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










Publication year range
1.
bioRxiv ; 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38883778

ABSTRACT

Viral internal ribosomal entry sites (IRESs) form several classes that use distinct mechanisms to mediate end-independent initiation of translation. The origin of viral IRESs is a longstanding question. The simplest IRESs comprise tandem pseudoknots and occur in the intergenic region (IGR) of Dicistroviridae genomes (order Picornavirales ). Larger IGR IRESs contain additional elements that determine specific properties such as binding to the head of the ribosoma l 40S subunit. Metagenomic analyses reported here identified novel groups of structurally distinct IGR-like IRESs. The smallest of these (∼120nt long) comprise three pseudoknots and bind directly to the ribosomal P site. Others are up to 260nt long: insertions occurred at specific loci, possibly reflecting non-templated nucleotide insertion during replication. Various groups can be arranged in order, differing by the cumulative addition of single structural elements, suggesting an accretion mechanism for the structural elaboration of IRESs. Identification of chimeric IRESs implicates recombinational exchange of domains as a second mechanism for the diversification of IRES structure. Recombination likely also accounts for the presence of IGR-like IRESs at the 5'-end of some dicistrovirus-like genomes (e.g. Hangzhou dicistrovirus 3) and in the RNA genomes of Tombusviridae (order Tolivirales ), Marnaviridae (order Picornavirale s), and the 'Ripiresk' picorna-like clade (order Picornavirale s).

2.
Genes Dev ; 37(17-18): 844-860, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37821106

ABSTRACT

SARS CoV-2 nonstructural protein 1 (Nsp1) is the major pathogenesis factor that inhibits host translation using a dual strategy of impairing initiation and inducing endonucleolytic cleavage of cellular mRNAs. To investigate the mechanism of cleavage, we reconstituted it in vitro on ß-globin, EMCV IRES, and CrPV IRES mRNAs that use unrelated initiation mechanisms. In all instances, cleavage required Nsp1 and only canonical translational components (40S subunits and initiation factors), arguing against involvement of a putative cellular RNA endonuclease. Requirements for initiation factors differed for these mRNAs, reflecting their requirements for ribosomal attachment. Cleavage of CrPV IRES mRNA was supported by a minimal set of components consisting of 40S subunits and eIF3g's RRM domain. The cleavage site was located in the coding region 18 nt downstream from the mRNA entrance, indicating that cleavage occurs on the solvent side of the 40S subunit. Mutational analysis identified a positively charged surface on Nsp1's N-terminal domain (NTD) and a surface above the mRNA-binding channel on eIF3g's RRM domain that contain residues essential for cleavage. These residues were required for cleavage on all three mRNAs, highlighting general roles of the Nsp1 NTD and eIF3g's RRM domain in cleavage per se, irrespective of the mode of ribosomal attachment.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , RNA, Messenger/metabolism , Peptide Initiation Factors/genetics , Peptide Initiation Factors/metabolism , Protein Biosynthesis
3.
Nucleic Acids Res ; 51(17): 9294-9313, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37427788

ABSTRACT

Internal ribosomal entry sites (IRESs) engage with the eukaryotic translation apparatus to promote end-independent initiation. We identified a conserved class of ∼150 nt long intergenic region (IGR) IRESs in dicistrovirus genomes derived from members of the phyla Arthropoda, Bryozoa, Cnidaria, Echinodermata, Entoprocta, Mollusca and Porifera. These IRESs, exemplified by Wenling picorna-like virus 2, resemble the canonical cricket paralysis virus (CrPV) IGR IRES in comprising two nested pseudoknots (PKII/PKIII) and a 3'-terminal pseudoknot (PKI) that mimics a tRNA anticodon stem-loop base-paired to mRNA. However, they are ∼50 nt shorter than CrPV-like IRESs, and PKIII is an H-type pseudoknot that lacks the SLIV and SLV stem-loops that are primarily responsible for the affinity of CrPV-like IRESs for the 40S ribosomal subunit and that restrict initial binding of PKI to its aminoacyl (A) site. Wenling-class IRESs bound strongly to 80S ribosomes but only weakly to 40S subunits. Whereas CrPV-like IRESs must be translocated from the A site to the peptidyl (P) site by elongation factor 2 for elongation to commence, Wenling-class IRESs bound directly to the P site of 80S ribosomes, and decoding begins without a prior translocation step. A chimeric CrPV clone containing a Wenling-class IRES was infectious, confirming that the IRES functioned in cells.


Subject(s)
Internal Ribosome Entry Sites , RNA Viruses , Base Sequence , DNA, Intergenic/genetics , DNA, Intergenic/metabolism , Ribosomes/metabolism , RNA Viruses/genetics , RNA, Viral/metabolism , Protein Biosynthesis
4.
bioRxiv ; 2023 May 26.
Article in English | MEDLINE | ID: mdl-37292671

ABSTRACT

SARS CoV-2 nonstructural protein 1 (Nsp1) is the major pathogenesis factor that inhibits host translation using a dual strategy of impairing initiation and inducing endonucleolytic cleavage of cellular mRNAs. To investigate the mechanism of cleavage, we reconstituted it in vitro on ß-globin, EMCV IRES and CrPV IRES mRNAs that use unrelated initiation mechanisms. In all instances, cleavage required Nsp1 and only canonical translational components (40S subunits and initiation factors), arguing against involvement of a putative cellular RNA endonuclease. Requirements for initiation factors differed for these mRNAs, reflecting their requirements for ribosomal attachment. Cleavage of CrPV IRES mRNA was supported by a minimal set of components consisting of 40S subunits and eIF3g's RRM domain. The cleavage site was located in the coding region 18 nucleotides downstream from the mRNA entrance indicating that cleavage occurs on the solvent side of the 40S subunit. Mutational analysis identified a positively charged surface on Nsp1's N-terminal domain (NTD) and a surface above the mRNA-binding channel on eIF3g's RRM domain that contain residues essential for cleavage. These residues were required for cleavage on all three mRNAs, highlighting general roles of Nsp1-NTD and eIF3g's RRM domain in cleavage per se, irrespective of the mode of ribosomal attachment.

5.
RNA ; 29(7): 1051-1068, 2023 07.
Article in English | MEDLINE | ID: mdl-37041031

ABSTRACT

Initiation of translation on many viral mRNAs occurs by noncanonical mechanisms that involve 5' end-independent binding of ribosomes to an internal ribosome entry site (IRES). The ∼190-nt-long intergenic region (IGR) IRES of dicistroviruses such as cricket paralysis virus (CrPV) initiates translation without Met-tRNAi Met or initiation factors. Advances in metagenomics have revealed numerous dicistrovirus-like genomes with shorter, structurally distinct IGRs, such as nedicistrovirus (NediV) and Antarctic picorna-like virus 1 (APLV1). Like canonical IGR IRESs, the ∼165-nt-long NediV-like IGRs comprise three domains, but they lack key canonical motifs, including L1.1a/L1.1b loops (which bind to the L1 stalk of the ribosomal 60S subunit) and the apex of stem-loop V (SLV) (which binds to the head of the 40S subunit). Domain 2 consists of a compact, highly conserved pseudoknot (PKIII) that contains a UACUA loop motif and a protruding CrPV-like stem--loop SLIV. In vitro reconstitution experiments showed that NediV-like IRESs initiate translation from a non-AUG codon and form elongation-competent 80S ribosomal complexes in the absence of initiation factors and Met-tRNAi Met Unlike canonical IGR IRESs, NediV-like IRESs bind directly to the peptidyl (P) site of ribosomes leaving the aminoacyl (A) site accessible for decoding. The related structures of NediV-like IRESs and their common mechanism of action indicate that they exemplify a distinct class of IGR IRES.


Subject(s)
Internal Ribosome Entry Sites , Ribosomes , Internal Ribosome Entry Sites/genetics , DNA, Intergenic/genetics , DNA, Intergenic/metabolism , Ribosomes/metabolism , Peptide Initiation Factors , RNA, Transfer/chemistry , RNA, Viral/genetics , RNA, Viral/chemistry , Protein Biosynthesis
6.
EMBO J ; 41(16): e110581, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35822879

ABSTRACT

Hepatitis C virus mRNA contains an internal ribosome entry site (IRES) that mediates end-independent translation initiation, requiring a subset of eukaryotic initiation factors (eIFs). Biochemical studies revealed that direct binding of the IRES to the 40S ribosomal subunit places the initiation codon into the P site, where it base pairs with eIF2-bound Met-tRNAiMet forming a 48S initiation complex. Subsequently, eIF5 and eIF5B mediate subunit joining, yielding an elongation-competent 80S ribosome. Initiation can also proceed without eIF2, in which case Met-tRNAiMet is recruited directly by eIF5B. However, the structures of initiation complexes assembled on the HCV IRES, the transitions between different states, and the accompanying conformational changes have remained unknown. To fill these gaps, we now obtained cryo-EM structures of IRES initiation complexes, at resolutions up to 3.5 Å, that cover all major stages from the initial ribosomal association, through eIF2-containing 48S initiation complexes, to eIF5B-containing complexes immediately prior to subunit joining. These structures provide insights into the dynamic network of 40S/IRES contacts, highlight the role of IRES domain II, and reveal conformational changes that occur during the transition from eIF2- to eIF5B-containing 48S complexes and prepare them for subunit joining.


Subject(s)
Hepacivirus , Hepatitis C , Eukaryotic Initiation Factor-2/metabolism , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatitis C/metabolism , Humans , Internal Ribosome Entry Sites , Protein Biosynthesis , RNA, Viral/genetics , RNA, Viral/metabolism , Ribosomes/metabolism
7.
Cell Rep ; 33(10): 108476, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33296660

ABSTRACT

Dicistrovirus intergenic region internal ribosomal entry sites (IGR IRESs) do not require initiator tRNA, an AUG codon, or initiation factors and jumpstart translation from the middle of the elongation cycle via formation of IRES/80S complexes resembling the pre-translocation state. eEF2 then translocates the [codon-anticodon]-mimicking pseudoknot I (PKI) from ribosomal A sites to P sites, bringing the first sense codon into the decoding center. Halastavi árva virus (HalV) contains an IGR that is related to previously described IGR IRESs but lacks domain 2, which enables these IRESs to bind to individual 40S ribosomal subunits. By using in vitro reconstitution and cryoelectron microscopy (cryo-EM), we now report that the HalV IGR IRES functions by the simplest initiation mechanism that involves binding to 80S ribosomes such that PKI is placed in the P site, so that the A site contains the first codon that is directly accessible for decoding without prior eEF2-mediated translocation of PKI.


Subject(s)
Internal Ribosome Entry Sites/genetics , Peptide Chain Initiation, Translational/genetics , Positive-Strand RNA Viruses/genetics , Anticodon , Codon/metabolism , Cryoelectron Microscopy/methods , DNA, Intergenic/metabolism , Internal Ribosome Entry Sites/physiology , Peptide Chain Initiation, Translational/physiology , Peptide Elongation Factor 2/metabolism , Peptide Initiation Factors/genetics , Positive-Strand RNA Viruses/metabolism , Protein Biosynthesis/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , Ribosomes/metabolism , Virus Replication/genetics , Virus Replication/physiology , Viruses/metabolism
8.
Mol Cell ; 77(6): 1340-1349.e6, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32006463

ABSTRACT

The evolutionarily conserved Ski2-Ski3-Ski8 (Ski) complex containing the 3'→5' RNA helicase Ski2 binds to 80S ribosomes near the mRNA entrance and facilitates 3'→5' exosomal degradation of mRNA during ribosome-associated mRNA surveillance pathways. Here, we assayed Ski's activity using an in vitro reconstituted translation system and report that this complex efficiently extracts mRNA from 80S ribosomes in the 3'→5' direction in a nucleotide-by-nucleotide manner. The process is ATP dependent and can occur on pre- and post-translocation ribosomal complexes. The Ski complex can engage productively with mRNA and extract it from 80S complexes containing as few as 19 (but not 13) 3'-terminal mRNA nucleotides starting from the P site. The mRNA-extracting activity of the Ski complex suggests that its role in mRNA quality control pathways is not limited to acceleration of exosomal degradation and could include clearance of stalled ribosomes from mRNA, poising mRNA for degradation and rendering stalled ribosomes recyclable by Pelota/Hbs1/ABCE1.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Exosomes/metabolism , GTP-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Messenger/isolation & purification , Ribosomes/metabolism , ATP-Binding Cassette Transporters/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Exosomes/genetics , GTP-Binding Proteins/genetics , Humans , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/genetics
9.
Nucleic Acids Res ; 44(5): 2362-77, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26783202

ABSTRACT

Halastavi árva virus (HalV) has a positive-sense RNA genome, with an 827 nt-long 5' UTR and an intergenic region separating two open reading frames. Whereas the encoded proteins are most homologous to Dicistrovirus polyproteins, its 5' UTR is distinct. Here, we report that the HalV 5' UTR comprises small stem-loop domains separated by long single-stranded areas and a large A-rich unstructured region surrounding the initiation codon AUG828, and possesses cross-kingdom internal ribosome entry site (IRES) activity. In contrast to most viral IRESs, it does not depend on structural integrity and specific interaction of a structured element with a translational component, and is instead determined by the unstructured region flanking AUG828. eIF2, eIF3, eIF1 and eIF1A promote efficient 48S initiation complex formation at AUG828, which is reduced ∼5-fold on omission of eIF1 and eIF1A. Initiation involves direct attachment of 43S preinitiation complexes within a short window at or immediately downstream of AUG828. 40S and eIF3 are sufficient for initial binding. After attachment, 43S complexes undergo retrograde scanning, strongly dependent on eIF1 and eIF1A. eIF4A/eIF4G stimulated initiation only at low temperatures or on mutants, in which areas surrounding AUG828 had been replaced by heterologous sequences. However, they strongly promoted initiation at AUG872, yielding a proline-rich oligopeptide.


Subject(s)
Genome, Viral , Peptide Chain Initiation, Translational , RNA, Double-Stranded/metabolism , RNA, Viral/metabolism , Ribosomes/metabolism , Viral Proteins/biosynthesis , Viruses, Unclassified/metabolism , 5' Untranslated Regions , Animals , Cell-Free System , Cloning, Molecular , Codon, Initiator/chemistry , Codon, Initiator/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Gene Expression , Nucleic Acid Conformation , Open Reading Frames , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/genetics , RNA, Viral/chemistry , RNA, Viral/genetics , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reticulocytes/chemistry , Ribosomes/chemistry , Spodoptera/chemistry , Viral Proteins/genetics , Viruses, Unclassified/genetics
10.
EMBO J ; 33(1): 76-92, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24357634

ABSTRACT

Picornavirus Type 1 IRESs comprise five principal domains (dII-dVI). Whereas dV binds eIF4G, a conserved AUG in dVI was suggested to stimulate attachment of 43S ribosomal preinitiation complexes, which then scan to the initiation codon. Initiation on Type 1 IRESs also requires IRES trans-acting factors (ITAFs), and several candidates have been proposed. Here, we report the in vitro reconstitution of initiation on three Type 1 IRESs: poliovirus (PV), enterovirus 71 (EV71), and bovine enterovirus (BEV). All of them require eIF2, eIF3, eIF4A, eIF4G, eIF4B, eIF1A, and a single ITAF, poly(C) binding protein 2 (PCBP2). In each instance, initiation starts with binding of eIF4G/eIF4A. Subsequent recruitment of 43S complexes strictly requires direct interaction of their eIF3 constituent with eIF4G. The following events can differ between IRESs, depending on the stability of dVI. If it is unstructured (BEV), all ribosomes scan through dVI to the initiation codon, requiring eIF1 to bypass its AUG. If it is structured (PV, EV71), most initiation events occur without inspection of dVI, implying that its AUG does not determine ribosomal attachment.


Subject(s)
Enterovirus A, Human/physiology , Enterovirus, Bovine/physiology , Peptide Chain Initiation, Translational , Poliovirus/physiology , Codon, Initiator/metabolism , Enterovirus A, Human/genetics , Enterovirus A, Human/metabolism , Enterovirus, Bovine/genetics , Enterovirus, Bovine/metabolism , Eukaryotic Initiation Factors/metabolism , Poliovirus/genetics , Poliovirus/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Ribosomes/metabolism
11.
Nucleic Acids Res ; 39(11): 4851-65, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21306989

ABSTRACT

Type 2 internal ribosomal entry sites (IRESs) of encephalomyocarditis virus (EMCV), foot-and-mouth disease virus (FMDV) and other picornaviruses comprise five major domains H-L. Initiation of translation on these IRESs begins with specific binding of the central domain of initiation factor, eIF4G to the J-K domains, which is stimulated by eIF4A. eIF4G/eIF4A then restructure the region of ribosomal attachment on the IRES and promote recruitment of ribosomal 43S pre-initiation complexes. In addition to canonical translation factors, type 2 IRESs also require IRES trans-acting factors (ITAFs) that are hypothesized to stabilize the optimal IRES conformation that supports efficient ribosomal recruitment: the EMCV IRES is stimulated by pyrimidine tract binding protein (PTB), whereas the FMDV IRES requires PTB and ITAF(45). To test this hypothesis, we assessed the effect of ITAFs on the conformations of EMCV and FMDV IRESs by comparing their influence on hydroxyl radical cleavage of these IRESs from the central domain of eIF4G. The observed changes in cleavage patterns suggest that cognate ITAFs promote similar conformational changes that are consistent with adoption by the IRESs of comparable, more compact structures, in which domain J undergoes local conformational changes and is brought into closer proximity to the base of domain I.


Subject(s)
5' Untranslated Regions , Encephalomyocarditis virus/genetics , Eukaryotic Initiation Factors/metabolism , Foot-and-Mouth Disease Virus/genetics , Peptide Chain Initiation, Translational , RNA, Viral/chemistry , Base Sequence , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , Polypyrimidine Tract-Binding Protein/metabolism , RNA, Viral/metabolism , Ribosomes/chemistry , Ribosomes/metabolism
12.
EMBO J ; 30(1): 115-29, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21113134

ABSTRACT

Initiation codon selection in eukaryotes involves base-by-base inspection of the 5'-untranslated region of mRNA by scanning ribosomal 43S preinitiation complexes. We employed in vitro reconstitution to investigate factor requirements for this process and report that in the absence of eIF1 and DHX29, eIFs 4A, 4B and 4G promote efficient bypassing of stable stems by scanning 43S complexes and formation of 48S initiation complexes on AUG codons immediately upstream and downstream of such stems, without their unwinding. However, intact stems are not threaded through the entire mRNA Exit channel of the 40S subunit, resulting in incorrect positioning of mRNA upstream of the ribosomal P site in 48S complexes formed on AUG codons following intact stems, which renders them susceptible to dissociation by eIF1. In 48S complexes formed on AUG codons preceding intact stems, the stems are accommodated in the A site. Such aberrant complexes are destabilized by DHX29, which also ensures that mRNA enters the mRNA-binding cleft in a single-stranded form and therefore undergoes base-by-base inspection during scanning.


Subject(s)
Eukaryotic Initiation Factors/metabolism , RNA, Messenger/metabolism , Ribosomes/metabolism , Animals , Base Sequence , Codon, Initiator , Humans , Protein Biosynthesis , RNA, Messenger/genetics , Ribosome Subunits/genetics , Ribosome Subunits/metabolism , Ribosomes/genetics , beta-Globins/genetics
13.
EMBO J ; 27(7): 1060-72, 2008 Apr 09.
Article in English | MEDLINE | ID: mdl-18337746

ABSTRACT

Specific interactions of the classical swine fever virus internal ribosomal entry site (IRES) with 40S ribosomal subunits and eukaryotic translation initiation factor (eIF)3 enable 43S preinitiation complexes containing eIF3 and eIF2-GTP-Met-tRNA(iMet) to bind directly to the initiation codon, yielding 48S initiation complexes. We report that eIF5B or eIF5B/eIF3 also promote Met-tRNA(iMet) binding to IRES-40S complexes, forming 48S complexes that can assemble elongation-competent ribosomes. Although 48S complexes assembled both by eIF2/eIF3- and eIF5B/eIF3-mediated Met-tRNA(iMet) recruitment were destabilized by eIF1, dissociation of 48S complexes formed with eIF2 could be out-competed by efficient subunit joining. Deletion of IRES domain II, which is responsible for conformational changes induced in 40S subunits by IRES binding, eliminated the sensitivity of 48S complexes assembled by eIF2/eIF3- and eIF5B/eIF3-mediated mechanisms to eIF1-induced destabilization. However, 48S complexes formed by the eIF5B/eIF3-mediated mechanism on the truncated IRES could not undergo efficient subunit joining, as reported previously for analogous complexes assembled with eIF2, indicating that domain II is essential for general conformational changes in 48S complexes, irrespective of how they were assembled, that are required for eIF5-induced hydrolysis of eIF2-bound GTP and/or subunit joining.


Subject(s)
Classical Swine Fever Virus/genetics , Eukaryotic Initiation Factor-2/chemistry , Eukaryotic Initiation Factor-2/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Animals , Hepacivirus/genetics , Peptide Chain Initiation, Translational , Phosphorylation , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/metabolism , Rabbits , Ribosomes/metabolism
14.
Mol Cell Biol ; 25(3): 1089-99, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15657435

ABSTRACT

Animal cells counteract oxidative stress and electrophilic attack through coordinated expression of a set of detoxifying and antioxidant enzyme genes mediated by transcription factor Nrf2. In unstressed cells, Nrf2 appears to be sequestered in the cytoplasm via association with an inhibitor protein, Keap1. Here, by using the yeast two-hybrid screen, human Keap1 has been identified as a partner of the nuclear protein prothymosin alpha. The in vivo and in vitro data indicated that the prothymosin alpha-Keap1 interaction is direct, highly specific, and functionally relevant. Furthermore, we showed that Keap1 is a nuclear-cytoplasmic shuttling protein equipped with a nuclear export signal that is important for its inhibitory action. Prothymosin alpha was able to liberate Nrf2 from the Nrf2-Keap1 inhibitory complex in vitro through competition with Nrf2 for binding to the same domain of Keap1. In vivo, the level of Nrf2-dependent transcription was correlated with the intracellular level of prothymosin alpha by using prothymosin alpha overproduction and mRNA interference approaches. Our data attribute to prothymosin alpha the role of intranuclear dissociator of the Nrf2-Keap1 complex, thus revealing a novel function for prothymosin alpha and adding a new dimension to the molecular mechanisms underlying expression of oxidative stress-protecting genes.


Subject(s)
DNA-Binding Proteins/metabolism , Oxidative Stress/genetics , Protein Precursors/metabolism , Proteins/metabolism , Thymosin/analogs & derivatives , Thymosin/metabolism , Trans-Activators/metabolism , Transcriptional Activation/genetics , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/physiology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Oxidative Stress/physiology , Protein Binding , RNA, Small Interfering/metabolism , Transcriptional Activation/physiology , Tumor Cells, Cultured , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...