Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 184(7): 1865-1883.e20, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33636127

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of the ongoing coronavirus disease 2019 (COVID-19) pandemic. Understanding of the RNA virus and its interactions with host proteins could improve therapeutic interventions for COVID-19. By using icSHAPE, we determined the structural landscape of SARS-CoV-2 RNA in infected human cells and from refolded RNAs, as well as the regulatory untranslated regions of SARS-CoV-2 and six other coronaviruses. We validated several structural elements predicted in silico and discovered structural features that affect the translation and abundance of subgenomic viral RNAs in cells. The structural data informed a deep-learning tool to predict 42 host proteins that bind to SARS-CoV-2 RNA. Strikingly, antisense oligonucleotides targeting the structural elements and FDA-approved drugs inhibiting the SARS-CoV-2 RNA binding proteins dramatically reduced SARS-CoV-2 infection in cells derived from human liver and lung tumors. Our findings thus shed light on coronavirus and reveal multiple candidate therapeutics for COVID-19 treatment.


Subject(s)
COVID-19 Drug Treatment , Drug Repositioning , RNA, Viral , RNA-Binding Proteins/antagonists & inhibitors , SARS-CoV-2 , Animals , Cell Line , Chlorocebus aethiops , Deep Learning , Humans , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA-Binding Proteins/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/genetics
2.
Mol Cell ; 82(6): 1089-1106.e12, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35231400

ABSTRACT

The recruitment of signaling proteins into activated receptor tyrosine kinases (RTKs) to produce rapid, high-fidelity downstream response is exposed to the ambiguity of random diffusion to the target site. Liquid-liquid phase separation (LLPS) overcomes this by providing elevated, localized concentrations of the required proteins while impeding competitor ligands. Here, we show a subset of phosphorylation-dependent RTK-mediated LLPS states. We then investigate the formation of phase-separated droplets comprising a ternary complex including the RTK, (FGFR2); the phosphatase, SHP2; and the phospholipase, PLCγ1, which assembles in response to receptor phosphorylation. SHP2 and activated PLCγ1 interact through their tandem SH2 domains via a previously undescribed interface. The complex of FGFR2 and SHP2 combines kinase and phosphatase activities to control the phosphorylation state of the assembly while providing a scaffold for active PLCγ1 to facilitate access to its plasma membrane substrate. Thus, LLPS modulates RTK signaling, with potential consequences for therapeutic intervention.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Signal Transduction , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Tyrosine/metabolism , src Homology Domains
3.
Cell ; 157(6): 1253-1254, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24906143

ABSTRACT

The generation of piRNAs from long primary transcripts requires specialized factors that distinguish these precursors from canonical RNA polymerase II transcripts. Mohn et al. and Zhang et al. provide evidence that in Drosophila melanogaster noncanonical transcription coupled with splicing inhibition differentiates piRNA precursors from mRNAs and ensures their correct processing.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Microtubule-Associated Proteins/metabolism , RNA Splicing , RNA, Small Interfering/genetics , RNA-Binding Proteins/metabolism , Transcription, Genetic , Animals , Female
4.
Mol Cell ; 80(6): 1067-1077.e5, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33259809

ABSTRACT

The Coronaviridae is a family of positive-strand RNA viruses that includes SARS-CoV-2, the etiologic agent of the COVID-19 pandemic. Bearing the largest single-stranded RNA genomes in nature, coronaviruses are critically dependent on long-distance RNA-RNA interactions to regulate the viral transcription and replication pathways. Here we experimentally mapped the in vivo RNA-RNA interactome of the full-length SARS-CoV-2 genome and subgenomic mRNAs. We uncovered a network of RNA-RNA interactions spanning tens of thousands of nucleotides. These interactions reveal that the viral genome and subgenomes adopt alternative topologies inside cells and engage in different interactions with host RNAs. Notably, we discovered a long-range RNA-RNA interaction, the FSE-arch, that encircles the programmed ribosomal frameshifting element. The FSE-arch is conserved in the related MERS-CoV and is under purifying selection. Our findings illuminate RNA structure-based mechanisms governing replication, discontinuous transcription, and translation of coronaviruses and will aid future efforts to develop antiviral strategies.


Subject(s)
COVID-19/metabolism , Frameshifting, Ribosomal , Genome, Viral/physiology , RNA, Viral/biosynthesis , SARS-CoV-2/physiology , Virus Replication/physiology , Animals , COVID-19/genetics , Chlorocebus aethiops , Humans , Protein Biosynthesis , RNA, Viral/genetics , Transcription, Genetic , Vero Cells
5.
Mol Cell ; 78(5): 862-875.e8, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32348780

ABSTRACT

Nuclear RNA interference (RNAi) pathways work together with histone modifications to regulate gene expression and enact an adaptive response to transposable RNA elements. In the germline, nuclear RNAi can lead to trans-generational epigenetic inheritance (TEI) of gene silencing. We identified and characterized a family of nuclear Argonaute-interacting proteins (ENRIs) that control the strength and target specificity of nuclear RNAi in C. elegans, ensuring faithful inheritance of epigenetic memories. ENRI-1/2 prevent misloading of the nuclear Argonaute NRDE-3 with small RNAs that normally effect maternal piRNAs, which prevents precocious nuclear translocation of NRDE-3 in the early embryo. Additionally, they are negative regulators of nuclear RNAi triggered from exogenous sources. Loss of ENRI-3, an unstable protein expressed mostly in the male germline, misdirects the RNAi response to transposable elements and impairs TEI. The ENRIs determine the potency and specificity of nuclear RNAi responses by gating small RNAs into specific nuclear Argonautes.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Gene Silencing/physiology , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cell Nucleus/metabolism , Germ Cells/metabolism , Nuclear Proteins/metabolism , RNA Interference/physiology , RNA, Double-Stranded/metabolism , RNA, Nuclear/metabolism , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics
6.
Cell ; 150(1): 88-99, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22738725

ABSTRACT

Transgenerational effects have wide-ranging implications for human health, biological adaptation, and evolution; however, their mechanisms and biology remain poorly understood. Here, we demonstrate that a germline nuclear small RNA/chromatin pathway can maintain stable inheritance for many generations when triggered by a piRNA-dependent foreign RNA response in C. elegans. Using forward genetic screens and candidate approaches, we find that a core set of nuclear RNAi and chromatin factors is required for multigenerational inheritance of environmental RNAi and piRNA silencing. These include a germline-specific nuclear Argonaute HRDE1/WAGO-9, a HP1 ortholog HPL-2, and two putative histone methyltransferases, SET-25 and SET-32. piRNAs can trigger highly stable long-term silencing lasting at least 20 generations. Once established, this long-term memory becomes independent of the piRNA trigger but remains dependent on the nuclear RNAi/chromatin pathway. Our data present a multigenerational epigenetic inheritance mechanism induced by piRNAs.


Subject(s)
Caenorhabditis elegans/genetics , Epigenomics , RNA Interference , RNA, Helminth/metabolism , RNA, Small Interfering/metabolism , Animals , Caenorhabditis elegans/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Female , Germ Cells/metabolism , Male , Transgenes
7.
Mol Cell ; 74(3): 598-608.e6, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31051140

ABSTRACT

RNA flow between organisms has been documented within and among different kingdoms of life. Recently, we demonstrated horizontal RNA transfer between honeybees involving secretion and ingestion of worker and royal jellies. However, how the jelly facilitates transfer of RNA is still unknown. Here, we show that worker and royal jellies harbor robust RNA-binding activity. We report that a highly abundant jelly component, major royal jelly protein 3 (MRJP-3), acts as an extracellular non-sequence-specific RNA-aggregating factor. Multivalent RNA binding stimulates higher-order assembly of MRJP-3 into extracellular ribonucleoprotein granules that protect RNA from degradation and enhance RNA bioavailability. These findings reveal that honeybees have evolved a secreted dietary RNA-binding factor to concentrate, stabilize, and share RNA among individuals. Our work identifies high-order ribonucleoprotein assemblies with functions outside cells and organisms.


Subject(s)
Bees/genetics , Fatty Acids/genetics , Gene Transfer, Horizontal/genetics , Glycoproteins/genetics , Insect Proteins/genetics , Animals , Fatty Acids/biosynthesis , Phase Transition , RNA/genetics , RNA Transport/genetics , RNA-Binding Proteins/genetics
8.
Genes Dev ; 33(1-2): 90-102, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30567997

ABSTRACT

Piwi-interacting RNAs (piRNAs) engage Piwi proteins to suppress transposons and nonself nucleic acids and maintain genome integrity and are essential for fertility in a variety of organisms. In Caenorhabditis elegans, most piRNA precursors are transcribed from two genomic clusters that contain thousands of individual piRNA transcription units. While a few genes have been shown to be required for piRNA biogenesis, the mechanism of piRNA transcription remains elusive. Here we used functional proteomics approaches to identify an upstream sequence transcription complex (USTC) that is essential for piRNA biogenesis. The USTC contains piRNA silencing-defective 1 (PRDE-1), SNPC-4, twenty-one-U fouled-up 4 (TOFU-4), and TOFU-5. The USTC forms unique piRNA foci in germline nuclei and coats the piRNA cluster genomic loci. USTC factors associate with the Ruby motif just upstream of type I piRNA genes. USTC factors are also mutually dependent for binding to the piRNA clusters and forming the piRNA foci. Interestingly, USTC components bind differentially to piRNAs in the clusters and other noncoding RNA genes. These results reveal the USTC as a striking example of the repurposing of a general transcription factor complex to aid in genome defense against transposons.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Gene Expression Regulation/genetics , RNA, Small Interfering/genetics , Amino Acid Motifs , Animals , Caenorhabditis elegans Proteins/genetics , Genome, Helminth/genetics , Protein Binding , Proteomics , RNA, Small Interfering/biosynthesis
9.
Nat Rev Mol Cell Biol ; 15(8): 525-35, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25053358

ABSTRACT

Small RNAs that function in a non-cell autonomous manner are becoming increasingly recognized as regulatory molecules with the potential to transmit information between cells, organisms and species. In plants and nematodes, small RNA mobility can be genetically dissected to provide information about the nature of the mobile RNA species, their distribution in the organism and inside cells, as well as the cellular machinery required for mobility, including channel proteins and cellular trafficking factors. Mobile RNAs function in antiviral defence, cell signalling and gene expression regulation, and might also mediate transgenerational epigenetic inheritance.


Subject(s)
RNA, Small Untranslated/metabolism , Animals , Biological Transport , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Epigenesis, Genetic , Humans , Immunity/genetics , Plants/genetics , Plants/metabolism , RNA, Small Interfering , RNA, Small Untranslated/genetics , Signal Transduction/genetics , Virus Diseases/genetics , Virus Diseases/immunology
10.
Nucleic Acids Res ; 2024 May 29.
Article in English | MEDLINE | ID: mdl-38808663

ABSTRACT

pre-mRNA splicing is a critical feature of eukaryotic gene expression. Both cis- and trans-splicing rely on accurately recognising splice site sequences by spliceosomal U snRNAs and associated proteins. Spliceosomal snRNAs carry multiple RNA modifications with the potential to affect different stages of pre-mRNA splicing. Here, we show that the conserved U6 snRNA m6A methyltransferase METT-10 is required for accurate and efficient cis- and trans-splicing of C. elegans pre-mRNAs. The absence of METT-10 in C. elegans and METTL16 in humans primarily leads to alternative splicing at 5' splice sites with an adenosine at +4 position. In addition, METT-10 is required for splicing of weak 3' cis- and trans-splice sites. We identified a significant overlap between METT-10 and the conserved splicing factor SNRNP27K in regulating 5' splice sites with +4A. Finally, we show that editing endogenous 5' splice site +4A positions to +4U restores splicing to wild-type positions in a mett-10 mutant background, supporting a direct role for U6 snRNA m6A modification in 5' splice site recognition. We conclude that the U6 snRNA m6A modification is important for accurate and efficient pre-mRNA splicing.

11.
Proc Natl Acad Sci U S A ; 120(5): e2217992120, 2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36689659

ABSTRACT

SWItch/sucrose non-fermenting (SWI/SNF) complexes are a family of chromatin remodelers that are conserved across eukaryotes. Mutations in subunits of SWI/SNF cause a multitude of different developmental disorders in humans, most of which have no current treatment options. Here, we identify an alanine-to-valine-causing mutation in the SWI/SNF subunit snfc-5 (SMARCB1 in humans) that prevents embryonic lethality in Caenorhabditis elegans nematodes harboring a loss-of-function mutation in the SWI/SNF subunit swsn-1 (SMARCC1/2 in humans). Furthermore, we found that the combination of this specific mutation in snfc-5 and a loss-of-function mutation in either of the E3 ubiquitin ligases ubr-5 (UBR5 in humans) or hecd-1 (HECTD1 in humans) can restore development to adulthood in swsn-1 loss-of-function mutants that otherwise die as embryos. Using these mutant models, we established a set of 335 genes that are dysregulated in SWI/SNF mutants that arrest their development embryonically but exhibit near wild-type levels of expression in the presence of suppressor mutations that prevent embryonic lethality, suggesting that SWI/SNF promotes development by regulating some subset of these 335 genes. In addition, we show that SWI/SNF protein levels are reduced in swsn-1; snfc-5 double mutants and partly restored to wild-type levels in swsn-1; snfc-5; ubr-5 triple mutants, consistent with a model in which UBR-5 regulates SWI/SNF levels by tagging the complex for proteasomal degradation. Our findings establish a link between two E3 ubiquitin ligases and SWI/SNF function and suggest that UBR5 and HECTD1 could be potential therapeutic targets for the many developmental disorders caused by missense mutations in SWI/SNF subunits.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Humans , Caenorhabditis elegans/metabolism , Ubiquitin-Protein Ligases/metabolism , Caenorhabditis elegans Proteins/metabolism , Ubiquitins/metabolism
12.
Trends Genet ; 38(6): 529-553, 2022 06.
Article in English | MEDLINE | ID: mdl-35307201

ABSTRACT

Transposable element (TE)-derived sequences are ubiquitous in most eukaryotic genomes known to date. Because their expression and mobility can lead to genomic instability, several pathways have evolved to control TEs. Nevertheless, TEs represent an important source of genomic novelty and are often co-opted for novel functions that are relevant for phenotypic divergence and adaptation. Here, we review how animals, in particular vertebrates, mitigate TE mobility and expression, alongside known examples of TE domestication. We argue that the next frontier is to understand the determinants and dynamics of TE domestication: how they shift from 'non-self' targets of epigenetic silencing to 'self' genetic elements. New technologies enable avenues of research that may close the gap between epigenetic silencing and domestication of TEs.


Subject(s)
DNA Transposable Elements , Domestication , Animals , DNA Transposable Elements/genetics , Epigenesis, Genetic/genetics , Eukaryota/genetics , Evolution, Molecular , Vertebrates/genetics
13.
EMBO J ; 40(5): e105565, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33533030

ABSTRACT

PIWI-interacting RNAs (piRNAs) are genome-encoded small RNAs that regulate germ cell development and maintain germline integrity in many animals. Mature piRNAs engage Piwi Argonaute proteins to silence complementary transcripts, including transposable elements and endogenous genes. piRNA biogenesis mechanisms are diverse and remain poorly understood. Here, we identify the RNA polymerase II (RNA Pol II) core subunit RPB-9 as required for piRNA-mediated silencing in the nematode Caenorhabditis elegans. We show that rpb-9 initiates heritable piRNA-mediated gene silencing at two DNA transposon families and at a subset of somatic genes in the germline. We provide genetic and biochemical evidence that RPB-9 is required for piRNA biogenesis by recruiting the Integrator complex at piRNA genes, hence promoting transcriptional termination. We conclude that, as a part of its rapid evolution, the piRNA pathway has co-opted an ancient machinery for high-fidelity transcription.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Gene Expression Regulation , Gene Silencing , RNA Polymerase II/metabolism , RNA, Small Interfering/metabolism , Transcription, Genetic , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Germ Cells , Promoter Regions, Genetic , Protein Subunits , RNA Polymerase II/genetics , RNA, Small Interfering/genetics
14.
EMBO J ; 40(6): e105496, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33283887

ABSTRACT

Methylation of carbon-5 of cytosines (m5 C) is a post-transcriptional nucleotide modification of RNA found in all kingdoms of life. While individual m5 C-methyltransferases have been studied, the impact of the global cytosine-5 methylome on development, homeostasis and stress remains unknown. Here, using Caenorhabditis elegans, we generated the first organism devoid of m5 C in RNA, demonstrating that this modification is non-essential. Using this genetic tool, we determine the localisation and enzymatic specificity of m5 C sites in the RNome in vivo. We find that NSUN-4 acts as a dual rRNA and tRNA methyltransferase in C. elegans mitochondria. In agreement with leucine and proline being the most frequently methylated tRNA isoacceptors, loss of m5 C impacts the decoding of some triplets of these two amino acids, leading to reduced translation efficiency. Upon heat stress, m5 C loss leads to ribosome stalling at UUG triplets, the only codon translated by an m5 C34-modified tRNA. This leads to reduced translation efficiency of UUG-rich transcripts and impaired fertility, suggesting a role of m5 C tRNA wobble methylation in the adaptation to higher temperatures.


Subject(s)
5-Methylcytosine/metabolism , Adaptation, Physiological/genetics , Caenorhabditis elegans/genetics , Heat-Shock Response/genetics , RNA Processing, Post-Transcriptional/genetics , Animals , CRISPR-Cas Systems/genetics , Caenorhabditis elegans/physiology , Cytosine/chemistry , Gene Editing , Hot Temperature , Leucine/chemistry , Methyltransferases/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Proline/chemistry , Protein Biosynthesis/genetics , Protein Biosynthesis/physiology , RNA/chemistry , RNA/genetics , Ribosomes/metabolism
15.
Bioinformatics ; 40(4)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38597883

ABSTRACT

SUMMARY: RNA (ribonucleic acid) molecules have secondary and tertiary structures in vivo which play a crucial role in cellular processes such as the regulation of gene expression, RNA processing and localization. The ability to investigate these structures will enhance our understanding of their function and contribute to the diagnosis and treatment of diseases caused by RNA dysregulation. However, there are no mature pipelines or packages for processing and analyzing complex in vivo RNA structural data. Here, we present rnaCrosslinkOO (RNA Crosslink Object-Oriented), a novel software package for the comprehensive analysis of data derived from the COMRADES (Crosslinking of Matched RNA and Deep Sequencing) method. rnaCrosslinkOO offers a comprehensive pipeline from raw sequencing reads to the identification and comparison of RNA structural features. It includes read processing and alignment, clustering of duplexes, data exploration, folding and comparisons of RNA structures. rnaCrosslinkOO also enables comparisons between conditions, the identification of inter-RNA interactions, and the incorporation of reactivity data to improve structure prediction. AVAILABILITY AND IMPLEMENTATION: rnaCrosslinkOO is freely available to noncommercial users and implemented in R, with the source code and documentation accessible at https://CRAN.R-project.org/package=rnaCrosslinkOO. The software is supported on Linux, macOS, and Windows platforms.


Subject(s)
Nucleic Acid Conformation , RNA , Sequence Analysis, RNA , Software , RNA/chemistry , Sequence Analysis, RNA/methods , High-Throughput Nucleotide Sequencing/methods
16.
EMBO J ; 39(23): e104579, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33034389

ABSTRACT

Environmental factors can change phenotypes in exposed individuals and offspring and involve the germline, likely via biological signals in the periphery that communicate with germ cells. Here, using a mouse model of paternal exposure to traumatic stress, we identify circulating factors involving peroxisome proliferator-activated receptor (PPAR) pathways in the effects of exposure to the germline. We show that exposure alters metabolic functions and pathways, particularly lipid-derived metabolites, in exposed fathers and their offspring. We collected data in a human cohort exposed to childhood trauma and observed similar metabolic alterations in circulation, suggesting conserved effects. Chronic injection of serum from trauma-exposed males into controls recapitulates metabolic phenotypes in the offspring. We identify lipid-activated nuclear receptors PPARs as potential mediators of the effects from father to offspring. Pharmacological PPAR activation in vivo reproduces metabolic dysfunctions in the offspring and grand-offspring of injected males and affects the sperm transcriptome in fathers and sons. In germ-like cells in vitro, both serum and PPAR agonist induce PPAR activation. Together, these results highlight the role of circulating factors as potential communication vectors between the periphery and the germline.


Subject(s)
Germ Cells/metabolism , Paternal Exposure , Animals , Blood , Epigenesis, Genetic , Epigenomics , Fathers , Humans , Male , Mice , Mice, Inbred C57BL , Phenotype , Reproduction/physiology , Spermatozoa , Transcriptome , Wounds and Injuries
17.
Genome Res ; 31(12): 2290-2302, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34772700

ABSTRACT

Arbuscular mycorrhizal (AM) fungi form mutualistic relationships with most land plant species. AM fungi have long been considered as ancient asexuals. Long-term clonal evolution would be remarkable for a eukaryotic lineage and suggests the importance of alternative mechanisms to promote genetic variability facilitating adaptation. Here, we assessed the potential of transposable elements for generating such genomic diversity. The dynamic expression of TEs during Rhizophagus irregularis spore development suggests ongoing TE activity. We find Mutator-like elements located near genes belonging to highly expanded gene families. Whole-genome epigenomic profiling of R. irregularis provides direct evidence of DNA methylation and small RNA production occurring at TE loci. Our results support a model in which TE activity shapes the genome, while DNA methylation and small RNA-mediated silencing keep their overproliferation in check. We propose that a well-controlled TE activity directly contributes to genome evolution in AM fungi.

18.
RNA ; 28(3): 353-370, 2022 03.
Article in English | MEDLINE | ID: mdl-34949722

ABSTRACT

The human terminal uridyl transferases TUT4 and TUT7 (TUT4/7) catalyze the additions of uridines at the 3' end of RNAs, including the precursors of the tumor suppressor miRNA let-7 upon recruitment by the oncoprotein LIN28A. As a consequence, let-7 family miRNAs are down-regulated. Disruption of this TUT4/7 activity inhibits tumorigenesis. Hence, targeting TUT4/7 could be a potential anticancer therapy. In this study, we investigate TUT4/7-mediated RNA regulation in two cancer cell lines by establishing catalytic knockout models. Upon TUT4/7 mutation, we observe a significant reduction in miRNA uridylation, which results in defects in cancer cell properties such as cell proliferation and migration. With the loss of TUT4/7-mediated miRNA uridylation, the uridylated miRNA variants are replaced by adenylated isomiRs. Changes in miRNA modification profiles are accompanied by deregulation of expression levels in specific cases. Unlike let-7s, most miRNAs do not depend on LIN28A for TUT4/7-mediated regulation. Additionally, we identify TUT4/7-regulated cell-type-specific miRNA clusters and deregulation in their corresponding mRNA targets. Expression levels of miR-200c-3p and miR-141-3p are regulated by TUT4/7 in a cancer cell-type-specific manner. Subsequently, BCL2, which is a well-established target of miR-200c is up-regulated. Therefore, TUT4/7 loss causes deregulation of miRNA-mRNA networks in a cell-type-specific manner. Understanding of the underlying biology of such cell-type-specific deregulation will be an important aspect of targeting TUT4/7 for potential cancer therapies.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Neoplasms/metabolism , RNA Nucleotidyltransferases/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , MicroRNAs/genetics , Neoplasms/genetics , RNA Nucleotidyltransferases/genetics , RNA Processing, Post-Transcriptional
19.
Mol Syst Biol ; 19(11): e11835, 2023 Nov 09.
Article in English | MEDLINE | ID: mdl-37850520

ABSTRACT

How do the same mechanisms that faithfully regenerate complex developmental programmes in spite of environmental and genetic perturbations also allow responsiveness to environmental signals, adaptation and genetic evolution? Using the nematode Caenorhabditis elegans as a model, we explore the phenotypic space of growth and development in various genetic and environmental contexts. Our data are growth curves and developmental parameters obtained by automated microscopy. Using these, we show that among the traits that make up the developmental space, correlations within a particular context are predictive of correlations among different contexts. Furthermore, we find that the developmental variability of this animal can be captured on a relatively low-dimensional phenotypic manifold and that on this manifold, genetic and environmental contributions to plasticity can be deconvolved independently. Our perspective offers a new way of understanding the relationship between robustness and flexibility in complex systems, suggesting that projection and concentration of dimension can naturally align these forces as complementary rather than competing.


Subject(s)
Caenorhabditis elegans , Evolution, Molecular , Animals , Caenorhabditis elegans/genetics , Phenotype
20.
Nucleic Acids Res ; 50(11): e64, 2022 06 24.
Article in English | MEDLINE | ID: mdl-35234904

ABSTRACT

Most genomes harbor a large number of transposons, and they play an important role in evolution and gene regulation. They are also of interest to clinicians as they are involved in several diseases, including cancer and neurodegeneration. Although several methods for transposon identification are available, they are often highly specialised towards specific tasks or classes of transposons, and they lack common standards such as a unified taxonomy scheme and output file format. We present TransposonUltimate, a powerful bundle of three modules for transposon classification, annotation, and detection of transposition events. TransposonUltimate comes as a Conda package under the GPL-3.0 licence, is well documented and it is easy to install through https://github.com/DerKevinRiehl/TransposonUltimate. We benchmark the classification module on the large TransposonDB covering 891,051 sequences to demonstrate that it outperforms the currently best existing solutions. The annotation and detection modules combine sixteen existing softwares, and we illustrate its use by annotating Caenorhabditis elegans, Rhizophagus irregularis and Oryza sativa subs. japonica genomes. Finally, we use the detection module to discover 29 554 transposition events in the genomes of 20 wild type strains of C. elegans. Databases, assemblies, annotations and further findings can be downloaded from (https://doi.org/10.5281/zenodo.5518085).


Subject(s)
DNA Transposable Elements , Software , Animals , Benchmarking , Caenorhabditis elegans/genetics , Fungi/genetics , Genome , Molecular Sequence Annotation , Oryza/genetics , Reference Standards
SELECTION OF CITATIONS
SEARCH DETAIL