Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 11737, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37474590

ABSTRACT

Some strains of the inherited bacterium Wolbachia have been shown to be effective at reducing the transmission of dengue virus (DENV) and other RNA viruses by Aedes aegypti in both laboratory and field settings and are being deployed for DENV control. The degree of virus inhibition varies between Wolbachia strains. Density and tissue tropism can contribute to these differences but there are also indications that this is not the only factor involved: for example, strains wAu and wAlbA are maintained at similar intracellular densities but only wAu produces strong DENV inhibition. We previously reported perturbations in lipid transport dynamics, including sequestration of cholesterol in lipid droplets, with strains wMel/wMelPop in Ae. aegypti. To further investigate the cellular basis underlying these differences, proteomic analysis of midguts was carried out on Ae. aegypti lines carrying strains wAu and wAlbA: with the hypothesis that differences in perturbations may underline Wolbachia-mediated antiviral activity. Surprisingly, wAu-carrying midguts not only showed distinct proteome perturbations when compared to non-Wolbachia carrying and wAlbA-carrying midguts but also wMel-carrying midguts. There are changes in RNA processing pathways and upregulation of a specific set of RNA-binding proteins in the wAu-carrying line, including genes with known antiviral activity. Lipid transport and metabolism proteome changes also differ between strains, and we show that strain wAu does not produce the same cholesterol sequestration phenotype as wMel. Moreover, in contrast to wMel, wAu antiviral activity was not rescued by cyclodextrin treatment. Together these results suggest that wAu could show unique features in its inhibition of arboviruses compared to previously characterized Wolbachia strains.


Subject(s)
Aedes , Dengue Virus , Wolbachia , Animals , Dengue Virus/physiology , Proteome , Wolbachia/physiology , Antiviral Agents , Proteomics , Lipids
2.
Trends Biochem Sci ; 47(1): 23-38, 2022 01.
Article in English | MEDLINE | ID: mdl-34509361

ABSTRACT

RNA viruses interact with a wide range of cellular RNA-binding proteins (RBPs) during their life cycle. The prevalence of these host-virus interactions has been highlighted by new methods that elucidate the composition of viral ribonucleoproteins (vRNPs). Applied to 11 viruses so far, these approaches have revealed hundreds of cellular RBPs that interact with viral (v)RNA in infected cells. However, consistency across methods is limited, raising questions about methodological considerations when designing and interpreting these studies. Here, we discuss these caveats and, through comparing available vRNA interactomes, describe RBPs that are consistently identified as vRNP components and outline their potential roles in infection. In summary, these novel approaches have uncovered a new universe of host-virus interactions holding great therapeutic potential.


Subject(s)
Proteome , RNA, Viral , Cell Communication , Host Microbial Interactions , Host-Pathogen Interactions , Proteome/metabolism , RNA, Viral/genetics , Ribonucleoproteins/metabolism
3.
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
4.
Nat Protoc ; 16(1): 27-60, 2021 01.
Article in English | MEDLINE | ID: mdl-33208978

ABSTRACT

Interactions between RNA-binding proteins (RBPs) and RNAs are critical to cell biology. However, methods to comprehensively and quantitatively assess these interactions within cells were lacking. RNA interactome capture (RIC) uses in vivo UV crosslinking, oligo(dT) capture, and proteomics to identify RNA-binding proteomes. Recent advances have empowered RIC to quantify RBP responses to biological cues such as metabolic imbalance or virus infection. Enhanced RIC exploits the stronger binding of locked nucleic acid (LNA)-containing oligo(dT) probes to poly(A) tails to maximize RNA capture selectivity and efficiency, profoundly improving signal-to-noise ratios. The subsequent analytical use of SILAC and TMT proteomic approaches, together with high-sensitivity sample preparation and tailored statistical data analysis, substantially improves RIC's quantitative accuracy and reproducibility. This optimized approach is an extension of the original RIC protocol. It takes 3 d plus 2 weeks for proteomics and data analysis and will enable the study of RBP dynamics under different physiological and pathological conditions.


Subject(s)
Proteomics/methods , RNA-Binding Proteins/metabolism , RNA/metabolism , Humans , Jurkat Cells , Oligonucleotides/metabolism , Protein Binding , Workflow
5.
Front Immunol ; 10: 2791, 2019.
Article in English | MEDLINE | ID: mdl-31849970

ABSTRACT

Virus infected immune cells can rapidly respond to the invader by activating the inflammasome and as a consequence release proinflammatory cytokines and eventually die by pyroptosis. In human adenovirus-5 (Ad5) infected THP-1 cells, inhibition of NLRP3 inflammasome activation was demonstrated by a decreased secretion of HMGB1 and matured forms of caspase-1and IL-1ß. An Ad5 mutant virus defective in expression of the non-coding VA RNAI failed to inhibit the NLRP3 inflammasome and in addition displayed formation of ASC specks and increased cell lysis. Importantly, in vitro synthesized VA RNAI was able to inhibit the NLRP3 inflammasome activity in THP-1 cells in the absence of an Ad5 infection, suggesting that VA RNAI binding to PKR and blocking its function is sufficient for inhibition of the NLRP3 inflammasome. Although the inhibition of NLRP3 inflammasome activation required the phylogenetically conserved base paired tetranucleotide sequence in the central stem of VA RNAI, we demonstrate that PKR binding to VA RNAI primarily protected the apical stem, but not the tetranucleotide sequence itself. VA RNAI did not influence the interaction between PKR and NLRP3. In contrast, we describe a novel interaction between PKR and ASC and further show that VA RNAI inhibited ASC phosphorylation and oligomerization. Collectively, our results indicate a novel role for Ad5 VA RNAI as an inhibitor of NLRP3 inflammasome activation by targeting the cellular pro-inflammatory protein PKR.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Multimerization , RNA, Viral/genetics , CARD Signaling Adaptor Proteins/chemistry , Cytokines/metabolism , Ectopic Gene Expression , Humans , Inflammation Mediators/metabolism , Protein Binding , RNA, Viral/chemistry , THP-1 Cells
6.
Proc Natl Acad Sci U S A ; 115(16): E3808-E3816, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29610341

ABSTRACT

The zinc finger CCCH-type containing 11A (ZC3H11A) gene encodes a well-conserved zinc finger protein that may function in mRNA export as it has been shown to associate with the transcription export (TREX) complex in proteomic screens. Here, we report that ZC3H11A is a stress-induced nuclear protein with RNA-binding capacity that localizes to nuclear splicing speckles. During an adenovirus infection, the ZC3H11A protein and splicing factor SRSF2 relocalize to nuclear regions where viral DNA replication and transcription take place. Knockout (KO) of ZC3H11A in HeLa cells demonstrated that several nuclear-replicating viruses are dependent on ZC3H11A for efficient growth (HIV, influenza virus, herpes simplex virus, and adenovirus), whereas cytoplasmic replicating viruses are not (vaccinia virus and Semliki Forest virus). High-throughput sequencing of ZC3H11A-cross-linked RNA showed that ZC3H11A binds to short purine-rich ribonucleotide stretches in cellular and adenoviral transcripts. We show that the RNA-binding property of ZC3H11A is crucial for its function and localization. In ZC3H11A KO cells, the adenovirus fiber mRNA accumulates in the cell nucleus. Our results suggest that ZC3H11A is important for maintaining nuclear export of mRNAs during stress and that several nuclear-replicating viruses take advantage of this mechanism to facilitate their replication.


Subject(s)
Cell Nucleus/virology , Nuclear Proteins/physiology , RNA, Messenger/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/physiology , Virus Replication , Zinc Fingers/physiology , Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , Binding Sites , Biological Transport , CRISPR-Cas Systems , Cell Nucleus/metabolism , Cytoplasm/virology , Gene Knockout Techniques , HeLa Cells , Heat-Shock Response/genetics , Heat-Shock Response/physiology , High-Throughput Nucleotide Sequencing , Host-Pathogen Interactions , Humans , Nuclear Proteins/antagonists & inhibitors , Protein Domains , Protein Stability , RNA-Binding Proteins/antagonists & inhibitors , Serine-Arginine Splicing Factors/physiology
7.
RNA ; 23(11): 1700-1711, 2017 11.
Article in English | MEDLINE | ID: mdl-28839112

ABSTRACT

Here we show that the adenovirus major late promoter produces a 31-nucleotide transcriptional start site small RNA (MLP-TSS-sRNA) that retains the 7-methylguanosine (m7G)-cap and is incorporated onto Ago2-containing RNA-induced silencing complexes (RISC) in human adenovirus-37 infected cells. RNA polymerase II CLIP (UV-cross linking immunoprecipitation) experiments suggest that the MLP-TSS-sRNA is produced by promoter proximal stalling/termination of RNA polymerase II transcription at the site of the small RNA 3' end. The MLP-TSS-sRNA is highly stable in cells and functionally active, down-regulating complementary targets in a sequence and dose-dependent manner. The MLP-TSS-sRNA is transcribed from the opposite strand to the adenoviral DNA polymerase and preterminal protein mRNAs, two essential viral replication proteins. We show that the MLP-TSS-sRNA act in trans to reduce DNA polymerase and preterminal protein mRNA expression. As a consequence of this, the MLP-TSS-sRNA has an inhibitory effect on the efficiency of viral DNA replication. Collectively, our results suggest that this novel sRNA may serve a regulatory function controlling viral genome replication during a lytic and/or persistent adenovirus infection in its natural host.


Subject(s)
Adenoviruses, Human/genetics , Adenoviruses, Human/metabolism , Argonaute Proteins/metabolism , DNA Replication/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , Virus Replication/genetics , Argonaute Proteins/genetics , Cell Line , Genes, Viral , HEK293 Cells , HeLa Cells , Humans , Nucleic Acid Conformation , Promoter Regions, Genetic , RNA Caps/chemistry , RNA Caps/genetics , RNA Caps/metabolism , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/chemistry , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism , Transcription Initiation Site
8.
PLoS One ; 12(5): e0177275, 2017.
Article in English | MEDLINE | ID: mdl-28542273

ABSTRACT

We have used high-throughput small RNA sequencing to characterize viral small RNA expression in purified tonsillar B and T lymphocytes isolated from patients tested positive for Epstein-Barr virus (EBV) or human adenovirus (HAdV) infections, respectively. In the small set of patients analyzed, the expression profile of EBV and HAdV miRNAs could not distinguish between patients diagnosed with tonsillar hypertrophy or chronic/recurrent tonsillitis. The EBV miR-BART expression profile among the patients diagnosed with tonsillar diseases resembles most closely the pattern seen in EBV+ tumors (Latency II/I). The miR-BARTs that appear to be absent in normal EBV infected cells are essentially all detectable in the diseased tonsillar B lymphocytes. In the EBV+ B cells we detected 44 EBV miR-BARTs derived from the proposed BART precursor hairpins whereof five are not annotated in miRBase v21. One previously undetected miRNA, BART16b-5p, originates from the miR-BART16 precursor hairpin as an alternative 5´ miR-BART16 located precisely upstream of the annotated miR-BART16-5p. Further, our analysis revealed an extensive sequence variation among the EBV miRNAs with isomiRs having a constant 5´ end but alternative 3´ ends. A range of small RNAs was also detected from the terminal stem of the EBER RNAs and the 3´ part of v-snoRNA1. During a lytic HAdV infection in established cell lines the terminal stem of the viral non-coding VA RNAs are processed to highly abundant viral miRNAs (mivaRNAs). In contrast, mivaRNA expression in HAdV positive tonsillar T lymphocytes was very low. The small RNA profile further showed that the 5´ mivaRNA from VA RNAI and the 3´ mivaRNA from VA RNAII were as predicted, whereas the 3´ mivaRNA from VA RNAI showed an aberrant processing upstream of the expected Dicer cleavage site.


Subject(s)
Adenoviruses, Human/genetics , B-Lymphocytes/virology , Gene Expression Regulation, Viral/genetics , Herpesvirus 4, Human/genetics , MicroRNAs/genetics , Palatine Tonsil/virology , T-Lymphocytes/virology , Adolescent , Adult , Child , Child, Preschool , Epstein-Barr Virus Infections/virology , Female , Humans , Male , RNA, Viral/genetics , Virus Latency/genetics , Young Adult
9.
Nucleic Acids Res ; 45(4): 1731-1742, 2017 02 28.
Article in English | MEDLINE | ID: mdl-27899607

ABSTRACT

The adenovirus L4-22K protein both activates and suppresses transcription from the adenovirus major late promoter (MLP) by binding to DNA elements located downstream of the MLP transcriptional start site: the so-called DE element (positive) and the R1 region (negative). Here we show that L4-22K preferentially binds to the RNA form of the R1 region, both to the double-stranded RNA and the single-stranded RNA of the same polarity as the nascent MLP transcript. Further, L4-22K binds to a 5΄-CAAA-3΄ motif in the single-stranded RNA, which is identical to the sequence motif characterized for L4-22K DNA binding. L4-22K binding to single-stranded RNA results in an enhancement of U1 snRNA recruitment to the major late first leader 5΄ splice site. This increase in U1 snRNA binding results in a suppression of MLP transcription and a concurrent stimulation of major late first intron splicing.


Subject(s)
Gene Expression Regulation , RNA Splicing , RNA-Binding Proteins/metabolism , Transcription, Genetic , Viral Nonstructural Proteins/metabolism , Adenoviruses, Human/genetics , Adenoviruses, Human/metabolism , Base Sequence , Binding Sites , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Gene Expression Regulation, Viral , Gene Order , Humans , Introns , Models, Biological , Nucleotide Motifs , Protein Binding , RNA Splice Sites , Ribonucleoprotein, U1 Small Nuclear/metabolism
10.
Virology ; 485: 25-35, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26196231

ABSTRACT

Human adenoviruses (HAdVs) encode for multifunctional non-coding virus-associated (VA) RNAs, which function as powerful suppressors of the cellular interferon (IFN) and RNA interference (RNAi) systems. In this study we tested the ability of various plant and animal virus encoded RNAi and IFN suppressor proteins to functionally substitute for the HAdV-5 VA RNAI. Our results revealed that only the Vaccinia virus (VACV) E3L protein was able to substitute for the HAdV-5 VA RNAI functions in virus-infected cells. Interestingly, the E3L protein rescues the translational defect but does not stimulate viral capsid mRNA accumulation observed with VA RNA. We further show that the E3L C-terminal region containing the dsRNA-binding domain is needed to enhance VA RNAI mutant virus replication. Additionally, we show that the HAdV-4 and HAdV-37 VA RNAI are more effective than the HAdV-5 VA RNAI in rescuing virus replication.


Subject(s)
Adenoviruses, Human/genetics , Gene Expression Regulation, Viral , RNA Interference , RNA, Messenger/genetics , RNA, Viral/genetics , RNA-Binding Proteins/genetics , Vaccinia virus/genetics , Viral Proteins/genetics , Adenoviruses, Human/metabolism , Capsid/metabolism , Genetic Complementation Test , HEK293 Cells , Host-Pathogen Interactions , Humans , Oligopeptides/metabolism , Plasmids/chemistry , Plasmids/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , RNA, Viral/antagonists & inhibitors , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Serogroup , Transfection , Vaccinia virus/metabolism , Viral Proteins/metabolism , Virus Replication
11.
FEBS Lett ; 589(12): 1383-8, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25907535

ABSTRACT

Cytochrome P450 family member CYP51A1 is a key enzyme in cholesterol biosynthesis whose deregulation is implicated in numerous diseases, including retinal degeneration. Here we describe that HAdV-37 infection leads to downregulation of CYP51A1 expression and overexpression of its antisense non-coding Alu element (AluCYP51A1) in retinal pigment epithelium (RPE) cells. This change in gene expression is associated with a reversed accumulation of a positive histone mark at the CYP51A1 and AluCYP51A1 promoters. Further, transient AluCYP51A1 RNA overexpression correlates with reduced CYP51A1 mRNA accumulation. Collectively, our data suggest that AluCYP51A1 might control CYP51A1 gene expression in HAdV-37-infected RPE cells.


Subject(s)
Mechanical Phenomena , Microscopy, Atomic Force/methods , Molecular Imaging/methods
12.
PLoS One ; 9(8): e105746, 2014.
Article in English | MEDLINE | ID: mdl-25144466

ABSTRACT

Human adenoviruses (HAds) encode for one or two highly abundant virus-associated RNAs, designated VA RNAI and VA RNAII, which fold into stable hairpin structures resembling miRNA precursors. Here we show that the terminal stem of the VA RNAs originating from Ad4, Ad5, Ad11 and Ad37, all undergo Dicer dependent processing into virus-specific miRNAs (so-called mivaRNAs). We further show that the mivaRNA duplex is subjected to a highly asymmetric RISC loading with the 3'-strand from all VA RNAs being the favored strand, except for the Ad37 VA RNAII, where the 5'-mivaRNAII strand was preferentially assembled into RISC. Although the mivaRNA seed sequences are not fully conserved between the HAds a bioinformatics prediction approach suggests that a large fraction of the VA RNAII-, but not the VA RNAI-derived mivaRNAs still are able to target the same cellular genes. Using small RNA deep sequencing we demonstrate that the Dicer processing event in the terminal stem of the VA RNAs is not unique and generates 3'-mivaRNAs with a slight variation of the position of the 5' terminal nucleotide in the RISC loaded guide strand. Also, we show that all analyzed VA RNAs, except Ad37 VA RNAI and Ad5 VA RNAII, utilize an alternative upstream A start site in addition to the classical +1 G start site. Further, the 5'-mivaRNAs with an A start appears to be preferentially incorporated into RISC. Although the majority of mivaRNA research has been done using Ad5 as the model system our analysis demonstrates that the mivaRNAs expressed in Ad11- and Ad37-infected cells are the most abundant mivaRNAs associated with Ago2-containing RISC. Collectively, our results show an unexpected variability in Dicer processing of the VA RNAs and a serotype-specific loading of mivaRNAs into Ago2-based RISC.


Subject(s)
Adenoviridae/metabolism , MicroRNAs/metabolism , RNA, Viral/metabolism , Adenoviridae/genetics , HeLa Cells , Humans , MicroRNAs/genetics , Nucleic Acid Conformation , RNA, Viral/genetics , Ribonuclease III/genetics , Ribonuclease III/metabolism
13.
J Endod ; 40(3): 446-50, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24565669

ABSTRACT

INTRODUCTION: The aim of this study was to compare by scanning electron microscopy the presence of smear layer and debris on root canal walls after preparation with the single-file system WaveOne (Dentsply Maillefer, Ballaigues, Switzerland) versus the rotary ProTaper system (Dentsply Maillefer, Ballaigues, Switzerland) under 2 final irrigant regimens. METHODS: Forty freshly extracted single-rooted human teeth were randomly divided into 4 groups (n = 10). The ProTaper and ProTaper and rotary CanalBrush (Coltène Whaledent GmbH+ Co KG, Langenau, Germany) groups were instrumented with the ProTaper system. Groups WaveOne and WaveOne and rotary CanalBrush were instrumented with the WaveOne system. The irrigant in all groups was 2 mL 5.25% sodium hypochlorite (NaOCl) solution, whereas the final irrigation after preparation in the ProTaper and WaveOne groups was 1 mL Smear Clear solution (Sybron Endo, Orange, CA) and then 5.25% NaOCl applied with a plastic syringe, and in the ProTaper and rotary CanalBrush and WaveOne and rotary CanalBrush groups, it was 1 mL Smear Clear solution and then 5.25% NaOCl (rotary CanalBrush agitation). Roots were processed for scanning electron microscopic examination for debris and smear layer scoring. Data were statistically analyzed. RESULTS: All groups showed more efficient smear layer and debris removal coronally than in the middle and apical regions, whereas the mean total debris score and the mean smear layer score in all groups were less in the WaveOne and rotary CanalBrush groups than the ProTaper and rotary CanalBrush and the WaveOne and ProTaper groups. CONCLUSIONS: Using the rotary CanalBrush in canals prepared with WaveOne produced the cleanest canal walls, and the WaveOne system gave superior results compared with the ProTaper system.


Subject(s)
Dental Pulp Cavity/drug effects , Edetic Acid/therapeutic use , Root Canal Irrigants/therapeutic use , Root Canal Preparation/instrumentation , Smear Layer/drug therapy , Dental Pulp Cavity/ultrastructure , Dentin/drug effects , Dentin/ultrastructure , Equipment Design , Humans , Materials Testing , Microscopy, Electron, Scanning , Random Allocation , Smear Layer/pathology , Sodium Hypochlorite/therapeutic use , Tooth Apex/drug effects , Tooth Apex/ultrastructure , Treatment Outcome
14.
Nucleic Acids Res ; 41(9): 4802-12, 2013 May.
Article in English | MEDLINE | ID: mdl-23525465

ABSTRACT

At late times during a lytic infection human adenovirus type 5 produces ∼10(8) copies per cell of virus-associated RNA I (VA RNAI). This short highly structured RNA polymerase III transcript has previously been shown to be essential for lytic virus growth. A fraction of VA RNAI is processed by Dicer into small RNAs, so-called mivaRNAIs, which are efficiently incorporated into the RNA-induced silencing complex. Here, we constructed recombinant adenoviruses with mutations in the seed sequence of both the 5'- and the 3'-strand of the mivaRNAI duplex. The results showed that late viral protein synthesis, as well as new virus progeny formation, was essentially unaffected by the seed sequence mutations under lytic replicative conditions in HeLa or HEK293 cells. Collectively, our results suggest that either strand of the mivaRNAI duplex does not have target mRNA interactions that are critical for the establishment of virus growth under lytic conditions. Further, by depletion of protein kinase R (PKR) in HEK293 cells, we show that the suppressive effect of VA RNAI on the interferon-induced PKR pathway is most critical for late gene expression.


Subject(s)
Adenoviruses, Human/genetics , MicroRNAs/metabolism , RNA, Viral/metabolism , Adenoviruses, Human/growth & development , Base Sequence , Genetic Vectors , HEK293 Cells , HeLa Cells , Humans , MicroRNAs/chemistry , Molecular Sequence Data , Mutation , RNA, Viral/chemistry , RNA-Induced Silencing Complex/metabolism , eIF-2 Kinase/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...