA and mRNA, revealing distribution features of the internal m7G methylome in human cells. We also identified METTL1 as a methyltransferase that installs a subset of m7G within mRNA and showed that internal m7G methylation could affect mRNA translation.


Subject(s)
Chromosome Mapping/methods , Guanosine/analogs & derivatives , Methyltransferases/genetics , RNA, Messenger/genetics , RNA, Transfer/genetics , Transcriptome , Animals , Base Sequence , Cell Line , Fibroblasts/cytology , Fibroblasts/metabolism , Guanosine/metabolism , HEK293 Cells , HeLa Cells , Hep G2 Cells , High-Throughput Nucleotide Sequencing , Humans , Methylation , Methyltransferases/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , RNA, Transfer/metabolism , Reverse Transcription
6.
Mol Cell ; 69(6): 1028-1038.e6, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29547716

ABSTRACT

N6-methyladenosine (m6A) is an abundant modification in eukaryotic mRNA, regulating mRNA dynamics by influencing mRNA stability, splicing, export, and translation. However, the precise m6A regulating machinery still remains incompletely understood. Here we demonstrate that ZC3H13, a zinc-finger protein, plays an important role in modulating RNA m6A methylation in the nucleus. We show that knockdown of Zc3h13 in mouse embryonic stem cell significantly decreases global m6A level on mRNA. Upon Zc3h13 knockdown, a great majority of WTAP, Virilizer, and Hakai translocate to the cytoplasm, suggesting that Zc3h13 is required for nuclear localization of the Zc3h13-WTAP-Virilizer-Hakai complex, which is important for RNA m6A methylation. Finally, Zc3h13 depletion, as does WTAP, Virilizer, or Hakai, impairs self-renewal and triggers mESC differentiation. Taken together, our findings demonstrate that Zc3h13 plays a critical role in anchoring WTAP, Virilizer, and Hakai in the nucleus to facilitate m6A methylation and to regulate mESC self-renewal.


Subject(s)
Adenosine/analogs & derivatives , Cell Nucleus/metabolism , Cell Proliferation , Cell Self Renewal , Mouse Embryonic Stem Cells/metabolism , Nuclear Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , 3' Untranslated Regions , Active Transport, Cell Nucleus , Adenosine/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Differentiation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Methylation , Mice , Nuclear Proteins/genetics , RNA Splicing Factors , RNA Stability , RNA, Messenger/genetics , RNA-Binding Proteins , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
7.
Hum Mol Genet ; 30(22): 2110-2122, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34196368

ABSTRACT

The well-established functions of UHRF1 converge to DNA biological processes, as exemplified by DNA methylation maintenance and DNA damage repair during cell cycles. However, the potential effect of UHRF1 on RNA metabolism is largely unexplored. Here, we revealed that UHRF1 serves as a novel alternative RNA splicing regulator. The protein interactome of UHRF1 identified various splicing factors. Among them, SF3B3 could interact with UHRF1 directly and participate in UHRF1-regulated alternative splicing events. Furthermore, we interrogated the RNA interactome of UHRF1, and surprisingly, we identified U snRNAs, the canonical spliceosome components, in the purified UHRF1 complex. Unexpectedly, we found H3R2 methylation status determines the binding preference of U snRNAs, especially U2 snRNAs. The involvement of U snRNAs in UHRF1-containing complex and their binding preference to specific chromatin configuration imply a finely orchestrated mechanism at play. Our results provided the resources and pinpointed the molecular basis of UHRF1-mediated alternative RNA splicing, which will help us better our understanding of the physiological and pathological roles of UHRF1 in disease development.


Subject(s)
Alternative Splicing , CCAAT-Enhancer-Binding Proteins/metabolism , Histones/metabolism , RNA Splicing Factors/metabolism , RNA, Small Nuclear/genetics , Ubiquitin-Protein Ligases/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Humans , Methylation , Multiprotein Complexes , Nucleic Acid Conformation , Protein Binding , RNA, Small Nuclear/metabolism , Ubiquitin-Protein Ligases/genetics
8.
J Am Chem Soc ; 144(3): 1323-1331, 2022 01 26.
Article in English | MEDLINE | ID: mdl-35037455

ABSTRACT

As an aberrant base in DNA, uracil is generated by either deoxyuridine (dU) misincorporation or cytosine deamination, and involved in multiple physiological and pathological processes. Genome-wide profiles of uracil are important for study of these processes. Current methods for whole-genome mapping of uracil all rely on uracil-DNA N-glycosylase (UNG) and are limited in resolution, specificity, and/or sensitivity. Here, we developed a UdgX cross-linking and polymerase stalling sequencing ("Ucaps-seq") method to detect dU at single-nucleotide resolution. First, the specificity of Ucaps-seq was confirmed on synthetic DNA. Then the effectiveness of the approach was verified on two genomes from different sources. Ucaps-seq not only identified the enrichment of dU at dT sites in pemetrexed-treated cancer cells with globally elevated uracil but also detected dU at dC sites within the "WRC" motif in activated B cells which have increased dU in specific regions. Finally, Ucaps-seq was utilized to detect dU introduced by the cytosine base editor (nCas9-APOBEC) and identified a novel off-target site in cellular context. In conclusion, Ucaps-seq is a powerful tool with many potential applications, especially in evaluation of base editing fidelity.


Subject(s)
Nucleotides
9.
Biochem Biophys Res Commun ; 625: 31-37, 2022 10 15.
Article in English | MEDLINE | ID: mdl-35944361

ABSTRACT

Dilated cardiomyopathy, a type of heart muscle disease defined by the presence of left ventricular dilatation and contractile dysfunction, is an important cause of sudden cardiac death and heart failure. O-GlcNAcylation is an important post-translational modification of proteins by the addition of O-GlcNAc moieties at serine or threonine residues. Several studies have shown that proper control of O-GlcNAcylation is required for maintaining physiological function of heart by using Ogt (O-GlcNAc transferase) cardiomyocyte-specific knockout mouse models. In this study, we generated a new mouse model (αSMA-Ogt KO) in which Ogt was deleted in both cardiomyocytes and smooth muscle cells by crossing Ogt floxed mice with αSMA-Cre mice. αSMA-Cre-mediated Ogt deletion in mice led to severe postnatal lethality; the survived mice were smaller than control mice, had dilated hearts, and showed observable signs of heart failure. Moreover, the αSMA-Ogt KO heart had more apoptotic cells and fibrosis. The arteries of αSMA-Ogt KO mice exhibited significantly reduced expression of contractile genes and a trend towards arterial stiffness. In conclusion, our data emphasize the importance of OGT in maintaining normal heart function and reveal a novel role of OGT in regulating arterial contractility.


Subject(s)
Heart Failure , Muscle, Smooth, Vascular , Animals , Heart Failure/genetics , Heart Failure/metabolism , Integrases , Mice , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/metabolism , N-Acetylglucosaminyltransferases/metabolism
10.
Nat Chem Biol ; 16(5): 489-492, 2020 05.
Article in English | MEDLINE | ID: mdl-32015521

ABSTRACT

RNA secondary structure is critical to RNA regulation and function. We report a new N3-kethoxal reagent that allows fast and reversible labeling of single-stranded guanine bases in live cells. This N3-kethoxal-based chemistry allows efficient RNA labeling under mild conditions and transcriptome-wide RNA secondary structure mapping.


Subject(s)
Aldehydes/chemistry , RNA/chemistry , Animals , Butanones , Embryonic Stem Cells , Guanine/chemistry , HeLa Cells , High-Throughput Nucleotide Sequencing , Humans , Mice , Nucleic Acid Conformation , Nucleic Acid Heteroduplexes , RNA Folding , Transcriptome
11.
Circ Res ; 126(12): 1706-1720, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32212953

ABSTRACT

RATIONALE: The stress response of heart rate, which is determined by the plasticity of the sinoatrial node (SAN), is essential for cardiac function and survival in mammals. As an RNA-binding protein, CIRP (cold-inducible RNA-binding protein) can act as a stress regulator. Previously, we have documented that CIRP regulates cardiac electrophysiology at posttranscriptional level, suggesting its role in SAN plasticity, especially upon stress conditions. OBJECTIVE: Our aim was to clarify the role of CIRP in SAN plasticity and heart rate regulation under stress conditions. METHODS AND RESULTS: Telemetric ECG monitoring demonstrated an excessive acceleration of heart rate under isoprenaline stimulation in conscious CIRP-KO (knockout) rats. Patch-clamp analysis and confocal microscopic Ca2+ imaging of isolated SAN cells demonstrated that isoprenaline stimulation induced a faster spontaneous firing rate in CIRP-KO SAN cells than that in WT (wild type) SAN cells. A higher concentration of cAMP-the key mediator of pacemaker activity-was detected in CIRP-KO SAN tissues than in WT SAN tissues. RNA sequencing and quantitative real-time polymerase chain reaction analyses of single cells revealed that the 4B and 4D subtypes of PDE (phosphodiesterase), which controls cAMP degradation, were significantly decreased in CIRP-KO SAN cells. A PDE4 inhibitor (rolipram) abolished the difference in beating rate resulting from CIRP deficiency. The mechanistic study showed that CIRP stabilized the mRNA of Pde4b and Pde4d by direct mRNA binding, thereby regulating the protein expression of PDE4B and PDE4D at posttranscriptional level. CONCLUSIONS: CIRP acts as an mRNA stabilizer of specific PDEs to control the cAMP concentration in SAN, maintaining the appropriate heart rate stress response.


Subject(s)
Cold Shock Proteins and Peptides/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Heart Rate , Myocytes, Cardiac/metabolism , RNA-Binding Proteins/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Cells, Cultured , Cold Shock Proteins and Peptides/genetics , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Isoproterenol/pharmacology , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Phosphodiesterase Inhibitors/pharmacology , RNA Stability , RNA-Binding Proteins/genetics , Rats , Rats, Sprague-Dawley , Rolipram/pharmacology , Sinoatrial Node/cytology , Sinoatrial Node/metabolism , Sinoatrial Node/physiology , Stress, Physiological
12.
EMBO Rep ; 21(10): e49863, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32783360

ABSTRACT

RNA modifications represent a novel layer of regulation of gene expression. Functional experiments revealed that N6 -methyladenosine (m6 A) on messenger RNA (mRNA) plays critical roles in cell fate determination and development. m6 A mark also resides in the decoding center of 18S ribosomal RNA (rRNA); however, the biological function of m6 A on 18S rRNA is still poorly understood. Here, we report that methyltransferase-like 5 (METTL5) methylates 18S rRNA both in vivo and in vitro, which is consistent with previous reports. Deletion of Mettl5 causes a dramatic differentiation defect in mouse embryonic stem cells (mESCs). Mechanistically, the m6 A deposited by METTL5 is involved in regulating the efficient translation of F-box and WD repeat domain-containing 7 (FBXW7), a key regulator of cell differentiation. Deficiency of METTL5 reduces FBXW7 levels and leads to the accumulation of its substrate c-MYC, thereby delaying the onset of mESC differentiation. Our study uncovers an important role of METTL5-mediated 18S m6 A in mESC differentiation through translation regulation and provides new insight into the functional significance of rRNA m6 A.


Subject(s)
Methyltransferases , Mouse Embryonic Stem Cells , Animals , Cell Differentiation/genetics , Methyltransferases/genetics , Mice , RNA, Messenger , RNA, Ribosomal, 18S/genetics
13.
J Cell Mol Med ; 25(23): 10879-10891, 2021 12.
Article in English | MEDLINE | ID: mdl-34716659

ABSTRACT

N6 -methyladenosine (m6 A) is the most prevalent modification in mRNA and engages in multiple biological processes. Previous studies indicated that m6 A methyltransferase METTL3 ('writer') and demethylase FTO ('eraser') play critical roles in heart-related disease. However, in the heart, the function of m6 A 'reader', such as YTH (YT521-B homology) domain-containing proteins remains unclear. Here, we report that the defect in YTHDC1 but not other YTH family members contributes to dilated cardiomyopathy (DCM) in mice. Cardiac-specific conditional Ythdc1 knockout led to obvious left ventricular chamber enlargement and severe systolic dysfunction. YTHDC1 deficiency also resulted in the decrease of cardiomyocyte contractility and disordered sarcomere arrangement. By means of integrating multiple high-throughput sequence technologies, including m6 A-MeRIP, RIP-seq and mRNA-seq, we identified 42 transcripts as potential downstream targets of YTHDC1. Amongst them, we found that Titin mRNA was decorated with m6 A modification and depletion of YTHDC1 resulted in aberrant splicing of Titin. Our study suggests that Ythdc1 plays crucial role in regulating the normal contractile function and the development of DCM. These findings clarify the essential role of m6 A reader in cardiac biofunction and provide a novel potential target for the treatment of DCM.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Methyltransferases/metabolism , Nerve Tissue Proteins/metabolism , Protein Kinases/metabolism , RNA Splicing Factors/metabolism , Adenosine/metabolism , Animals , Connectin/metabolism , Male , Mice , RNA-Binding Proteins/metabolism , Serine-Arginine Splicing Factors/metabolism
14.
J Am Chem Soc ; 143(29): 10902-10909, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34254797

ABSTRACT

The asymmetric hydroboration of alkenes has proven to be among the most powerful methods for the synthesis of chiral boron compounds. This protocol is well suitable for activated alkenes such as vinylarenes and alkenes bearing directing groups. However, the catalytic enantioselective hydroboration of O-substituted alkenes has remained unprecedented. Here we report a Rh-catalyzed enantioselective hydroboration of silyl enol ethers (SEEs) that utilizes two new chiral phosphine ligands we developed. This approach features mild reaction conditions and a broad substrate scope as well as excellent functional group tolerance, and enables highly efficient preparation of synthetically valuable chiral borylethers.

15.
Biochem Biophys Res Commun ; 578: 142-149, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34562654

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) acts as a central regulator of metabolic pathways that drive cellular growth. Abnormal activation of mTORC1 occurs at high frequency in human and mouse hepatocellular carcinoma (HCC). DEP domain-containing protein 5 (DEPDC5), a component of GATOR1 complex, is a repressor of amino acid-sensing branch of the mTORC1 pathway. In the current study, we found that persistent activation of hepatic mTORC1 signaling caused by Depdc5 ablation was sufficient to induce a pathological program of liver damage, inflammation and fibrosis that triggers spontaneous HCC development. Take advantage of the combinatory treatment with a single dose of diethylnitrosamine (DEN) and chronic feeding with high-fat diet (HFD), we demonstrated that hepatic depdc5 deletion did not aggravate DEN&HFD induced liver tumorigenesis, probably due to its protective effects on diet-induced liver steatosis. In addition, we further showed that chronic rapamycin treatment did not have any apparent tumor-suppressing effects on DEN&HFD treated control mice, whereas it dramatically reduced the tumor burden in mice with hepatic Depdc5 ablation. This study provides the novel in vivo evidence for Depdc5 deletion mediated mTORC1 hyperactivation in liver tumorigenesis caused by aging or DEN&HFD treatment. Moreover, our findings also propose that pharmacological inhibition of mTORC1 signaling maybe a promising strategy to treat HCC patients with mutations in DEPDC5 gene.


Subject(s)
Carcinoma, Hepatocellular/pathology , Diet, High-Fat , Diethylnitrosamine/toxicity , Fatty Liver/pathology , GTPase-Activating Proteins/physiology , Liver/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Alkylating Agents/toxicity , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Disease Models, Animal , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/metabolism , Female , Inflammation/immunology , Inflammation/metabolism , Liver/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Tumor Burden
16.
Biochem Biophys Res Commun ; 569: 118-124, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34243067

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1) is a crucial regulator of adipogenesis and systemic energy metabolism. Its dysregulation leads to a diversity of metabolic diseases, including obesity and type 2 diabetes. DEP-domain containing 5 (DEPDC5) is a critical component of GATOR1 complex that functions as a key inhibitor of mTORC1. So far, its function in adipose tissue remains largely unknown. Herein we evaluated how persistent mTORC1 activation in adipocyte via Depdc5 knockout modulates adiposity in vivo. Our data indicated that adipocyte-specific knockout of Depdc5 in aged mice led to reduced visceral fat, aggravated insulin resistance and enhanced adipose tissue inflammation. Moreover, we found that Depdc5 ablation resulted in upregulation of adipose triglyceride lipase (ATGL) in adipocytes and elevated levels of serum free fatty acids (FFAs). Intriguingly, rapamycin treatment did not reverse insulin resistance but alleviated adipose tissue inflammation caused by Depdc5 deletion. Taken together, our findings revealed that mTORC1 activation caused by Depdc5 deletion promotes lipolysis process and further exacerbates insulin resistance and adipose tissue inflammation in mice.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , GTPase-Activating Proteins/genetics , Inflammation/genetics , Insulin Resistance/genetics , Adipogenesis/genetics , Adipose Tissue/pathology , Age Factors , Animals , Blotting, Western , Diet, High-Fat/adverse effects , GTPase-Activating Proteins/deficiency , Inflammation/metabolism , Intra-Abdominal Fat/metabolism , Lipase/genetics , Lipase/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Knockout , Mice, Transgenic , Obesity/etiology , Obesity/genetics , Obesity/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics
17.
Nat Chem Biol ; 15(5): 549, 2019 05.
Article in English | MEDLINE | ID: mdl-30737498

ABSTRACT

In the version of this article originally published, the references were incorrectly re-ordered during production. The hyphen in "N6-methyladenosine" in the title was also superscript. The errors have been corrected in the HTML and PDF versions of the paper.

18.
Nat Chem Biol ; 15(1): 88-94, 2019 01.
Article in English | MEDLINE | ID: mdl-30531910

ABSTRACT

N6-Methyladenosine (m6A) RNA modification is present in messenger RNAs (mRNA), ribosomal RNAs (rRNA), and spliceosomal RNAs (snRNA) in humans. Although mRNA m6A modifications have been extensively studied and shown to play critical roles in many cellular processes, the identity of m6A methyltransferases for rRNAs and the function of rRNA m6A modifications are unknown. Here we report a new m6A methyltransferase, ZCCHC4, which primarily methylates human 28S rRNA and also interacts with a subset of mRNAs. ZCCHC4 knockout eliminates m6A4220 modification in 28S rRNA, reduces global translation, and inhibits cell proliferation. We also find that ZCCHC4 protein is overexpressed in hepatocellular carcinoma tumors, and ZCCHC4 knockout significantly reduces tumor size in a xenograft mouse model. Our results highlight the functional significance of an rRNA m6A modification in translation and in tumor biology.


Subject(s)
Adenosine/analogs & derivatives , Liver Neoplasms/metabolism , Methyltransferases/metabolism , RNA, Ribosomal, 28S/metabolism , Adenosine/genetics , Adenosine/metabolism , Animals , Cell Proliferation , Humans , Liver Neoplasms/pathology , Male , Methylation , Methyltransferases/genetics , Mice, Inbred BALB C , Protein Biosynthesis , Xenograft Model Antitumor Assays
19.
Pharmacol Res ; 169: 105642, 2021 07.
Article in English | MEDLINE | ID: mdl-33933636

ABSTRACT

Anthracyclines including doxorubicin (DOX) are still the most widely used and efficacious antitumor drugs, although their cardiotoxicity is a significant cause of heart failure. Despite considerable efforts being made to minimize anthracycline-induced cardiac adverse effects, little progress has been achieved. In this study, we aimed to explore the role and underlying mechanism of SNX17 in DOX-induced cardiotoxicity. We found that SNX17 was downregulated in cardiomyocytes treated with DOX both in vitro and in vivo. DOX treatment combined with SNX17 interference worsened the damage to neonatal rat ventricular myocytes (NRVMs). Furthermore, the rats with SNX17 deficiency manifested increased susceptibility to DOX-induced cardiotoxicity (myocardial damage and fibrosis, impaired contractility and cardiac death). Mechanistic investigation revealed that SNX17 interacted with leiomodin-2 (LMOD2), a key regulator of the thin filament length in muscles, via its C-TERM domain and SNX17 deficiency exacerbated DOX-induced cardiac systolic dysfunction by promoting aberrant LMOD2 degradation through lysosomal pathway. In conclusion, these findings highlight that SNX17 plays a protective role in DOX-induced cardiotoxicity, which provides an attractive target for the prevention and treatment of anthracycline induced cardiotoxicity.


Subject(s)
Cardiotoxins/toxicity , Doxorubicin/toxicity , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/drug effects , Sorting Nexins/metabolism , Animals , Blotting, Western , Cardiotoxins/antagonists & inhibitors , Doxorubicin/antagonists & inhibitors , Fluorescent Antibody Technique , HEK293 Cells , Humans , Immunoprecipitation , Male , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Sorting Nexins/physiology
20.
Genes Dev ; 26(12): 1364-75, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22713873

ABSTRACT

The histone H3 Lys 27 (H3K27) demethylase JMJD3 has been shown to play important roles in transcriptional regulation and cell differentiation. However, the mechanism underlying JMJD3-mediated transcriptional regulation remains incompletely understood. Here we show that JMJD3 is associated with KIAA1718, whose substrates include dimethylated H3K27 (H3K27me2), and proteins involved in transcriptional elongation. JMJD3 and KIAA1718 directly bind to and regulate the expression of a plethora of common target genes in both a demethylase activity-dependent and -independent manner in the human promyelocytic leukemia cell line HL-60. We found that JMJD3 and KIAA1718 collaborate to demethylate trimethylated H3K27 (H3K27me3) on a subset of their target genes, some of which are bivalently marked by H3K4me3 and H3K27me3 and associated with promoter-proximal, paused RNA polymerase II (Pol II) before activation. Reduction of either JMJD3 or KIAA1718 diminishes Pol II traveling along the gene bodies of the affected genes while having no effect on the promoter-proximal Pol II. Furthermore, JMJD3 and KIAA1718 also play a role in localizing elongation factors SPT6 and SPT16 to the target genes. Our results support the model whereby JMJD3 activates bivalent gene transcription by demethylating H3K27me3 and promoting transcriptional elongation. Taken together, these findings provide new insight into the mechanisms by which JMJD3 regulates gene expression.


Subject(s)
Gene Expression Regulation, Neoplastic , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine/metabolism , Transcription, Genetic , Binding Sites , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Gene Expression Regulation, Neoplastic/drug effects , HL-60 Cells , Humans , Macrophages/cytology , Methylation/drug effects , Models, Biological , Phenotype , RNA Polymerase II/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/metabolism , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL