Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Cell ; 186(22): 4834-4850.e23, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37794589

ABSTRACT

Regulation of viral RNA biogenesis is fundamental to productive SARS-CoV-2 infection. To characterize host RNA-binding proteins (RBPs) involved in this process, we biochemically identified proteins bound to genomic and subgenomic SARS-CoV-2 RNAs. We find that the host protein SND1 binds the 5' end of negative-sense viral RNA and is required for SARS-CoV-2 RNA synthesis. SND1-depleted cells form smaller replication organelles and display diminished virus growth kinetics. We discover that NSP9, a viral RBP and direct SND1 interaction partner, is covalently linked to the 5' ends of positive- and negative-sense RNAs produced during infection. These linkages occur at replication-transcription initiation sites, consistent with NSP9 priming viral RNA synthesis. Mechanistically, SND1 remodels NSP9 occupancy and alters the covalent linkage of NSP9 to initiating nucleotides in viral RNA. Our findings implicate NSP9 in the initiation of SARS-CoV-2 RNA synthesis and unravel an unsuspected role of a cellular protein in orchestrating viral RNA production.


Subject(s)
COVID-19 , RNA, Viral , Humans , COVID-19/metabolism , Endonucleases/metabolism , RNA, Viral/metabolism , SARS-CoV-2/genetics , Virus Replication
2.
Cell ; 173(1): 90-103.e19, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29551269

ABSTRACT

Blood cell formation is classically thought to occur through a hierarchical differentiation process, although recent studies have shown that lineage commitment may occur earlier in hematopoietic stem and progenitor cells (HSPCs). The relevance to human blood diseases and the underlying regulation of these refined models remain poorly understood. By studying a genetic blood disorder, Diamond-Blackfan anemia (DBA), where the majority of mutations affect ribosomal proteins and the erythroid lineage is selectively perturbed, we are able to gain mechanistic insight into how lineage commitment is programmed normally and disrupted in disease. We show that in DBA, the pool of available ribosomes is limited, while ribosome composition remains constant. Surprisingly, this global reduction in ribosome levels more profoundly alters translation of a select subset of transcripts. We show how the reduced translation of select transcripts in HSPCs can impair erythroid lineage commitment, illuminating a regulatory role for ribosome levels in cellular differentiation.


Subject(s)
Anemia, Diamond-Blackfan/pathology , Ribosomes/metabolism , 5' Untranslated Regions , Anemia, Diamond-Blackfan/genetics , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bone Marrow Cells/metabolism , Cells, Cultured , Female , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Male , Mutation, Missense , RNA Interference , RNA, Small Interfering/metabolism , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Genes Dev ; 33(23-24): 1673-1687, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31699777

ABSTRACT

Knockout of the ubiquitously expressed miRNA-17∼92 cluster in mice produces a lethal developmental lung defect, skeletal abnormalities, and blocked B lymphopoiesis. A shared target of miR-17∼92 miRNAs is the pro-apoptotic protein BIM, central to life-death decisions in mammalian cells. To clarify the contribution of miR-17∼92:Bim interactions to the complex miR-17∼92 knockout phenotype, we used a system of conditional mutagenesis of the nine Bim 3' UTR miR-17∼92 seed matches. Blocking miR-17∼92:Bim interactions early in development phenocopied the lethal lung phenotype of miR-17∼92 ablation and generated a skeletal kinky tail. In the hematopoietic system, instead of causing the predicted B cell developmental block, it produced a selective inability of B cells to resist cellular stress; and prevented B and T cell hyperplasia caused by Bim haploinsufficiency. Thus, the interaction of miR-17∼92 with a single target is essential for life, and BIM regulation by miRNAs serves as a rheostat controlling cell survival in specific physiological contexts.


Subject(s)
B-Lymphocytes/cytology , Bcl-2-Like Protein 11/metabolism , Cell Survival/genetics , Gene Expression Regulation, Developmental/genetics , Hematopoiesis/genetics , MicroRNAs/metabolism , 3' Untranslated Regions/genetics , Animals , B-Lymphocytes/pathology , Bcl-2-Like Protein 11/genetics , Gene Knockout Techniques , Lung/embryology , Mice , MicroRNAs/genetics , Mutation , Stress, Physiological
4.
Nucleic Acids Res ; 52(5): e26, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38281241

ABSTRACT

RNA-protein interactions determine the cellular fate of RNA and are central to regulating gene expression outcomes in health and disease. To date, no method exists that is able to identify proteins that interact with specific regions within endogenous RNAs in live cells. Here, we develop SHIFTR (Selective RNase H-mediated interactome framing for target RNA regions), an efficient and scalable approach to identify proteins bound to selected regions within endogenous RNAs using mass spectrometry. Compared to state-of-the-art techniques, SHIFTR is superior in accuracy, captures minimal background interactions and requires orders of magnitude lower input material. We establish SHIFTR workflows for targeting RNA classes of different length and abundance, including short and long non-coding RNAs, as well as mRNAs and demonstrate that SHIFTR is compatible with sequentially mapping interactomes for multiple target RNAs in a single experiment. Using SHIFTR, we comprehensively identify interactions of cis-regulatory elements located at the 5' and 3'-terminal regions of authentic SARS-CoV-2 RNAs in infected cells and accurately recover known and novel interactions linked to the function of these viral RNA elements. SHIFTR enables the systematic mapping of region-resolved RNA interactomes for any RNA in any cell type and has the potential to revolutionize our understanding of transcriptomes and their regulation.


Subject(s)
Proteomics , RNA-Binding Proteins , RNA , Software , RNA, Long Noncoding/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , Transcriptome , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , RNA/chemistry , RNA/metabolism , Proteomics/methods
5.
Nucleic Acids Res ; 52(6): 3050-3068, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38224452

ABSTRACT

RNA-binding proteins emerge as effectors of the DNA damage response (DDR). The multifunctional non-POU domain-containing octamer-binding protein NONO/p54nrb marks nuclear paraspeckles in unperturbed cells, but also undergoes re-localization to the nucleolus upon induction of DNA double-strand breaks (DSBs). However, NONO nucleolar re-localization is poorly understood. Here we show that the topoisomerase II inhibitor etoposide stimulates the production of RNA polymerase II-dependent, DNA damage-inducible antisense intergenic non-coding RNA (asincRNA) in human cancer cells. Such transcripts originate from distinct nucleolar intergenic spacer regions and form DNA-RNA hybrids to tether NONO to the nucleolus in an RNA recognition motif 1 domain-dependent manner. NONO occupancy at protein-coding gene promoters is reduced by etoposide, which attenuates pre-mRNA synthesis, enhances NONO binding to pre-mRNA transcripts and is accompanied by nucleolar detention of a subset of such transcripts. The depletion or mutation of NONO interferes with detention and prolongs DSB signalling. Together, we describe a nucleolar DDR pathway that shields NONO and aberrant transcripts from DSBs to promote DNA repair.


Subject(s)
DNA Breaks, Double-Stranded , DNA-Binding Proteins , Humans , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Etoposide/pharmacology , RNA Precursors/metabolism , Transcription Factors/metabolism , DNA , RNA-Binding Proteins/metabolism
6.
Cell ; 141(1): 129-41, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20371350

ABSTRACT

RNA transcripts are subject to posttranscriptional gene regulation involving hundreds of RNA-binding proteins (RBPs) and microRNA-containing ribonucleoprotein complexes (miRNPs) expressed in a cell-type dependent fashion. We developed a cell-based crosslinking approach to determine at high resolution and transcriptome-wide the binding sites of cellular RBPs and miRNPs. The crosslinked sites are revealed by thymidine to cytidine transitions in the cDNAs prepared from immunopurified RNPs of 4-thiouridine-treated cells. We determined the binding sites and regulatory consequences for several intensely studied RBPs and miRNPs, including PUM2, QKI, IGF2BP1-3, AGO/EIF2C1-4 and TNRC6A-C. Our study revealed that these factors bind thousands of sites containing defined sequence motifs and have distinct preferences for exonic versus intronic or coding versus untranslated transcript regions. The precise mapping of binding sites across the transcriptome will be critical to the interpretation of the rapidly emerging data on genetic variation between individuals and how these variations contribute to complex genetic diseases.


Subject(s)
Genetic Techniques , MicroRNAs/metabolism , RNA, Untranslated/genetics , RNA-Binding Proteins/metabolism , Regulatory Sequences, Ribonucleic Acid , Base Sequence , Cross-Linking Reagents/metabolism , Humans , Molecular Sequence Data , Nucleosides/metabolism , Point Mutation , Sequence Alignment
7.
Blood ; 139(16): 2534-2546, 2022 04 21.
Article in English | MEDLINE | ID: mdl-35030251

ABSTRACT

Master regulators, such as the hematopoietic transcription factor (TF) GATA1, play an essential role in orchestrating lineage commitment and differentiation. However, the precise mechanisms by which such TFs regulate transcription through interactions with specific cis-regulatory elements remain incompletely understood. Here, we describe a form of congenital hemolytic anemia caused by missense mutations in an intrinsically disordered region of GATA1, with a poorly understood role in transcriptional regulation. Through integrative functional approaches, we demonstrate that these mutations perturb GATA1 transcriptional activity by partially impairing nuclear localization and selectively altering precise chromatin occupancy by GATA1. These alterations in chromatin occupancy and concordant chromatin accessibility changes alter faithful gene expression, with failure to both effectively silence and activate select genes necessary for effective terminal red cell production. We demonstrate how disease-causing mutations can reveal regulatory mechanisms that enable the faithful genomic targeting of master TFs during cellular differentiation.


Subject(s)
Anemia , GATA1 Transcription Factor , Cell Differentiation/genetics , Chromatin/genetics , Chromatin Immunoprecipitation , Erythropoiesis/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Humans
9.
Nature ; 561(7721): 132-136, 2018 09.
Article in English | MEDLINE | ID: mdl-30150775

ABSTRACT

The human genome contains thousands of long non-coding RNAs1, but specific biological functions and biochemical mechanisms have been discovered for only about a dozen2-7. A specific long non-coding RNA-non-coding RNA activated by DNA damage (NORAD)-has recently been shown to be required for maintaining genomic stability8, but its molecular mechanism is unknown. Here we combine RNA antisense purification and quantitative mass spectrometry to identify proteins that directly interact with NORAD in living cells. We show that NORAD interacts with proteins involved in DNA replication and repair in steady-state cells and localizes to the nucleus upon stimulation with replication stress or DNA damage. In particular, NORAD interacts with RBMX, a component of the DNA-damage response, and contains the strongest RBMX-binding site in the transcriptome. We demonstrate that NORAD controls the ability of RBMX to assemble a ribonucleoprotein complex-which we term NORAD-activated ribonucleoprotein complex 1 (NARC1)-that contains the known suppressors of genomic instability topoisomerase I (TOP1), ALYREF and the PRPF19-CDC5L complex. Cells depleted for NORAD or RBMX display an increased frequency of chromosome segregation defects, reduced replication-fork velocity and altered cell-cycle progression-which represent phenotypes that are mechanistically linked to TOP1 and PRPF19-CDC5L function. Expression of NORAD in trans can rescue defects caused by NORAD depletion, but rescue is significantly impaired when the RBMX-binding site in NORAD is deleted. Our results demonstrate that the interaction between NORAD and RBMX is important for NORAD function, and that NORAD is required for the assembly of the previously unknown topoisomerase complex NARC1, which contributes to maintaining genomic stability. In addition, we uncover a previously unknown function for long non-coding RNAs in modulating the ability of an RNA-binding protein to assemble a higher-order ribonucleoprotein complex.


Subject(s)
DNA Topoisomerases, Type I/metabolism , Genomic Instability , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , Binding Sites , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Cell Survival , Chromosome Segregation , DNA Damage , DNA Repair , DNA Repair Enzymes/metabolism , DNA Replication , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Mass Spectrometry , Nuclear Proteins/metabolism , Protein Binding , RNA Splicing Factors/metabolism , RNA, Long Noncoding/genetics , Ribonucleoproteins/metabolism , Transcription Factors/metabolism
10.
Mol Cell ; 54(4): 573-85, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24726324

ABSTRACT

RNA helicases are important regulators of gene expression that act by remodeling RNA secondary structures and RNA-protein interactions. Here, we demonstrate that MOV10 has an ATP-dependent 5' to 3' in vitro RNA unwinding activity and determine the RNA-binding sites of MOV10 and its helicase mutants using PAR-CLIP. We find that MOV10 predominantly binds to 3' UTRs upstream of regions predicted to form local secondary structures and provide evidence that MOV10 helicase mutants are impaired in their ability to translocate 5' to 3' on their mRNA targets. MOV10 interacts with UPF1, the key component of the nonsense-mediated mRNA decay pathway. PAR-CLIP of UPF1 reveals that MOV10 and UPF1 bind to RNA in close proximity. Knockdown of MOV10 resulted in increased mRNA half-lives of MOV10-bound as well as UPF1-regulated transcripts, suggesting that MOV10 functions in UPF1-mediated mRNA degradation as an RNA clearance factor to resolve structures and displace proteins from 3' UTRs.


Subject(s)
3' Untranslated Regions , RNA Helicases/metabolism , RNA Stability , RNA, Messenger/metabolism , Trans-Activators/genetics , Amino Acid Motifs , Binding Sites , Gene Expression Regulation , Gene Knockdown Techniques , HEK293 Cells , Humans , Mutation , Nonsense Mediated mRNA Decay , Protein Transport , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism
11.
Biospektrum (Heidelb) ; 27(4): 376-379, 2021.
Article in German | MEDLINE | ID: mdl-34219983

ABSTRACT

Using RNA antisense purification and mass spectrometry, we identified more than 100 human proteins that directly and specifically bind SARS-CoV-2 RNA in infected cells. To gain insights into the functions of selected RNA interactors, we applied genetic perturbation and pharmacological inhibition experiments, and mapped the contact sites on the viral RNA. This led to the identification of host dependency factors and defense strategies, which can guide the design of novel therapeutics against SARS-CoV-2.

12.
Mol Cell ; 46(5): 674-90, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22681889

ABSTRACT

Protein-RNA interactions are fundamental to core biological processes, such as mRNA splicing, localization, degradation, and translation. We developed a photoreactive nucleotide-enhanced UV crosslinking and oligo(dT) purification approach to identify the mRNA-bound proteome using quantitative proteomics and to display the protein occupancy on mRNA transcripts by next-generation sequencing. Application to a human embryonic kidney cell line identified close to 800 proteins. To our knowledge, nearly one-third were not previously annotated as RNA binding, and about 15% were not predictable by computational methods to interact with RNA. Protein occupancy profiling provides a transcriptome-wide catalog of potential cis-regulatory regions on mammalian mRNAs and showed that large stretches in 3' UTRs can be contacted by the mRNA-bound proteome, with numerous putative binding sites in regions harboring disease-associated nucleotide polymorphisms. Our observations indicate the presence of a large number of mRNA binders with diverse molecular functions participating in combinatorial posttranscriptional gene-expression networks.


Subject(s)
Proteomics/methods , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Binding Sites , Cell Line , Humans , Mass Spectrometry , RNA-Binding Proteins/chemistry , Sequence Analysis, RNA
13.
Nature ; 495(7441): 333-8, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-23446348

ABSTRACT

Circular RNAs (circRNAs) in animals are an enigmatic class of RNA with unknown function. To explore circRNAs systematically, we sequenced and computationally analysed human, mouse and nematode RNA. We detected thousands of well-expressed, stable circRNAs, often showing tissue/developmental-stage-specific expression. Sequence analysis indicated important regulatory functions for circRNAs. We found that a human circRNA, antisense to the cerebellar degeneration-related protein 1 transcript (CDR1as), is densely bound by microRNA (miRNA) effector complexes and harbours 63 conserved binding sites for the ancient miRNA miR-7. Further analyses indicated that CDR1as functions to bind miR-7 in neuronal tissues. Human CDR1as expression in zebrafish impaired midbrain development, similar to knocking down miR-7, suggesting that CDR1as is a miRNA antagonist with a miRNA-binding capacity ten times higher than any other known transcript. Together, our data provide evidence that circRNAs form a large class of post-transcriptional regulators. Numerous circRNAs form by head-to-tail splicing of exons, suggesting previously unrecognized regulatory potential of coding sequences.


Subject(s)
Gene Expression Regulation , RNA/metabolism , Animals , Autoantigens/genetics , Autoantigens/metabolism , Binding Sites , Brain/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cell Line , Conserved Sequence , Female , HEK293 Cells , Humans , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA/genetics , RNA, Circular , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
14.
Nature ; 492(7429): 382-6, 2012 Dec 20.
Article in English | MEDLINE | ID: mdl-23235829

ABSTRACT

Fragile X syndrome (FXS) is a multi-organ disease that leads to mental retardation, macro-orchidism in males and premature ovarian insufficiency in female carriers. FXS is also a prominent monogenic disease associated with autism spectrum disorders (ASDs). FXS is typically caused by the loss of fragile X mental retardation 1 (FMR1) expression, which codes for the RNA-binding protein FMRP. Here we report the discovery of distinct RNA-recognition elements that correspond to the two independent RNA-binding domains of FMRP, in addition to the binding sites within the messenger RNA targets for wild-type and I304N mutant FMRP isoforms and the FMRP paralogues FXR1P and FXR2P (also known as FXR1 and FXR2). RNA-recognition-element frequency, ratio and distribution determine target mRNA association with FMRP. Among highly enriched targets, we identify many genes involved in ASD and show that FMRP affects their protein levels in human cell culture, mouse ovaries and human brain. Notably, we discovered that these targets are also dysregulated in Fmr1(-/-) mouse ovaries showing signs of premature follicular overdevelopment. These results indicate that FMRP targets share signalling pathways across different cellular contexts. As the importance of signalling pathways in both FXS and ASD is becoming increasingly apparent, our results provide a ranked list of genes as basis for the pursuit of new therapeutic targets for these neurological disorders.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Gene Expression Regulation/genetics , Protein Biosynthesis/genetics , RNA, Messenger/genetics , Regulatory Sequences, Ribonucleic Acid/genetics , Animals , Base Sequence , Binding Sites , Brain/metabolism , Child , Child Development Disorders, Pervasive/genetics , Child Development Disorders, Pervasive/metabolism , Cross-Linking Reagents , Female , HEK293 Cells , Humans , Immunoprecipitation , Mice , Molecular Sequence Data , Multigene Family , Mutation , Ovary/metabolism , Ovary/pathology , RNA, Messenger/metabolism , Response Elements/genetics , Signal Transduction , Substrate Specificity
16.
Am J Hematol ; 92(9): E513-E519, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28568895

ABSTRACT

Immunodeficient mouse models have been valuable for studies of human hematopoiesis, but high-fidelity recapitulation of erythropoiesis in most xenograft recipients remains elusive. Recently developed immunodeficient and Kit mutant mice, however, have provided a suitable background to achieve higher-level human erythropoiesis after long-term hematopoietic engraftment. While there has been some characterization of human erythropoiesis in these models, a comprehensive analysis from various human developmental stages has not yet been reported. Here, we have utilized cell surface phenotypes, morphologic analyses, and molecular studies to fully characterize human erythropoiesis from multiple developmental stages in immunodeficient and Kit mutant mouse models following long-term hematopoietic stem and progenitor cell engraftment. We show that human erythropoiesis in such models demonstrates complete maturation and enucleation, as well as developmentally appropriate globin gene expression. These results provide a framework for future studies to utilize this model system for interrogating disorders affecting human erythropoiesis and for developing improved therapeutic approaches.


Subject(s)
Erythropoiesis , Hematopoietic Stem Cell Transplantation , Mutation , Proto-Oncogene Proteins c-kit/metabolism , Animals , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Proto-Oncogene Proteins c-kit/genetics
17.
Methods ; 65(3): 302-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24096003

ABSTRACT

A key prerequisite to understand how gene regulatory processes are controlled by the interplay of RNA-binding proteins and ribonucleoprotein complexes with RNAs is the generation of comprehensive high-resolution maps of protein-RNA interactions. Recent advances in next-generation sequencing technology accelerated the development of various crosslinking and immunoprecipitation (CLIP) approaches to broadly identify RNA regions contacted by RNA-binding proteins. However these methods only consider single RNA-binding proteins and their contact sites, irrespective of the overall cis-regulatory sequence space contacted by other RNA interacting factors. Here we describe the application of protein occupancy profiling, a novel approach that globally displays the RNA contact sites of the poly(A)+ RNA-bound proteome. Protein occupancy profiling enables the generation of transcriptome-wide maps of protein-RNA interactions on polyadenylated transcripts and narrows the sequence search space for transcript regions involved in cis-regulation of gene expression in response to internal or external stimuli, altered cellular programs or disease.


Subject(s)
Gene Expression Profiling/methods , Proteome/chemistry , RNA, Messenger/chemistry , RNA-Binding Proteins/chemistry , Transcriptome/genetics , Animals , Binding Sites , Cell Line, Tumor , Embryonic Stem Cells/chemistry , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Gene Library , Human Umbilical Vein Endothelial Cells/chemistry , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Protein Binding , Proteome/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism
18.
RNA Biol ; 10(7): 1146-59, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23770886

ABSTRACT

The conserved human LIN28 RNA-binding proteins function in development, maintenance of pluripotency and oncogenesis. We used PAR-CLIP and a newly developed variant of this method, iDo-PAR-CLIP, to identify LIN28B targets as well as sites bound by the individual RNA-binding domains of LIN28B in the human transcriptome at nucleotide resolution. The position of target binding sites reflected the known structural relative orientation of individual LIN28B-binding domains, validating iDo-PAR-CLIP. Our data suggest that LIN28B directly interacts with most expressed mRNAs and members of the let-7 microRNA family. The Lin28-binding motif detected in pre-let-7 was enriched in mRNA sequences bound by LIN28B. Upon LIN28B knockdown, cell proliferation and the cell cycle were strongly impaired. Quantitative shotgun proteomics of LIN28B depleted cells revealed significant reduction of protein synthesis from its RNA targets. Computational analyses provided evidence that the strength of protein synthesis reduction correlated with the location of LIN28B binding sites within target transcripts.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Base Sequence , Binding Sites , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Line , Consensus Sequence , Humans , MicroRNAs/chemistry , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , Nucleotide Motifs , Position-Specific Scoring Matrices , Protein Binding , Protein Biosynthesis , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Messenger/chemistry , RNA-Binding Proteins/metabolism , Reproducibility of Results
19.
Nat Commun ; 13(1): 1018, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35197461

ABSTRACT

The antiviral immune response to SARS-CoV-2 infection can limit viral spread and prevent development of pneumonic COVID-19. However, the protective immunological response associated with successful viral containment in the upper airways remains unclear. Here, we combine a multi-omics approach with longitudinal sampling to reveal temporally resolved protective immune signatures in non-pneumonic and ambulatory SARS-CoV-2 infected patients and associate specific immune trajectories with upper airway viral containment. We see a distinct systemic rather than local immune state associated with viral containment, characterized by interferon stimulated gene (ISG) upregulation across circulating immune cell subsets in non-pneumonic SARS-CoV2 infection. We report reduced cytotoxic potential of Natural Killer (NK) and T cells, and an immune-modulatory monocyte phenotype associated with protective immunity in COVID-19. Together, we show protective immune trajectories in SARS-CoV2 infection, which have important implications for patient prognosis and the development of immunomodulatory therapies.


Subject(s)
COVID-19/immunology , Adult , Aged , Aged, 80 and over , Ambulatory Care , Cytokines/blood , Female , Gene Expression Regulation , Gene Regulatory Networks , Humans , Interferons/immunology , Killer Cells, Natural/immunology , Longitudinal Studies , Male , Middle Aged , Monocytes/immunology , Nasopharynx/immunology , Nasopharynx/virology , SARS-CoV-2/physiology , T-Lymphocytes/immunology
20.
mBio ; 12(3)2021 05 04.
Article in English | MEDLINE | ID: mdl-33947766

ABSTRACT

Interferon-stimulated gene products (ISGs) play a crucial role in early infection control. The ISG zinc finger CCCH-type antiviral protein 1 (ZAP/ZC3HAV1) antagonizes several RNA viruses by binding to CG-rich RNA sequences, whereas its effect on DNA viruses is less well understood. Here, we decipher the role of ZAP in the context of human cytomegalovirus (HCMV) infection, a ß-herpesvirus that is associated with high morbidity in immunosuppressed individuals and newborns. We show that expression of the two major isoforms of ZAP, ZAP-S and ZAP-L, is induced during HCMV infection and that both negatively affect HCMV replication. Transcriptome and proteome analyses demonstrated that the expression of ZAP results in reduced viral mRNA and protein levels and decelerates the progression of HCMV infection. Metabolic RNA labeling combined with high-throughput sequencing (SLAM-seq) revealed that most of the gene expression changes late in infection result from the general attenuation of HCMV. Furthermore, at early stages of infection, ZAP restricts HCMV by destabilizing a distinct subset of viral mRNAs, particularly those from the previously uncharacterized UL4-UL6 HCMV gene locus. Through enhanced cross-linking immunoprecipitation and sequencing analysis (eCLIP-seq), we identified the transcripts expressed from this HCMV locus as the direct targets of ZAP. Moreover, our data show that ZAP preferentially recognizes not only CG, but also other cytosine-rich sequences, thereby expanding its target specificity. In summary, this report is the first to reveal direct targets of ZAP during HCMV infection, which strongly indicates that transcripts from the UL4-UL6 locus may play an important role for HCMV replication.IMPORTANCE Viral infections have a large impact on society, leading to major human and economic losses and even global instability. So far, many viral infections, including human cytomegalovirus (HCMV) infection, are treated with a small repertoire of drugs, often accompanied by the occurrence of resistant mutants. There is no licensed HCMV vaccine in sight to protect those most at risk, particularly immunocompromised individuals or pregnant women who might otherwise transmit the virus to the fetus. Thus, the identification of novel intervention strategies is urgently required. In this study, we show that ZAP decelerates the viral gene expression cascade, presumably by selectively handpicking a distinct set of viral transcripts for degradation. Our study illustrates the potent role of ZAP as an HCMV restriction factor and sheds light on a possible role for UL4 and/or UL5 early during infection, paving a new avenue for the exploration of potential targets for novel therapies.


Subject(s)
Cytomegalovirus/genetics , Host Microbial Interactions/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Viral Envelope Proteins/metabolism , Viral Proteins/metabolism , Cell Line , Cells, Cultured , Cytomegalovirus/physiology , Fibroblasts/virology , HEK293 Cells , Humans , Protein Isoforms/genetics , RNA-Binding Proteins/pharmacology , Viral Envelope Proteins/genetics , Viral Proteins/genetics , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL