Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 791
Filter
Add more filters

Publication year range
1.
Cell ; 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38942014

ABSTRACT

Neuroimmune interactions mediate intercellular communication and underlie critical brain functions. Microglia, CNS-resident macrophages, modulate the brain through direct physical interactions and the secretion of molecules. One such secreted factor, the complement protein C1q, contributes to complement-mediated synapse elimination in both developmental and disease models, yet brain C1q protein levels increase significantly throughout aging. Here, we report that C1q interacts with neuronal ribonucleoprotein (RNP) complexes in an age-dependent manner. Purified C1q protein undergoes RNA-dependent liquid-liquid phase separation (LLPS) in vitro, and the interaction of C1q with neuronal RNP complexes in vivo is dependent on RNA and endocytosis. Mice lacking C1q have age-specific alterations in neuronal protein synthesis in vivo and impaired fear memory extinction. Together, our findings reveal a biophysical property of C1q that underlies RNA- and age-dependent neuronal interactions and demonstrate a role of C1q in critical intracellular neuronal processes.

2.
Annu Rev Biochem ; 91: 197-219, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35303788

ABSTRACT

DEAD-box ATPases constitute a very large protein family present in all cells, often in great abundance. From bacteria to humans, they play critical roles in many aspects of RNA metabolism, and due to their widespread importance in RNA biology, they have been characterized in great detail at both the structural and biochemical levels. DEAD-box proteins function as RNA-dependent ATPases that can unwind short duplexes of RNA, remodel ribonucleoprotein (RNP) complexes, or act as clamps to promote RNP assembly. Yet, it often remains enigmatic how individual DEAD-box proteins mechanistically contribute to specific RNA-processing steps. Here, we review the role of DEAD-box ATPases in the regulation of gene expression and propose that one common function of these enzymes is in the regulation of liquid-liquid phase separation of RNP condensates.


Subject(s)
DEAD-box RNA Helicases , RNA , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , DEAD-box RNA Helicases/chemistry , Gene Expression , Humans , RNA/metabolism
3.
Cell ; 185(8): 1308-1324.e23, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35325593

ABSTRACT

Asymmetric localization of oskar ribonucleoprotein (RNP) granules to the oocyte posterior is crucial for abdominal patterning and germline formation in the Drosophila embryo. We show that oskar RNP granules in the oocyte are condensates with solid-like physical properties. Using purified oskar RNA and scaffold proteins Bruno and Hrp48, we confirm in vitro that oskar granules undergo a liquid-to-solid phase transition. Whereas the liquid phase allows RNA incorporation, the solid phase precludes incorporation of additional RNA while allowing RNA-dependent partitioning of client proteins. Genetic modification of scaffold granule proteins or tethering the intrinsically disordered region of human fused in sarcoma (FUS) to oskar mRNA allowed modulation of granule material properties in vivo. The resulting liquid-like properties impaired oskar localization and translation with severe consequences on embryonic development. Our study reflects how physiological phase transitions shape RNA-protein condensates to regulate the localization and expression of a maternal RNA that instructs germline formation.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Embryo, Nonmammalian/metabolism , Animals , Cytoplasmic Ribonucleoprotein Granules , Drosophila/embryology , Drosophila Proteins/genetics , Embryonic Development , Oocytes/metabolism , RNA/metabolism
4.
Cell ; 183(3): 730-738.e13, 2020 10 29.
Article in English | MEDLINE | ID: mdl-32979942

ABSTRACT

SARS-CoV-2 is an enveloped virus responsible for the COVID-19 pandemic. Despite recent advances in the structural elucidation of SARS-CoV-2 proteins, the detailed architecture of the intact virus remains to be unveiled. Here we report the molecular assembly of the authentic SARS-CoV-2 virus using cryoelectron tomography (cryo-ET) and subtomogram averaging (STA). Native structures of the S proteins in pre- and postfusion conformations were determined to average resolutions of 8.7-11 Å. Compositions of the N-linked glycans from the native spikes were analyzed by mass spectrometry, which revealed overall processing states of the native glycans highly similar to that of the recombinant glycoprotein glycans. The native conformation of the ribonucleoproteins (RNPs) and their higher-order assemblies were revealed. Overall, these characterizations revealed the architecture of the SARS-CoV-2 virus in exceptional detail and shed light on how the virus packs its ∼30-kb-long single-segmented RNA in the ∼80-nm-diameter lumen.


Subject(s)
Betacoronavirus/physiology , Betacoronavirus/ultrastructure , Virus Assembly , Animals , Chlorocebus aethiops , Cryoelectron Microscopy , Humans , Mass Spectrometry , Models, Molecular , Protein Conformation , SARS-CoV-2 , Vero Cells , Viral Proteins/chemistry , Viral Proteins/ultrastructure , Virus Cultivation
5.
Cell ; 180(3): 411-426.e16, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31928844

ABSTRACT

Stress granules are condensates of non-translating mRNAs and proteins involved in the stress response and neurodegenerative diseases. Stress granules form in part through intermolecular RNA-RNA interactions, and to better understand how RNA-based condensation occurs, we demonstrate that RNA is effectively recruited to the surfaces of RNA or RNP condensates in vitro. We demonstrate that, through ATP-dependent RNA binding, the DEAD-box protein eIF4A reduces RNA condensation in vitro and limits stress granule formation in cells. This defines a function for eIF4A to limit intermolecular RNA-RNA interactions in cells. These results establish an important role for eIF4A, and potentially other DEAD-box proteins, as ATP-dependent RNA chaperones that limit the condensation of RNA, analogous to the function of proteins like HSP70 in combatting protein aggregates.


Subject(s)
DEAD-box RNA Helicases/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4F/metabolism , RNA Helicases/metabolism , RNA, Fungal/metabolism , Ribonucleoproteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphate/metabolism , HeLa Cells , Humans , Microscopy, Confocal , Protein Binding , RNA, Fungal/isolation & purification , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Time-Lapse Imaging
6.
Cell ; 177(2): 446-462.e16, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30951671

ABSTRACT

Poor reproducibility within and across studies arising from lack of knowledge regarding the performance of extracellular RNA (exRNA) isolation methods has hindered progress in the exRNA field. A systematic comparison of 10 exRNA isolation methods across 5 biofluids revealed marked differences in the complexity and reproducibility of the resulting small RNA-seq profiles. The relative efficiency with which each method accessed different exRNA carrier subclasses was determined by estimating the proportions of extracellular vesicle (EV)-, ribonucleoprotein (RNP)-, and high-density lipoprotein (HDL)-specific miRNA signatures in each profile. An interactive web-based application (miRDaR) was developed to help investigators select the optimal exRNA isolation method for their studies. miRDar provides comparative statistics for all expressed miRNAs or a selected subset of miRNAs in the desired biofluid for each exRNA isolation method and returns a ranked list of exRNA isolation methods prioritized by complexity, expression level, and reproducibility. These results will improve reproducibility and stimulate further progress in exRNA biomarker development.


Subject(s)
Cell-Free Nucleic Acids/isolation & purification , Circulating MicroRNA/isolation & purification , RNA/isolation & purification , Adult , Body Fluids/chemistry , Cell Line , Extracellular Vesicles/metabolism , Female , Healthy Volunteers , Humans , Male , MicroRNAs/isolation & purification , MicroRNAs/metabolism , RNA/metabolism , Reproducibility of Results , Sequence Analysis, RNA/methods
7.
Cell ; 179(6): 1357-1369.e16, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31761533

ABSTRACT

Ribosome assembly is an efficient but complex and heterogeneous process during which ribosomal proteins assemble on the nascent rRNA during transcription. Understanding how the interplay between nascent RNA folding and protein binding determines the fate of transcripts remains a major challenge. Here, using single-molecule fluorescence microscopy, we follow assembly of the entire 3' domain of the bacterial small ribosomal subunit in real time. We find that co-transcriptional rRNA folding is complicated by the formation of long-range RNA interactions and that r-proteins self-chaperone the rRNA folding process prior to stable incorporation into a ribonucleoprotein (RNP) complex. Assembly is initiated by transient rather than stable protein binding, and the protein-RNA binding dynamics gradually decrease during assembly. This work questions the paradigm of strictly sequential and cooperative ribosome assembly and suggests that transient binding of RNA binding proteins to cellular RNAs could provide a general mechanism to shape nascent RNA folding during RNP assembly.


Subject(s)
RNA Folding , RNA, Ribosomal/metabolism , RNA-Binding Proteins/metabolism , Models, Biological , Nucleic Acid Conformation , Protein Binding , RNA Stability , RNA, Ribosomal/chemistry , Transcription, Genetic
8.
Cell ; 179(6): 1370-1381.e12, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31761536

ABSTRACT

The synthesis of new ribosomes begins during transcription of the rRNA and is widely assumed to follow an orderly 5' to 3' gradient. To visualize co-transcriptional assembly of ribosomal protein-RNA complexes in real time, we developed a single-molecule platform that simultaneously monitors transcription and protein association with the elongating transcript. Unexpectedly, the early assembly protein uS4 binds newly made pre-16S rRNA only transiently, likely due to non-native folding of the rRNA during transcription. Stable uS4 binding became more probable only in the presence of additional ribosomal proteins that bind upstream and downstream of protein uS4 by allowing productive assembly intermediates to form earlier. We propose that dynamic sampling of elongating RNA by multiple proteins overcomes heterogeneous RNA folding, preventing assembly bottlenecks and initiating assembly within the transcription time window. This may be a common feature of transcription-coupled RNP assembly.


Subject(s)
Ribonucleoproteins/metabolism , Transcription, Genetic , Fluorescence , Models, Biological , Protein Binding , Protein Stability , RNA Precursors/biosynthesis , RNA Precursors/chemistry , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Transcription Elongation, Genetic
9.
Cell ; 172(3): 590-604.e13, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29373831

ABSTRACT

Stress granules (SGs) are transient ribonucleoprotein (RNP) aggregates that form during cellular stress and are increasingly implicated in human neurodegeneration. To study the proteome and compositional diversity of SGs in different cell types and in the context of neurodegeneration-linked mutations, we used ascorbate peroxidase (APEX) proximity labeling, mass spectrometry, and immunofluorescence to identify ∼150 previously unknown human SG components. A highly integrated, pre-existing SG protein interaction network in unstressed cells facilitates rapid coalescence into larger SGs. Approximately 20% of SG diversity is stress or cell-type dependent, with neuronal SGs displaying a particularly complex repertoire of proteins enriched in chaperones and autophagy factors. Strengthening the link between SGs and neurodegeneration, we demonstrate aberrant dynamics, composition, and subcellular distribution of SGs in cells from amyotrophic lateral sclerosis (ALS) patients. Using three Drosophila ALS/FTD models, we identify SG-associated modifiers of neurotoxicity in vivo. Altogether, our results highlight SG proteins as central to understanding and ultimately targeting neurodegeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Cytoplasmic Granules/metabolism , Protein Interaction Maps , Ribonucleoproteins/metabolism , Stress, Physiological , Animals , Drosophila melanogaster , HEK293 Cells , HeLa Cells , Humans , Neurons/metabolism , Protein Transport
10.
Cell ; 173(3): 720-734.e15, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29677515

ABSTRACT

Reversible phase separation underpins the role of FUS in ribonucleoprotein granules and other membrane-free organelles and is, in part, driven by the intrinsically disordered low-complexity (LC) domain of FUS. Here, we report that cooperative cation-π interactions between tyrosines in the LC domain and arginines in structured C-terminal domains also contribute to phase separation. These interactions are modulated by post-translational arginine methylation, wherein arginine hypomethylation strongly promotes phase separation and gelation. Indeed, significant hypomethylation, which occurs in FUS-associated frontotemporal lobar degeneration (FTLD), induces FUS condensation into stable intermolecular ß-sheet-rich hydrogels that disrupt RNP granule function and impair new protein synthesis in neuron terminals. We show that transportin acts as a physiological molecular chaperone of FUS in neuron terminals, reducing phase separation and gelation of methylated and hypomethylated FUS and rescuing protein synthesis. These results demonstrate how FUS condensation is physiologically regulated and how perturbations in these mechanisms can lead to disease.


Subject(s)
Arginine/chemistry , Molecular Chaperones/chemistry , RNA-Binding Protein FUS/chemistry , Amyotrophic Lateral Sclerosis/metabolism , Animals , Cations , DNA Methylation , Frontotemporal Dementia/metabolism , Frontotemporal Lobar Degeneration/metabolism , Humans , Microscopy, Atomic Force , Microscopy, Fluorescence , Protein Binding , Protein Domains , Protein Processing, Post-Translational , Protein Structure, Secondary , RNA-Binding Protein FUS/metabolism , Tyrosine/chemistry , Xenopus laevis
11.
Annu Rev Biochem ; 86: 439-460, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28141967

ABSTRACT

Telomerase is the essential reverse transcriptase required for linear chromosome maintenance in most eukaryotes. Telomerase supplements the tandem array of simple-sequence repeats at chromosome ends to compensate for the DNA erosion inherent in genome replication. The template for telomerase reverse transcriptase is within the RNA subunit of the ribonucleoprotein complex, which in cells contains additional telomerase holoenzyme proteins that assemble the active ribonucleoprotein and promote its function at telomeres. Telomerase is distinct among polymerases in its reiterative reuse of an internal template. The template is precisely defined, processively copied, and regenerated by release of single-stranded product DNA. New specificities of nucleic acid handling that underlie the catalytic cycle of repeat synthesis derive from both active site specialization and new motif elaborations in protein and RNA subunits. Studies of telomerase provide unique insights into cellular requirements for genome stability, tissue renewal, and tumorigenesis as well as new perspectives on dynamic ribonucleoprotein machines.


Subject(s)
DNA Replication , DNA, Single-Stranded/metabolism , RNA/metabolism , Ribonucleoproteins/metabolism , Telomerase/metabolism , Telomere/enzymology , Animals , Catalytic Domain , DNA, Single-Stranded/genetics , Gene Expression Regulation , Humans , Microsatellite Repeats , Nucleic Acid Conformation , Oxytricha/genetics , Oxytricha/metabolism , RNA/genetics , Ribonucleoproteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Telomerase/genetics , Telomere/chemistry , Tetrahymena thermophila/genetics , Tetrahymena thermophila/metabolism
12.
Cell ; 168(1-2): 159-171.e14, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-28041848

ABSTRACT

Phase transitions driven by intrinsically disordered protein regions (IDRs) have emerged as a ubiquitous mechanism for assembling liquid-like RNA/protein (RNP) bodies and other membrane-less organelles. However, a lack of tools to control intracellular phase transitions limits our ability to understand their role in cell physiology and disease. Here, we introduce an optogenetic platform that uses light to activate IDR-mediated phase transitions in living cells. We use this "optoDroplet" system to study condensed phases driven by the IDRs of various RNP body proteins, including FUS, DDX4, and HNRNPA1. Above a concentration threshold, these constructs undergo light-activated phase separation, forming spatiotemporally definable liquid optoDroplets. FUS optoDroplet assembly is fully reversible even after multiple activation cycles. However, cells driven deep within the phase boundary form solid-like gels that undergo aging into irreversible aggregates. This system can thus elucidate not only physiological phase transitions but also their link to pathological aggregates.


Subject(s)
Molecular Imaging/methods , Phase Transition , Proteins/chemistry , Animals , Arabidopsis Proteins , Cryptochromes , Intrinsically Disordered Proteins , Kinetics , Light , Mice , Models, Chemical , NIH 3T3 Cells , Optogenetics , Protein Interaction Maps , Proteins/metabolism
13.
Mol Cell ; 83(17): 3095-3107.e9, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37683610

ABSTRACT

The nucleolus is the largest biomolecular condensate and facilitates transcription, processing, and assembly of ribosomal RNA (rRNA). Although nucleolar function is thought to require multiphase liquid-like properties, nucleolar fluidity and its connection to the highly coordinated transport and biogenesis of ribosomal subunits are poorly understood. Here, we use quantitative imaging, mathematical modeling, and pulse-chase nucleotide labeling to examine nucleolar material properties and rRNA dynamics. The mobility of rRNA is several orders of magnitude slower than that of nucleolar proteins, with rRNA steadily moving away from the transcriptional sites in a slow (∼1 Å/s), radially directed fashion. This constrained but directional mobility, together with polymer physics-based calculations, suggests that nascent rRNA forms an entangled gel, whose constant production drives outward flow. We propose a model in which progressive maturation of nascent rRNA reduces its initial entanglement, fluidizing the nucleolar periphery to facilitate the release of assembled pre-ribosomal particles.


Subject(s)
RNA, Ribosomal , RNA , RNA/genetics , RNA, Ribosomal/genetics , Biomolecular Condensates , Cell Nucleolus/genetics , Nuclear Proteins/genetics
14.
Annu Rev Biochem ; 84: 381-404, 2015.
Article in English | MEDLINE | ID: mdl-25747400

ABSTRACT

The RNA polymerase II transcription cycle is often divided into three major stages: initiation, elongation, and termination. Research over the last decade has blurred these divisions and emphasized the tightly regulated transitions that occur as RNA polymerase II synthesizes a transcript from start to finish. Transcription termination, the process that marks the end of transcription elongation, is regulated by proteins that interact with the polymerase, nascent transcript, and/or chromatin template. The failure to terminate transcription can cause accumulation of aberrant transcripts and interfere with transcription at downstream genes. Here, we review the mechanism, regulation, and physiological impact of a termination pathway that targets small noncoding transcripts produced by RNA polymerase II. We emphasize the Nrd1-Nab3-Sen1 pathway in yeast, in which the process has been extensively studied. The importance of understanding small RNA termination pathways is underscored by the need to control noncoding transcription in eukaryotic genomes.


Subject(s)
RNA Polymerase II/metabolism , RNA, Small Untranslated/metabolism , Transcription Termination, Genetic , Animals , Cell Physiological Phenomena , Humans , RNA Processing, Post-Transcriptional , Saccharomyces cerevisiae/genetics
15.
Genes Dev ; 37(9-10): 354-376, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37137715

ABSTRACT

RNA granules are mesoscale assemblies that form in the absence of limiting membranes. RNA granules contain factors for RNA biogenesis and turnover and are often assumed to represent specialized compartments for RNA biochemistry. Recent evidence suggests that RNA granules assemble by phase separation of subsoluble ribonucleoprotein (RNP) complexes that partially demix from the cytoplasm or nucleoplasm. We explore the possibility that some RNA granules are nonessential condensation by-products that arise when RNP complexes exceed their solubility limit as a consequence of cellular activity, stress, or aging. We describe the use of evolutionary and mutational analyses and single-molecule techniques to distinguish functional RNA granules from "incidental condensates."


Subject(s)
Cytoplasmic Granules , Ribonucleoproteins , Ribonucleoproteins/genetics , Cytoplasmic Ribonucleoprotein Granules , RNA/chemistry
16.
Genes Dev ; 37(11-12): 505-517, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37399331

ABSTRACT

Messenger RNAs (mRNAs) are at the center of the central dogma of molecular biology. In eukaryotic cells, these long ribonucleic acid polymers do not exist as naked transcripts; rather, they associate with mRNA-binding proteins to form messenger ribonucleoprotein (mRNP) complexes. Recently, global proteomic and transcriptomic studies have provided comprehensive inventories of mRNP components. However, knowledge of the molecular features of distinct mRNP populations has remained elusive. We purified endogenous nuclear mRNPs from Saccharomyces cerevisiae by harnessing the mRNP biogenesis factors THO and Sub2 in biochemical procedures optimized to preserve the integrity of these transient ribonucleoprotein assemblies. We found that these mRNPs are compact particles that contain multiple copies of Yra1, an essential protein with RNA-annealing properties. To investigate their molecular and architectural organization, we used a combination of proteomics, RNA sequencing, cryo-electron microscopy, cross-linking mass spectrometry, structural models, and biochemical assays. Our findings indicate that yeast nuclear mRNPs are packaged around an intricate network of interconnected proteins capable of promoting RNA-RNA interactions via their positively charged intrinsically disordered regions. The evolutionary conservation of the major mRNA-packaging factor (yeast Yra1 and Aly/REF in metazoans) points toward a general paradigm governing nuclear mRNP packaging.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Cryoelectron Microscopy , Proteomics , Saccharomyces cerevisiae Proteins/metabolism , Ribonucleoproteins/genetics , RNA, Messenger/metabolism
17.
Mol Cell ; 81(14): 2901-2913.e5, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34157309

ABSTRACT

Polynucleotide phosphorylase (PNPase) is an ancient exoribonuclease conserved in the course of evolution and is found in species as diverse as bacteria and humans. Paradoxically, Escherichia coli PNPase can act not only as an RNA degrading enzyme but also by an unknown mechanism as a chaperone for small regulatory RNAs (sRNAs), with pleiotropic consequences for gene regulation. We present structures of the ternary assembly formed by PNPase, the RNA chaperone Hfq, and sRNA and show that this complex boosts sRNA stability in vitro. Comparison of structures for PNPase in RNA carrier and degradation modes reveals how the RNA is rerouted away from the active site through interactions with Hfq and the KH and S1 domains. Together, these data explain how PNPase is repurposed to protect sRNAs from cellular ribonucleases such as RNase E and could aid RNA presentation to facilitate regulatory actions on target genes.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/genetics , Host Factor 1 Protein/genetics , Polyribonucleotide Nucleotidyltransferase/genetics , RNA, Bacterial/genetics , Catalytic Domain/genetics , Endoribonucleases/genetics , Exoribonucleases/genetics , Gene Expression Regulation, Bacterial/genetics , Molecular Chaperones/genetics , RNA Stability/genetics , RNA, Small Untranslated/genetics
18.
Mol Cell ; 81(13): 2851-2867.e7, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34118193

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19). SARS-CoV-2 relies on cellular RNA-binding proteins (RBPs) to replicate and spread, although which RBPs control its life cycle remains largely unknown. Here, we employ a multi-omic approach to identify systematically and comprehensively the cellular and viral RBPs that are involved in SARS-CoV-2 infection. We reveal that SARS-CoV-2 infection profoundly remodels the cellular RNA-bound proteome, which includes wide-ranging effects on RNA metabolic pathways, non-canonical RBPs, and antiviral factors. Moreover, we apply a new method to identify the proteins that directly interact with viral RNA, uncovering dozens of cellular RBPs and six viral proteins. Among them are several components of the tRNA ligase complex, which we show regulate SARS-CoV-2 infection. Furthermore, we discover that available drugs targeting host RBPs that interact with SARS-CoV-2 RNA inhibit infection. Collectively, our results uncover a new universe of host-virus interactions with potential for new antiviral therapies against COVID-19.


Subject(s)
COVID-19/metabolism , Proteome/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , SARS-CoV-2/physiology , Viral Proteins/metabolism , Virus Replication/physiology , A549 Cells , COVID-19/genetics , Humans , Proteome/genetics , RNA, Viral/genetics , RNA-Binding Proteins/genetics , Viral Proteins/genetics
19.
Trends Biochem Sci ; 49(7): 611-621, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38677920

ABSTRACT

YTHDF proteins are main cytoplasmic 'reader' proteins of RNA N6-methyladenosine (m6A) methylation in mammals. They are largely responsible for m6A-mediated regulation in the cell cytosol by controlling both mRNA translation and degradation. Recent functional and mechanistic investigations of the YTHDF proteins revealed that these proteins have different functions to enable versatile regulation of the epitranscriptome. Their divergent functions largely originate from their different amino acid sequences in the low-complexity N termini. Consequently, they have different phase separation propensities and possess distinct post-translational modifications (PTMs). Different PTMs, subcellular localizations, and competition among partner proteins have emerged as three major mechanisms that control the functions of these YTHDF proteins. We also summarize recent progress on critical roles of these YTHDF proteins in anticancer immunity and the potential for targeting these proteins for developing new anticancer therapies.


Subject(s)
Adenosine , RNA-Binding Proteins , Humans , Animals , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Adenosine/metabolism , Adenosine/analogs & derivatives , Protein Processing, Post-Translational , RNA/metabolism , Methylation , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology
20.
Trends Biochem Sci ; 49(3): 199-207, 2024 03.
Article in English | MEDLINE | ID: mdl-38071089

ABSTRACT

Gene expression is a complex process requiring many control mechanisms to achieve a desired phenotype. DNA accessibility within chromatin is well established as an important determinant of gene expression. By contrast, while mRNA also associates with a complement of proteins, the exact nature of messenger ribonucleoprotein (mRNP) packaging and its functional relevance is not as clear. Recent reports indicate that exon junction complex (EJC)-mediated mRNP packaging renders exon junction-proximal regions inaccessible for m6A methylation, and that EJCs reside within the inaccessible interior of globular transcription and export (TREX) complex-associated nuclear mRNPs. We propose that 'mRNA accessibility' within mRNPs is an important determinant of gene expression that may modulate the specificity of a broad array of regulatory processes including but not limited to m6A methylation.


Subject(s)
Cell Nucleus , Ribonucleoproteins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Cell Nucleus/metabolism , Gene Expression
SELECTION OF CITATIONS
SEARCH DETAIL