Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Cancer Discov ; 13(10): 2228-2247, 2023 10 05.
Article En | MEDLINE | ID: mdl-37548590

Therapies that enhance antitumor immunity have altered the natural history of many cancers. Consequently, leveraging nonoverlapping mechanisms to increase immunogenicity of cancer cells remains a priority. Using a novel enzymatic inhibitor of the RNA methyl-transferase METTL3, we demonstrate a global decrease in N6-methyladenosine (m6A) results in double-stranded RNA (dsRNA) formation and a profound cell-intrinsic interferon response. Through unbiased CRISPR screens, we establish dsRNA-sensing and interferon signaling are primary mediators that potentiate T-cell killing of cancer cells following METTL3 inhibition. We show in a range of immunocompetent mouse models that although METTL3 inhibition is equally efficacious to anti-PD-1 therapy, the combination has far greater preclinical activity. Using SPLINTR barcoding, we demonstrate that anti-PD-1 therapy and METTL3 inhibition target distinct malignant clones, and the combination of these therapies overcomes clones insensitive to the single agents. These data provide the mole-cular and preclinical rationale for employing METTL3 inhibitors to promote antitumor immunity in the clinic. SIGNIFICANCE: This work demonstrates that METTL3 inhibition stimulates a cell-intrinsic interferon response through dsRNA formation. This immunomodulatory mechanism is distinct from current immunotherapeutic agents and provides the molecular rationale for combination with anti-PD-1 immune-checkpoint blockade to augment antitumor immunity. This article is featured in Selected Articles from This Issue, p. 2109.


Interferons , Methyltransferases , Animals , Mice , Interferons/genetics , Methyltransferases/genetics , Methyltransferases/metabolism , RNA, Double-Stranded
3.
Mol Ther Nucleic Acids ; 27: 1092-1102, 2022 Mar 08.
Article En | MEDLINE | ID: mdl-35228902

SINEUPs are a novel class of natural and synthetic non-coding antisense RNA molecules able to increase the translation of a target mRNA. They present a modular organization comprising an unstructured antisense target-specific domain, which sets the specificity of each individual SINEUP, and a structured effector domain, which is responsible for the translation enhancement. In order to design a fully functional in vitro transcribed SINEUP for therapeutics applications, SINEUP RNAs were synthesized in vitro with a variety of chemical modifications and screened for their activity on endogenous target mRNA upon transfection. Three combinations of modified ribonucleotides-2'O methyl-ATP (Am), N6 methyl-ATP (m6A), and pseudo-UTP (ψ)-conferred SINEUP activity to naked RNA. The best combination tested in this study was fully modified with m6A and ψ. Aside from functionality, this combination conferred improved stability upon transfection and higher thermal stability. Common structural determinants of activity were identified by circular dichroisms, defining a core functional structure that is achieved with different combinations of modifications.

4.
Nat Commun ; 13(1): 929, 2022 02 17.
Article En | MEDLINE | ID: mdl-35177605

Many cellular processes, including ribosome biogenesis, are regulated through post-transcriptional RNA modifications. Here, a genome-wide analysis of the human mitochondrial transcriptome shows that 2'-O-methylation is limited to residues of the mitoribosomal large subunit (mtLSU) 16S mt-rRNA, introduced by MRM1, MRM2 and MRM3, with the modifications installed by the latter two proteins being interdependent. MRM2 controls mitochondrial respiration by regulating mitoribosome biogenesis. In its absence, mtLSU particles (visualized by cryo-EM at the resolution of 2.6 Å) present disordered RNA domains, partial occupancy of bL36m and bound MALSU1:L0R8F8:mtACP anti-association module, allowing five mtLSU biogenesis intermediates with different intersubunit interface configurations to be placed along the assembly pathway. However, mitoribosome biogenesis does not depend on the methyltransferase activity of MRM2. Disruption of the MRM2 Drosophila melanogaster orthologue leads to mitochondria-related developmental arrest. This work identifies a key checkpoint during mtLSU assembly, essential to maintain mitochondrial homeostasis.


Drosophila Proteins/metabolism , Methyltransferases/metabolism , Mitochondrial Ribosomes/metabolism , Protein Biosynthesis , Ribosome Subunits, Large/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Gene Knockout Techniques , HEK293 Cells , Humans , Male , Methylation , Methyltransferases/genetics , RNA, Ribosomal, 16S/metabolism , Ribosomal Proteins/metabolism
5.
Mol Cell Proteomics ; 21(2): 100192, 2022 02.
Article En | MEDLINE | ID: mdl-34979241

The amount of any given protein in the brain is determined by the rates of its synthesis and destruction, which are regulated by different cellular mechanisms. Here, we combine metabolic labeling in live mice with global proteomic profiling to simultaneously quantify both the flux and amount of proteins in mouse models of neurodegeneration. In multiple models, protein turnover increases were associated with increasing pathology. This method distinguishes changes in protein expression mediated by synthesis from those mediated by degradation. In the AppNL-F knockin mouse model of Alzheimer's disease, increased turnover resulted from imbalances in both synthesis and degradation, converging on proteins associated with synaptic vesicle recycling (Dnm1, Cltc, Rims1) and mitochondria (Fis1, Ndufv1). In contrast to disease models, aging in wild-type mice caused a widespread decrease in protein recycling associated with a decrease in autophagic flux. Overall, this simple multidimensional approach enables a comprehensive mapping of proteome dynamics and identifies affected proteins in mouse models of disease and other live animal test settings.


Alzheimer Disease , Proteome , Aging , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Mammals/metabolism , Mice , Mice, Transgenic , Proteome/metabolism , Proteomics/methods
6.
Nature ; 593(7860): 597-601, 2021 05.
Article En | MEDLINE | ID: mdl-33902106

N6-methyladenosine (m6A) is an abundant internal RNA modification1,2 that is catalysed predominantly by the METTL3-METTL14 methyltransferase complex3,4. The m6A methyltransferase METTL3 has been linked to the initiation and maintenance of acute myeloid leukaemia (AML), but the potential of therapeutic applications targeting this enzyme remains unknown5-7. Here we present the identification and characterization of STM2457, a highly potent and selective first-in-class catalytic inhibitor of METTL3, and a crystal structure of STM2457 in complex with METTL3-METTL14. Treatment of tumours with STM2457 leads to reduced AML growth and an increase in differentiation and apoptosis. These cellular effects are accompanied by selective reduction of m6A levels on known leukaemogenic mRNAs and a decrease in their expression consistent with a translational defect. We demonstrate that pharmacological inhibition of METTL3 in vivo leads to impaired engraftment and prolonged survival in various mouse models of AML, specifically targeting key stem cell subpopulations of AML. Collectively, these results reveal the inhibition of METTL3 as a potential therapeutic strategy against AML, and provide proof of concept that the targeting of RNA-modifying enzymes represents a promising avenue for anticancer therapy.


Antineoplastic Agents/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Methyltransferases/antagonists & inhibitors , Adenosine/analogs & derivatives , Animals , Apoptosis , Cell Differentiation , Cell Line, Tumor , Female , Gene Expression Regulation, Leukemic/drug effects , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , Xenograft Model Antitumor Assays
8.
Nat Commun ; 11(1): 926, 2020 02 17.
Article En | MEDLINE | ID: mdl-32066737

The field of epitranscriptomics continues to reveal how post-transcriptional modification of RNA affects a wide variety of biological phenomena. A pivotal challenge in this area is the identification of modified RNA residues within their sequence contexts. Mass spectrometry (MS) offers a comprehensive solution by using analogous approaches to shotgun proteomics. However, software support for the analysis of RNA MS data is inadequate at present and does not allow high-throughput processing. Existing software solutions lack the raw performance and statistical grounding to efficiently handle the numerous modifications found on RNA. We present a free and open-source database search engine for RNA MS data, called NucleicAcidSearchEngine (NASE), that addresses these shortcomings. We demonstrate the capability of NASE to reliably identify a wide range of modified RNA sequences in four original datasets of varying complexity. In human tRNA, we characterize over 20 different modification types simultaneously and find many cases of incomplete modification.


Epigenomics/methods , High-Throughput Screening Assays/methods , RNA Processing, Post-Transcriptional/genetics , Search Engine , Tandem Mass Spectrometry/methods , Base Sequence/genetics , Databases, Factual/statistics & numerical data , Datasets as Topic , Humans , Oligonucleotides/chemistry , Oligonucleotides/genetics , Oligonucleotides/metabolism , RNA, Transfer/chemistry , RNA, Transfer/genetics , RNA, Transfer/metabolism , Reproducibility of Results
9.
Nucleic Acids Res ; 47(19): 10267-10281, 2019 11 04.
Article En | MEDLINE | ID: mdl-31665743

Post-transcriptional RNA modifications, the epitranscriptome, play important roles in modulating the functions of RNA species. Modifications of rRNA are key for ribosome production and function. Identification and characterization of enzymes involved in epitranscriptome shaping is instrumental for the elucidation of the functional roles of specific RNA modifications. Ten modified sites have been thus far identified in the mammalian mitochondrial rRNA. Enzymes responsible for two of these modifications have not been characterized. Here, we identify METTL15, show that it is the main N4-methylcytidine (m4C) methyltransferase in human cells and demonstrate that it is responsible for the methylation of position C839 in mitochondrial 12S rRNA. We show that the lack of METTL15 results in a reduction of the mitochondrial de novo protein synthesis and decreased steady-state levels of protein components of the oxidative phosphorylation system. Without functional METTL15, the assembly of the mitochondrial ribosome is decreased, with the late assembly components being unable to be incorporated efficiently into the small subunit. We speculate that m4C839 is involved in the stabilization of 12S rRNA folding, therefore facilitating the assembly of the mitochondrial small ribosomal subunits. Taken together our data show that METTL15 is a novel protein necessary for efficient translation in human mitochondria.


Methyltransferases/genetics , Mitochondria/genetics , Mitochondrial Ribosomes/chemistry , RNA, Ribosomal/genetics , Cytidine/genetics , Humans , Methylation , Mitochondria/chemistry , Oxidative Phosphorylation , Protein Biosynthesis/genetics , RNA Folding/genetics , RNA Processing, Post-Transcriptional/genetics , RNA, Ribosomal/chemistry
10.
Mol Cell ; 74(6): 1278-1290.e9, 2019 06 20.
Article En | MEDLINE | ID: mdl-31031083

7-methylguanosine (m7G) is present at mRNA caps and at defined internal positions within tRNAs and rRNAs. However, its detection within low-abundance mRNAs and microRNAs (miRNAs) has been hampered by a lack of sensitive detection strategies. Here, we adapt a chemical reactivity assay to detect internal m7G in miRNAs. Using this technique (Borohydride Reduction sequencing [BoRed-seq]) alongside RNA immunoprecipitation, we identify m7G within a subset of miRNAs that inhibit cell migration. We show that the METTL1 methyltransferase mediates m7G methylation within miRNAs and that this enzyme regulates cell migration via its catalytic activity. Using refined mass spectrometry methods, we map m7G to a single guanosine within the let-7e-5p miRNA. We show that METTL1-mediated methylation augments let-7 miRNA processing by disrupting an inhibitory secondary structure within the primary miRNA transcript (pri-miRNA). These results identify METTL1-dependent N7-methylation of guanosine as a new RNA modification pathway that regulates miRNA structure, biogenesis, and cell migration.


Guanosine/analogs & derivatives , Methyltransferases/genetics , MicroRNAs/genetics , RNA Processing, Post-Transcriptional , A549 Cells , Base Sequence , Biological Assay , Caco-2 Cells , Cell Movement , Cell Proliferation , Guanosine/metabolism , HEK293 Cells , Humans , Methylation , Methyltransferases/metabolism , MicroRNAs/metabolism , Nucleic Acid Conformation
11.
Cell ; 174(2): 325-337.e14, 2018 07 12.
Article En | MEDLINE | ID: mdl-29887380

Multiple proteins act co-operatively in mammalian clathrin-mediated endocytosis (CME) to generate endocytic vesicles from the plasma membrane. The principles controlling the activation and organization of the actin cytoskeleton during mammalian CME are, however, not fully understood. Here, we show that the protein FCHSD2 is a major activator of actin polymerization during CME. FCHSD2 deletion leads to decreased ligand uptake caused by slowed pit maturation. FCHSD2 is recruited to endocytic pits by the scaffold protein intersectin via an unusual SH3-SH3 interaction. Here, its flat F-BAR domain binds to the planar region of the plasma membrane surrounding the developing pit forming an annulus. When bound to the membrane, FCHSD2 activates actin polymerization by a mechanism that combines oligomerization and recruitment of N-WASP to PI(4,5)P2, thus promoting pit maturation. Our data therefore describe a molecular mechanism for linking spatiotemporally the plasma membrane to a force-generating actin platform guiding endocytic vesicle maturation.


Actin Cytoskeleton/physiology , Carrier Proteins/metabolism , Clathrin/metabolism , Membrane Proteins/metabolism , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Membrane/chemistry , Cell Membrane/metabolism , Clathrin-Coated Vesicles/metabolism , Endocytosis , HeLa Cells , Humans , Liposomes/chemistry , Liposomes/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Microscopy, Fluorescence , Models, Molecular , Mutagenesis, Site-Directed , RNA Interference , RNA, Small Interfering/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/chemistry , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , src Homology Domains
12.
Proc Natl Acad Sci U S A ; 110(47): 18934-9, 2013 Nov 19.
Article En | MEDLINE | ID: mdl-24191001

Mitochondrial respiratory complex I is a product of both the nuclear and mitochondrial genomes. The integration of seven subunits encoded in mitochondrial DNA into the inner membrane, their association with 14 nuclear-encoded membrane subunits, the construction of the extrinsic arm from 23 additional nuclear-encoded proteins, iron-sulfur clusters, and flavin mononucleotide cofactor require the participation of assembly factors. Some are intrinsic to the complex, whereas others participate transiently. The suppression of the expression of the NDUFA11 subunit of complex I disrupted the assembly of the complex, and subcomplexes with masses of 550 and 815 kDa accumulated. Eight of the known extrinsic assembly factors plus a hydrophobic protein, C3orf1, were associated with the subcomplexes. The characteristics of C3orf1, of another assembly factor, TMEM126B, and of NDUFA11 suggest that they all participate in constructing the membrane arm of complex I.


Electron Transport Complex I/metabolism , Gene Expression Regulation/physiology , Membrane Proteins/metabolism , Mitochondrial Membranes/metabolism , Protein Multimerization/physiology , Blotting, Western , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Humans , Mass Spectrometry , Microscopy, Confocal , Mitochondrial Membrane Transport Proteins , Mitochondrial Precursor Protein Import Complex Proteins , Oxygen Consumption/physiology
...