Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.846
Filter
1.
Cell ; 173(4): 839-850.e18, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29628142

ABSTRACT

Maize abnormal chromosome 10 (Ab10) encodes a classic example of true meiotic drive that converts heterochromatic regions called knobs into motile neocentromeres that are preferentially transmitted to egg cells. Here, we identify a cluster of eight genes on Ab10, called the Kinesin driver (Kindr) complex, that are required for both neocentromere motility and preferential transmission. Two meiotic drive mutants that lack neocentromere activity proved to be kindr epimutants with increased DNA methylation across the entire gene cluster. RNAi of Kindr induced a third epimutant and corresponding loss of meiotic drive. Kinesin gliding assays and immunolocalization revealed that KINDR is a functional minus-end-directed kinesin that localizes specifically to knobs containing 180 bp repeats. Sequence comparisons suggest that Kindr diverged from a Kinesin-14A ancestor ∼12 mya and has driven the accumulation of > 500 Mb of knob repeats and affected the segregation of thousands of genes linked to knobs on all 10 chromosomes.


Subject(s)
Centromere/metabolism , Kinesins/metabolism , Meiosis , Plant Proteins/metabolism , Zea mays/metabolism , Centromere/genetics , Chromosomes, Plant , Evolution, Molecular , Haplotypes , In Situ Hybridization, Fluorescence , Kinesins/antagonists & inhibitors , Kinesins/classification , Kinesins/genetics , Models, Genetic , Mutagenesis , Phylogeny , Plant Proteins/antagonists & inhibitors , Plant Proteins/classification , Plant Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Whole Genome Sequencing , Zea mays/genetics
2.
Annu Rev Cell Dev Biol ; 35: 407-431, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31403819

ABSTRACT

A large and significant portion of eukaryotic transcriptomes consists of noncoding RNAs (ncRNAs) that have minimal or no protein-coding capacity but are functional. Diverse ncRNAs, including both small RNAs and long ncRNAs (lncRNAs), play essential regulatory roles in almost all biological processes by modulating gene expression at the transcriptional and posttranscriptional levels. In this review, we summarize the current knowledge of plant small RNAs and lncRNAs, with a focus on their biogenesis, modes of action, local and systemic movement, and functions at the nexus of plant development and environmental responses. The complex connections among small RNAs, lncRNAs, and small peptides in plants are also discussed, along with the challenges of identifying and investigating new classes of ncRNAs.


Subject(s)
Plant Development/genetics , Plants/genetics , RNA, Long Noncoding/metabolism , RNA, Plant/metabolism , Stress, Physiological/genetics , Gene Expression Regulation, Plant/genetics , Gene Expression Regulation, Plant/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , RNA, Long Noncoding/genetics , RNA, Plant/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
3.
Mol Cell ; 84(15): 2918-2934.e11, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39025072

ABSTRACT

The RNA-induced silencing complex (RISC), which powers RNA interference (RNAi), consists of a guide RNA and an Argonaute protein that slices target RNAs complementary to the guide. We find that, for different guide-RNA sequences, slicing rates of perfectly complementary bound targets can be surprisingly different (>250-fold range), and that faster slicing confers better knockdown in cells. Nucleotide sequence identities at guide-RNA positions 7, 10, and 17 underlie much of this variation in slicing rates. Analysis of one of these determinants implicates a structural distortion at guide nucleotides 6-7 in promoting slicing. Moreover, slicing directed by different guide sequences has an unanticipated, 600-fold range in 3'-mismatch tolerance, attributable to guides with weak (AU-rich) central pairing requiring extensive 3' complementarity (pairing beyond position 16) to more fully populate the slicing-competent conformation. Together, our analyses identify sequence determinants of RISC activity and provide biochemical and conformational rationale for their action.


Subject(s)
Argonaute Proteins , Nucleic Acid Conformation , RNA, Guide, CRISPR-Cas Systems , RNA-Induced Silencing Complex , Argonaute Proteins/metabolism , Argonaute Proteins/genetics , Argonaute Proteins/chemistry , Humans , RNA-Induced Silencing Complex/metabolism , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/chemistry , Kinetics , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , RNA Interference , Base Sequence , HEK293 Cells
4.
Mol Cell ; 83(21): 3835-3851.e7, 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37875112

ABSTRACT

PIWI-interacting RNAs (piRNAs) guide transposable element repression in animal germ lines. In Drosophila, piRNAs are produced from heterochromatic loci, called piRNA clusters, which act as information repositories about genome invaders. piRNA generation by dual-strand clusters depends on the chromatin-bound Rhino-Deadlock-Cutoff (RDC) complex, which is deposited on clusters guided by piRNAs, forming a positive feedback loop in which piRNAs promote their own biogenesis. However, how piRNA clusters are formed before cognate piRNAs are present remains unknown. Here, we report spontaneous de novo piRNA cluster formation from repetitive transgenic sequences. Cluster formation occurs over several generations and requires continuous trans-generational maternal transmission of small RNAs. We discovered that maternally supplied small interfering RNAs (siRNAs) trigger de novo cluster activation in progeny. In contrast, siRNAs are dispensable for cluster function after its establishment. These results reveal an unexpected interplay between the siRNA and piRNA pathways and suggest a mechanism for de novo piRNA cluster formation triggered by siRNAs.


Subject(s)
Drosophila Proteins , Piwi-Interacting RNA , Animals , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Maternal Inheritance , Drosophila/genetics , Chromatin/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , DNA Transposable Elements/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism
5.
Mol Cell ; 83(12): 1983-2002.e11, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37295433

ABSTRACT

The evolutionarily conserved minor spliceosome (MiS) is required for protein expression of ∼714 minor intron-containing genes (MIGs) crucial for cell-cycle regulation, DNA repair, and MAP-kinase signaling. We explored the role of MIGs and MiS in cancer, taking prostate cancer (PCa) as an exemplar. Both androgen receptor signaling and elevated levels of U6atac, a MiS small nuclear RNA, regulate MiS activity, which is highest in advanced metastatic PCa. siU6atac-mediated MiS inhibition in PCa in vitro model systems resulted in aberrant minor intron splicing leading to cell-cycle G1 arrest. Small interfering RNA knocking down U6atac was ∼50% more efficient in lowering tumor burden in models of advanced therapy-resistant PCa compared with standard antiandrogen therapy. In lethal PCa, siU6atac disrupted the splicing of a crucial lineage dependency factor, the RE1-silencing factor (REST). Taken together, we have nominated MiS as a vulnerability for lethal PCa and potentially other cancers.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Male , Humans , Introns/genetics , Prostatic Neoplasms/metabolism , RNA Splicing/genetics , Spliceosomes/metabolism , Signal Transduction , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Cell Line, Tumor , Prostatic Neoplasms, Castration-Resistant/genetics
6.
Mol Cell ; 82(7): 1329-1342.e8, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35298909

ABSTRACT

Argonautes are nucleic acid-guided proteins that perform numerous cellular functions across all domains of life. Little is known about how distinct evolutionary pressures have shaped each Argonaute's biophysical properties. We applied high-throughput biochemistry to characterize how Thermus thermophilus Argonaute (TtAgo), a DNA-guided DNA endonuclease, finds, binds, and cleaves its targets. We found that TtAgo uses biophysical adaptations similar to those of eukaryotic Argonautes for rapid association but requires more extensive complementarity to achieve high-affinity target binding. Using these data, we constructed models for TtAgo association rates and equilibrium binding affinities that estimate the nucleic acid- and protein-mediated components of the target interaction energies. Finally, we showed that TtAgo cleavage rates vary widely based on the DNA guide, suggesting that only a subset of guides cleaves targets on physiologically relevant timescales.


Subject(s)
Argonaute Proteins , Thermus thermophilus , Argonaute Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA/genetics , Endonucleases/metabolism , Thermus thermophilus/genetics
7.
Mol Cell ; 81(23): 4826-4842.e8, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34626567

ABSTRACT

In animals, PIWI-interacting RNAs (piRNAs) silence transposons, fight viral infections, and regulate gene expression. piRNA biogenesis concludes with 3' terminal trimming and 2'-O-methylation. Both trimming and methylation influence piRNA stability. Our biochemical data show that multiple mechanisms destabilize unmethylated mouse piRNAs, depending on whether the piRNA 5' or 3' sequence is complementary to a trigger RNA. Unlike target-directed degradation of microRNAs, complementarity-dependent destabilization of piRNAs in mice and flies is blocked by 3' terminal 2'-O-methylation and does not require base pairing to both the piRNA seed and the 3' sequence. In flies, 2'-O-methylation also protects small interfering RNAs (siRNAs) from complementarity-dependent destruction. By contrast, pre-piRNA trimming protects mouse piRNAs from a degradation pathway unaffected by trigger complementarity. In testis lysate and in vivo, internal or 3' terminal uridine- or guanine-rich tracts accelerate pre-piRNA decay. Loss of both trimming and 2'-O-methylation causes the mouse piRNA pathway to collapse, demonstrating that these modifications collaborate to stabilize piRNAs.


Subject(s)
Argonaute Proteins/metabolism , RNA, Small Interfering/metabolism , Animals , Cell Separation , Drosophila melanogaster , Female , Flow Cytometry , Gene Expression , Gene Silencing , Genetic Techniques , Male , Methylation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Processing, Post-Translational , RNA, Double-Stranded , Spermatocytes/metabolism , Spermatogonia/metabolism , Testis/metabolism
8.
Trends Genet ; 40(3): 260-275, 2024 03.
Article in English | MEDLINE | ID: mdl-38296708

ABSTRACT

Intrinsically disordered proteins (IDPs) and proteins with intrinsically disordered regions (IDRs) possess low sequence complexity of amino acids and display non-globular tertiary structures. They can act as scaffolds, form regulatory hubs, or trigger biomolecular condensation to control diverse aspects of biology. Emerging evidence has recently implicated critical roles of IDPs and IDR-contained proteins in nuclear transcription and cytoplasmic post-transcriptional processes, among other molecular functions. We here summarize the concepts and organizing principles of IDPs. We then illustrate recent progress in understanding the roles of key IDPs in machineries that regulate transcriptional and post-transcriptional gene silencing (PTGS) in plants, aiming at highlighting new modes of action of IDPs in controlling biological processes.


Subject(s)
Intrinsically Disordered Proteins , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Plants/genetics , Plants/metabolism , Gene Silencing , Protein Conformation
9.
Mol Cell ; 75(4): 741-755.e11, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31324449

ABSTRACT

Argonaute proteins loaded with microRNAs (miRNAs) or small interfering RNAs (siRNAs) form the RNA-induced silencing complex (RISC), which represses target RNA expression. Predicting the biological targets, specificity, and efficiency of both miRNAs and siRNAs has been hamstrung by an incomplete understanding of the sequence determinants of RISC binding and cleavage. We applied high-throughput methods to measure the association kinetics, equilibrium binding energies, and single-turnover cleavage rates of mouse AGO2 RISC. We find that RISC readily tolerates insertions of up to 7 nt in its target opposite the central region of the guide. Our data uncover specific guide:target mismatches that enhance the rate of target cleavage, suggesting novel siRNA design strategies. Using these data, we derive quantitative models for RISC binding and target cleavage and show that our in vitro measurements and models predict knockdown in an engineered cellular system.


Subject(s)
Argonaute Proteins/chemistry , Models, Chemical , RNA, Small Interfering/chemistry , RNA-Induced Silencing Complex/chemistry , Animals , Mice
10.
EMBO Rep ; 25(7): 2896-2913, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38769420

ABSTRACT

Canonical RNA interference (RNAi) is sequence-specific mRNA degradation guided by small interfering RNAs (siRNAs) made by RNase III Dicer from long double-stranded RNA (dsRNA). RNAi roles include gene regulation, antiviral immunity or defense against transposable elements. In mammals, RNAi is constrained by Dicer's adaptation to produce another small RNA class-microRNAs. However, a truncated Dicer isoform (ΔHEL1) supporting RNAi exists in mouse oocytes. A homozygous mutation to express only the truncated ΔHEL1 variant causes dysregulation of microRNAs and perinatal lethality in mice. Here, we report the phenotype and canonical RNAi activity in DicerΔHEL1/wt mice, which are viable, show minimal miRNome changes, but their endogenous siRNA levels are an order of magnitude higher. We show that siRNA production in vivo is limited by available dsRNA, but not by Protein kinase R, a dsRNA sensor of innate immunity. dsRNA expression from a transgene yields sufficient siRNA levels to induce efficient RNAi in heart and muscle. DicerΔHEL1/wt mice with enhanced canonical RNAi offer a platform for examining potential and limits of mammalian RNAi in vivo.


Subject(s)
RNA Interference , RNA, Double-Stranded , RNA, Small Interfering , Ribonuclease III , Animals , Ribonuclease III/genetics , Ribonuclease III/metabolism , RNA, Double-Stranded/metabolism , RNA, Double-Stranded/genetics , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Female , Protein Isoforms/genetics , Protein Isoforms/metabolism
11.
Mol Cell ; 71(4): 629-636.e5, 2018 08 16.
Article in English | MEDLINE | ID: mdl-30118681

ABSTRACT

The kinases PERK and IRE1 alleviate endoplasmic reticulum (ER) stress by orchestrating the unfolded protein response (UPR). If stress mitigation fails, PERK promotes cell death by activating pro-apoptotic genes, including death receptor 5 (DR5). Conversely, IRE1-which harbors both kinase and endoribonuclease (RNase) modules-blocks apoptosis through regulated IRE1-dependent decay (RIDD) of DR5 mRNA. Under irresolvable ER stress, PERK activity persists, whereas IRE1 paradoxically attenuates, by mechanisms that remain obscure. Here, we report that PERK governs IRE1's attenuation through a phosphatase known as RPAP2 (RNA polymerase II-associated protein 2). RPAP2 reverses IRE1 phosphorylation, oligomerization, and RNase activation. This inhibits IRE1-mediated adaptive events, including activation of the cytoprotective transcription factor XBP1s, and ER-associated degradation of unfolded proteins. Furthermore, RIDD termination by RPAP2 unleashes DR5-mediated caspase activation and drives cell death. Thus, PERK attenuates IRE1 via RPAP2 to abort failed ER-stress adaptation and trigger apoptosis.


Subject(s)
Apoptosis/genetics , Carrier Proteins/genetics , Endoribonucleases/genetics , Protein Serine-Threonine Kinases/genetics , Unfolded Protein Response , eIF-2 Kinase/genetics , Carrier Proteins/metabolism , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/genetics , Endoribonucleases/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism , eIF-2 Kinase/metabolism
12.
Mol Cell ; 72(6): 1035-1049.e5, 2018 12 20.
Article in English | MEDLINE | ID: mdl-30503769

ABSTRACT

Membrane-less organelles (MLOs) are liquid-like subcellular compartments that form through phase separation of proteins and RNA. While their biophysical properties are increasingly understood, their regulation and the consequences of perturbed MLO states for cell physiology are less clear. To study the regulatory networks, we targeted 1,354 human genes and screened for morphological changes of nucleoli, Cajal bodies, splicing speckles, PML nuclear bodies (PML-NBs), cytoplasmic processing bodies, and stress granules. By multivariate analysis of MLO features we identified hundreds of genes that control MLO homeostasis. We discovered regulatory crosstalk between MLOs, and mapped hierarchical interactions between aberrant MLO states and cellular properties. We provide evidence that perturbation of pre-mRNA splicing results in stress granule formation and reveal that PML-NB abundance influences DNA replication rates and that PML-NBs are in turn controlled by HIP kinases. Together, our comprehensive dataset is an unprecedented resource for deciphering the regulation and biological functions of MLOs.


Subject(s)
Organelles/genetics , Stress, Physiological/genetics , Systems Biology/methods , Transcriptome , DNA Replication , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , HeLa Cells , Humans , Organelles/metabolism , Phase Transition , RNA Interference , RNA Precursors/genetics , RNA, Messenger/genetics , Signal Transduction/genetics , Single-Cell Analysis
13.
Mol Cell Proteomics ; 23(4): 100742, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38401707

ABSTRACT

Therapeutic RNAs are routinely modified during their synthesis to ensure proper drug uptake, stability, and efficacy. Phosphorothioate (PS) RNA, molecules in which one or more backbone phosphates are modified with a sulfur atom in place of standard nonbridging oxygen, is one of the most common modifications because of ease of synthesis and pharmacokinetic benefits. Quality assessment of RNA synthesis, including modification incorporation, is essential for drug selectivity and performance, and the synthetic nature of the PS linkage incorporation often reveals impurities. Here, we present a comprehensive analysis of PS RNA via tandem mass spectrometry (MS). We show that activated ion-negative electron transfer dissociation MS/MS is especially useful in diagnosing PS incorporation, producing diagnostic a- and z-type ions at PS linkage sites, beyond the standard d- and w-type ions. Analysis using resonant and beam-type collision-based activation reveals that, overall, more intense sequence ions and base-loss ions result when a PS modification is present. Furthermore, we report increased detection of b- and x-type product ions at sites of PS incorporation, in addition to the standard c- and y-type ions. This work reveals that the gas-phase chemical stability afforded by sulfur alters RNA dissociation and necessitates inclusion of additional product ions for MS/MS of PS RNA.


Subject(s)
RNA , Tandem Mass Spectrometry , Tandem Mass Spectrometry/methods , RNA/metabolism , Phosphorothioate Oligonucleotides/chemistry
14.
Proc Natl Acad Sci U S A ; 120(17): e2215253120, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37068229

ABSTRACT

Strategies to overcome irreversible cochlear hair cell (HC) damage and loss in mammals are of vital importance to hearing recovery in patients with permanent hearing loss. In mature mammalian cochlea, co-activation of Myc and Notch1 reprograms supporting cells (SC) and promotes HC regeneration. Understanding of the underlying mechanisms may aid the development of a clinically relevant approach to achieve HC regeneration in the nontransgenic mature cochlea. By single-cell RNAseq, we show that MYC/NICD "rejuvenates" the adult mouse cochlea by activating multiple pathways including Wnt and cyclase activator of cyclic AMP (cAMP), whose blockade suppresses HC-like cell regeneration despite Myc/Notch activation. We screened and identified a combination (the cocktail) of drug-like molecules composing of small molecules and small interfering RNAs to activate the pathways of Myc, Notch1, Wnt and cAMP. We show that the cocktail effectively replaces Myc and Notch1 transgenes and reprograms fully mature wild-type (WT) SCs for HC-like cells regeneration in vitro. Finally, we demonstrate the cocktail is capable of reprogramming adult cochlea for HC-like cells regeneration in WT mice with HC loss in vivo. Our study identifies a strategy by a clinically relevant approach to reprogram mature inner ear for HC-like cells regeneration, laying the foundation for hearing restoration by HC regeneration.


Subject(s)
Ear, Inner , Hair Cells, Auditory , Mice , Animals , Cell Proliferation/physiology , Hair Cells, Auditory/physiology , Ear, Inner/metabolism , Cochlea/physiology , Regeneration/physiology , Mammals
15.
Genes Dev ; 32(3-4): 271-282, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29483152

ABSTRACT

Cellular dsRNAs are edited by adenosine deaminases that act on RNA (ADARs). While editing can alter mRNA-coding potential, most editing occurs in noncoding sequences, the function of which is poorly understood. Using dsRNA immunoprecipitation (dsRIP) and RNA sequencing (RNA-seq), we identified 1523 regions of clustered A-to-I editing, termed editing-enriched regions (EERs), in four stages of Caenorhabditis elegans development, often with highest expression in embryos. Analyses of small RNA-seq data revealed 22- to 23-nucleotide (nt) siRNAs, reminiscent of viral siRNAs, that mapped to EERs and were abundant in adr-1;adr-2 mutant animals. Consistent with roles for these siRNAs in silencing, EER-associated genes (EAGs) were down-regulated in adr-1;adr-2 embryos, and this was dependent on associated EERs and the RNAi factor RDE-4. We observed that ADARs genetically interact with the 26G endogenous siRNA (endo-siRNA) pathway, which likely competes for RNAi components; deletion of factors required for this pathway (rrf-3 or ergo-1) in adr-1;adr-2 mutant strains caused a synthetic phenotype that was rescued by deleting antiviral RNAi factors. Poly(A)+ RNA-seq revealed EAG down-regulation and antiviral gene induction in adr-1;adr-2;rrf-3 embryos, and these expression changes were dependent on rde-1 and rde-4 Our data suggest that ADARs restrict antiviral silencing of cellular dsRNAs.


Subject(s)
Adenosine Deaminase/genetics , Caenorhabditis elegans Proteins/genetics , RNA Editing , RNA Interference , RNA, Double-Stranded/metabolism , Adenosine/metabolism , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Inosine/metabolism , Mutation , RNA, Small Interfering/metabolism , RNA-Dependent RNA Polymerase/genetics , Ribonuclease III/metabolism
16.
Genes Dev ; 32(3-4): 199-201, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29491134

ABSTRACT

Adenosine deaminases that act on RNA (ADARs) convert adenosines (A) to inosines (I) in stretches of dsRNA. The biological purpose of these editing events for the vast majority of ADAR substrates is largely unknown. In this issue of Genes & Development, Reich and colleagues (pp. 271-282) demonstrate that in Caenorhabditis elegans, A-to-I editing in double-stranded regions of protein-coding transcripts protects these RNAs from targeting by the RNAi pathway. Disruption of this safeguard through loss of ADAR activity coupled with enhanced RNAi results in developmental abnormalities and profound changes in gene expression that suggest aberrant induction of an antiviral response. Thus, editing of cellular dsRNA by ADAR helps prevent host RNA silencing and inadvertent antiviral activity.


Subject(s)
Antiviral Agents , Caenorhabditis elegans/genetics , Animals , RNA Editing , RNA Interference , RNA, Double-Stranded , Radar
17.
Trends Biochem Sci ; 46(5): 351-365, 2021 05.
Article in English | MEDLINE | ID: mdl-33309323

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is currently creating a global health emergency. This crisis is driving a worldwide effort to develop effective vaccines, prophylactics, and therapeutics. Nucleic acid (NA)-based treatments hold great potential to combat outbreaks of coronaviruses (CoVs) due to their rapid development, high target specificity, and the capacity to increase druggability. Here, we review key anti-CoV NA-based technologies, including antisense oligonucleotides (ASOs), siRNAs, RNA-targeting clustered regularly interspaced short palindromic repeats-CRISPR-associated protein (CRISPR-Cas), and mRNA vaccines, and discuss improved delivery methods and combination therapies with other antiviral drugs.


Subject(s)
COVID-19 Vaccines , CRISPR-Cas Systems , RNA, Messenger , RNA, Viral , SARS-CoV-2 , COVID-19/genetics , COVID-19/immunology , COVID-19/metabolism , COVID-19/therapy , COVID-19 Vaccines/genetics , COVID-19 Vaccines/immunology , COVID-19 Vaccines/therapeutic use , Humans , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/immunology , RNA, Viral/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/metabolism
18.
Plant J ; 117(1): 226-241, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37797206

ABSTRACT

Polyploids are common and have a wide geographical distribution and environmental adaptability. Allopolyploidy may lead to the activation of transposable elements (TE). However, the mechanism of epigenetic modification of TEs in the establishment and evolution of allopolyploids remains to be explored. We focused on the TEs of model allopolyploid Brassica napus (An An Cn Cn ), exploring the TE characteristics of the genome, epigenetic modifications of TEs during allopolyploidization, and regulation of gene expression by TE methylation. In B. napus, approximately 50% of the genome was composed of TEs. TEs increased with proximity to genes, especially DNA transposons. TE methylation levels were negatively correlated with gene expression, and changes in TE methylation levels were able to regulate the expression of neighboring genes related to responses to light intensity and stress, which promoted powerful adaptation of allopolyploids to new environments. TEs can be synergistically regulated by RNA-directed DNA methylation pathways and histone modifications. The epigenetic modification levels of TEs tended to be similar to those of the diploid parents during the genome evolution of B. napus. The TEs of the An subgenome were more likely to be modified, and the imbalance in TE number and epigenetic modification level in the An and Cn subgenomes may lead to the establishment of subgenome dominance. Our study analyzed the characteristics of TE location, DNA methylation, siRNA, and histone modification in B. napus and highlighted the importance of TE epigenetic modifications during the allopolyploidy process, providing support for revealing the mechanism of allopolyploid formation and evolution.


Subject(s)
Brassica napus , DNA Transposable Elements , DNA Transposable Elements/genetics , Brassica napus/genetics , Brassica napus/metabolism , Gene Expression Regulation, Plant/genetics , Genome, Plant/genetics , Epigenesis, Genetic/genetics , DNA Methylation/genetics , Genomics
19.
Plant J ; 117(4): 1206-1222, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38038953

ABSTRACT

MicroRNA (miRNA) target mimicry technologies, utilizing naturally occurring miRNA decoy molecules, represent a potent tool for analyzing miRNA function. In this study, we present a highly efficient small RNA (sRNA) target mimicry design based on G-U base-paired hairpin RNA (hpG:U), which allows for the simultaneous targeting of multiple sRNAs. The hpG:U constructs consistently generate high amounts of intact, polyadenylated stem-loop (SL) RNA outside the nuclei, in contrast to traditional hairpin RNA designs with canonical base pairing (hpWT), which were predominantly processed resulting in a loop. By incorporating a 460-bp G-U base-paired double-stranded stem and a 312-576 nt loop carrying multiple miRNA target mimicry sites (GUMIC), the hpG:U construct displayed effective repression of three Arabidopsis miRNAs, namely miR165/166, miR157, and miR160, both individually and in combination. Additionally, a GUMIC construct targeting a prominent cluster of siRNAs derived from cucumber mosaic virus (CMV) Y-satellite RNA (Y-Sat) effectively inhibited Y-Sat siRNA-directed silencing of the chlorophyll biosynthetic gene CHLI, thereby reducing the yellowing symptoms in infected Nicotiana plants. Therefore, the G-U base-paired hpRNA, characterized by differential processing compared to traditional hpRNA, acts as an efficient decoy for both miRNAs and siRNAs. This technology holds great potential for sRNA functional analysis and the management of sRNA-mediated diseases.


Subject(s)
Arabidopsis , MicroRNAs , Base Pairing/genetics , Plants, Genetically Modified/genetics , RNA, Small Interfering/genetics , MicroRNAs/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Double-Stranded , Arabidopsis/genetics
20.
J Cell Sci ; 136(23)2023 12 01.
Article in English | MEDLINE | ID: mdl-37997922

ABSTRACT

The correct inheritance of chromatin structure is key for maintaining genome function and cell identity and preventing cellular transformation. DEK, a conserved non-histone chromatin protein, has recognized tumor-promoting properties, its overexpression being associated with poor prognosis in various cancer types. At the cellular level, DEK displays pleiotropic functions, influencing differentiation, apoptosis and stemness, but a characteristic oncogenic mechanism has remained elusive. Here, we report the identification of DEK bodies, focal assemblies of DEK that regularly occur at specific, yet unidentified, sites of heterochromatin replication exclusively in late S-phase. In these bodies, DEK localizes in direct proximity to active replisomes in agreement with a function in the early maturation of heterochromatin. A high-throughput siRNA screen, supported by mutational and biochemical analyses, identifies SUMO as one regulator of DEK body formation, linking DEK to the complex SUMO protein network that controls chromatin states and cell fate. This work combines and refines our previous data on DEK as a factor essential for heterochromatin integrity and facilitating replication under stress, and delineates an avenue of further study for unraveling the contribution of DEK to cancer development.


Subject(s)
Heterochromatin , Neoplasms , Humans , Poly-ADP-Ribose Binding Proteins/genetics , Chromosomal Proteins, Non-Histone/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Chromatin
SELECTION OF CITATIONS
SEARCH DETAIL