Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4385, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782906

ABSTRACT

The parasite Toxoplasma gondii persists in its hosts by converting from replicating tachyzoites to latent bradyzoites housed in tissue cysts. The molecular mechanisms that mediate T. gondii differentiation remain poorly understood. Through a mutagenesis screen, we identified translation initiation factor eIF1.2 as a critical factor for T. gondii differentiation. A F97L mutation in eIF1.2 or the genetic ablation of eIF1.2 (∆eif1.2) markedly impeded bradyzoite cyst formation in vitro and in vivo. We demonstrated, at single-molecule level, that the eIF1.2 F97L mutation impacts the scanning process of the ribosome preinitiation complex on a model mRNA. RNA sequencing and ribosome profiling experiments unveiled that ∆eif1.2 parasites are defective in upregulating bradyzoite induction factors BFD1 and BFD2 during stress-induced differentiation. Forced expression of BFD1 or BFD2 significantly restored differentiation in ∆eif1.2 parasites. Together, our findings suggest that eIF1.2 functions by regulating the translation of key differentiation factors necessary to establish chronic toxoplasmosis.


Subject(s)
Toxoplasma , Toxoplasma/metabolism , Toxoplasma/genetics , Animals , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Toxoplasmosis/parasitology , Toxoplasmosis/metabolism , Mice , Mutation , Ribosomes/metabolism , Protein Biosynthesis , Female , RNA, Messenger/metabolism , RNA, Messenger/genetics , Cell Differentiation , Humans
2.
bioRxiv ; 2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37961607

ABSTRACT

The parasite Toxoplasma gondii persists in its hosts by converting from replicating tachyzoites to latent bradyzoites housed in tissue cysts. The molecular mechanisms that mediate T. gondii differentiation remain poorly understood. Through a mutagenesis screen, we identified translation initiation factor eIF1.2 as a critical factor for T. gondii differentiation. A F97L mutation in eIF1.2 or the genetic ablation of eIF1.2 (Δ eIF1.2 ) markedly impeded bradyzoite cyst formation in vitro and in vivo . We demonstrated, at single-molecule level, that the eIF1.2 F97L mutation impacts the scanning process of the ribosome preinitiation complex on a model mRNA. RNA sequencing and ribosome profiling experiments unveiled that Δ eIF1.2 parasites are defective in the upregulating bradyzoite induction factors BFD1 and BFD2 during stress-induced differentiation. Forced expression of BFD1 or BFD2 significantly restored differentiation in Δ eIF1.2 parasites. Together, our findings suggest that eIF1.2 functions by regulating the translation of key differentiation factors necessary to establish chronic toxoplasmosis.

3.
Nucleic Acids Res ; 50(14): 8240-8261, 2022 08 12.
Article in English | MEDLINE | ID: mdl-35871304

ABSTRACT

mRNA 5' cap recognition by eIF4F is a key element of eukaryotic translational control. Kinetic differences in eIF4F-mRNA interactions have long been proposed to mediate translation-efficiency differences between mRNAs, and recent transcriptome-wide studies have revealed significant heterogeneity in eIF4F engagement with differentially-translated mRNAs. However, detailed kinetic information exists only for eIF4F interactions with short model RNAs. We developed and applied single-molecule fluorescence approaches to directly observe real-time Saccharomyces cerevisiae eIF4F subunit interactions with full-length polyadenylated mRNAs. We found that eIF4E-mRNA association rates linearly anticorrelate with mRNA length. eIF4G-mRNA interaction accelerates eIF4E-mRNA association in proportion to mRNA length, as does an eIF4F-independent activity of eIF4A, though cap-proximal secondary structure still plays an important role in defining the final association rates. eIF4F-mRNA interactions remained dominated by effects of eIF4G, but were modulated to different extents for different mRNAs by the presence of eIF4A and ATP. We also found that eIF4A-catalyzed ATP hydrolysis ejects eIF4E, and likely eIF4E•eIF4G from the mRNA after initial eIF4F•mRNA complex formation, suggesting a mechanism to prepare the mRNA 5' end for ribosome recruitment. Our results support a role for mRNA-specific, factor-driven eIF4F association rates in kinetically controlling translation.


Subject(s)
Eukaryotic Initiation Factor-4F/metabolism , RNA Helicases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphate , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
4.
bioRxiv ; 2021 May 27.
Article in English | MEDLINE | ID: mdl-34075378

ABSTRACT

Coronaviruses initiate translation through recognition of the viral RNA 5' m 7 GpppA m cap by translation factor eIF4F. eIF4F is a heterotrimeric protein complex with cap-binding, RNA-binding, and RNA helicase activities. Modulating eIF4F function through cellular regulation or small-molecule inhibition impacts coronavirus replication, including for SARS-CoV-2. Translation initiation involves highly coordinated dynamics of translation factors with messenger or viral RNA. However, how the eIF4F subunits coordinate on the initiation timescale to define cap-binding efficiency remains incompletely understood. Here we report that translation supported by the SARS-CoV-2 5'-UTR is highly sensitive to eIF4A inhibition by rocaglamide. Through a single-molecule fluorescence approach that reports on eIF4E-cap interaction, we dissect how eIF4F subunits contribute to cap-recognition efficiency on the SARS-CoV-2 5' UTR. We find that free eIF4A enhances cap accessibility for eIF4E binding, but eIF4G alone does not change the kinetics of eIF4E-RNA interaction. Conversely, formation of the full eIF4F complex significantly alters eIF4E-cap interaction, suggesting that coordinated eIF4E and eIF4A activities establish the net eIF4F-cap recognition efficiency. Moreover, the eIF4F complex formed with phosphomimetic eIF4E(S209D) binds the viral UTR more efficiently than with wild-type eIF4E. These results highlight a dynamic interplay of eIF4F subunits and mRNA that determines cap-recognition efficiency.

5.
Nat Commun ; 12(1): 2490, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33941775

ABSTRACT

DNA methylation and trimethylated histone H4 Lysine 20 (H4K20me3) constitute two important heterochromatin-enriched marks that frequently cooperate in silencing repetitive elements of the mammalian genome. However, it remains elusive how these two chromatin modifications crosstalk. Here, we report that DNA methyltransferase 1 (DNMT1) specifically 'recognizes' H4K20me3 via its first bromo-adjacent-homology domain (DNMT1BAH1). Engagement of DNMT1BAH1-H4K20me3 ensures heterochromatin targeting of DNMT1 and DNA methylation at LINE-1 retrotransposons, and cooperates with the previously reported readout of histone H3 tail modifications (i.e., H3K9me3 and H3 ubiquitylation) by the RFTS domain to allosterically regulate DNMT1's activity. Interplay between RFTS and BAH1 domains of DNMT1 profoundly impacts DNA methylation at both global and focal levels and genomic resistance to radiation-induced damage. Together, our study establishes a direct link between H4K20me3 and DNA methylation, providing a mechanism in which multivalent recognition of repressive histone modifications by DNMT1 ensures appropriate DNA methylation patterning and genomic stability.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation/genetics , Heterochromatin/metabolism , Histones/metabolism , Long Interspersed Nucleotide Elements/genetics , Animals , Cell Line , Crystallography, X-Ray , Genome/genetics , Genomic Instability/genetics , Heterochromatin/genetics , Mice
6.
J Am Chem Soc ; 142(51): 21249-21253, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33315378

ABSTRACT

Dynamic RNA-protein interactions underpin numerous molecular control mechanisms in biology. However, relatively little is known about the kinetic landscape of protein interactions with full-length RNAs. The extent to which interaction kinetics vary for the same RNA element across the transcriptome and the molecular determinants of variability therefore remain poorly defined. Moreover, it is unclear how one protein-RNA interaction might be transduced by RNA to kinetically impact a second. We report a parallelized, real-time single-molecule fluorescence assay for protein interaction kinetics on eukaryotic mRNA populations obtained from cells. We observed ∼100-fold heterogeneity for interactions of the translation initiation factor eIF4E with the universal mRNA 5' cap structure, dominated by steric effects on barrier-height variability for association. We also found that an RNA helicase, eIF4A, independently accelerated eIF4E-cap association. These data support a kinetic mechanism for how mRNA can determine the sensitivity of its translation to reduction in cellular eIF4E concentrations. They also support the view that global RNA structure significantly modulates protein-RNA interaction dynamics and can facilitate real-time communication between protein interactions at distinct sites.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Single Molecule Imaging , Transcriptome , Kinetics , Protein Binding
7.
Nat Struct Mol Biol ; 27(10): 875-885, 2020 10.
Article in English | MEDLINE | ID: mdl-32778820

ABSTRACT

Suppressing cellular signal transducers of transcription 2 (STAT2) is a common strategy that viruses use to establish infections, yet the detailed mechanism remains elusive, owing to a lack of structural information about the viral-cellular complex involved. Here, we report the cryo-EM and crystal structures of human STAT2 (hSTAT2) in complex with the non-structural protein 5 (NS5) of Zika virus (ZIKV) and dengue virus (DENV), revealing two-pronged interactions between NS5 and hSTAT2. First, the NS5 methyltransferase and RNA-dependent RNA polymerase (RdRP) domains form a conserved interdomain cleft harboring the coiled-coil domain of hSTAT2, thus preventing association of hSTAT2 with interferon regulatory factor 9. Second, the NS5 RdRP domain also binds the amino-terminal domain of hSTAT2. Disruption of these ZIKV NS5-hSTAT2 interactions compromised NS5-mediated hSTAT2 degradation and interferon suppression, and viral infection under interferon-competent conditions. Taken together, these results clarify the mechanism underlying the functional antagonism of STAT2 by both ZIKV and DENV.


Subject(s)
STAT2 Transcription Factor/chemistry , STAT2 Transcription Factor/metabolism , Viral Nonstructural Proteins/chemistry , Cryoelectron Microscopy , Crystallography, X-Ray , Cytoplasm/metabolism , HEK293 Cells , Host-Pathogen Interactions , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Models, Molecular , Protein Conformation , STAT2 Transcription Factor/genetics , Viral Nonstructural Proteins/metabolism , Zika Virus Infection/virology
8.
Proc Natl Acad Sci U S A ; 117(31): 18439-18447, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32675241

ABSTRACT

In mammals, repressive histone modifications such as trimethylation of histone H3 Lys9 (H3K9me3), frequently coexist with DNA methylation, producing a more stable and silenced chromatin state. However, it remains elusive how these epigenetic modifications crosstalk. Here, through structural and biochemical characterizations, we identified the replication foci targeting sequence (RFTS) domain of maintenance DNA methyltransferase DNMT1, a module known to bind the ubiquitylated H3 (H3Ub), as a specific reader for H3K9me3/H3Ub, with the recognition mode distinct from the typical trimethyl-lysine reader. Disruption of the interaction between RFTS and the H3K9me3Ub affects the localization of DNMT1 in stem cells and profoundly impairs the global DNA methylation and genomic stability. Together, this study reveals a previously unappreciated pathway through which H3K9me3 directly reinforces DNMT1-mediated maintenance DNA methylation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , Heterochromatin/metabolism , Histones/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Heterochromatin/genetics , Histones/chemistry , Histones/genetics , Humans , Lysine/genetics , Lysine/metabolism , Methylation , Protein Processing, Post-Translational
9.
J Cell Biol ; 219(8)2020 08 03.
Article in English | MEDLINE | ID: mdl-32568386

ABSTRACT

The microtubule cytoskeleton serves as a dynamic structural framework for mitosis in eukaryotic cells. TANGLED1 (TAN1) is a microtubule-binding protein that localizes to the division site and mitotic microtubules and plays a critical role in division plane orientation in plants. Here, in vitro experiments demonstrate that TAN1 directly binds microtubules, mediating microtubule zippering or end-on microtubule interactions, depending on their contact angle. Maize tan1 mutant cells improperly position the preprophase band (PPB), which predicts the future division site. However, cell shape-based modeling indicates that PPB positioning defects are likely a consequence of abnormal cell shapes and not due to TAN1 absence. In telophase, colocalization of growing microtubules ends from the phragmoplast with TAN1 at the division site suggests that TAN1 interacts with microtubule tips end-on. Together, our results suggest that TAN1 contributes to microtubule organization to ensure proper division plane orientation.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Division , Microtubules/metabolism , Plant Proteins/metabolism , Plants, Genetically Modified/metabolism , Zea mays/metabolism , Binding Sites , Cell Cycle Proteins/genetics , Gene Expression Regulation, Plant , Microtubules/genetics , Plant Proteins/genetics , Plants, Genetically Modified/genetics , Protein Binding , Signal Transduction , Time Factors , Zea mays/genetics
10.
Nat Commun ; 9(1): 5087, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504830

ABSTRACT

Cellular protein-RNA complexes assemble on nascent transcripts, but methods to observe transcription and protein binding in real time and at physiological concentrations are not available. Here, we report a single-molecule approach based on zero-mode waveguides that simultaneously tracks transcription progress and the binding of ribosomal protein S15 to nascent RNA transcripts during early ribosome biogenesis. We observe stable binding of S15 to single RNAs immediately after transcription for the majority of the transcripts at 35 °C but for less than half at 20 °C. The remaining transcripts exhibit either rapid and transient binding or are unable to bind S15, likely due to RNA misfolding. Our work establishes the foundation for studying transcription and its coupled co-transcriptional processes, including RNA folding, ligand binding, and enzymatic activity such as in coupling of transcription to splicing, ribosome assembly or translation.


Subject(s)
RNA, Ribosomal/biosynthesis , RNA/biosynthesis , Ribosomal Proteins/metabolism , Nucleic Acid Conformation , Protein Binding , RNA/chemistry , RNA Folding , RNA Splicing/genetics , RNA, Ribosomal/metabolism , Ribosomal Proteins/genetics
11.
Nucleic Acids Res ; 45(19): 11043-11055, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-28977553

ABSTRACT

In prokaryotes, RNA polymerase and ribosomes can bind concurrently to the same RNA transcript, leading to the functional coupling of transcription and translation. The interactions between RNA polymerase and ribosomes are crucial for the coordination of transcription with translation. Here, we report that RNA polymerase directly binds ribosomes and isolated large and small ribosomal subunits. RNA polymerase and ribosomes form a one-to-one complex with a micromolar dissociation constant. The formation of the complex is modulated by the conformational and functional states of RNA polymerase and the ribosome. The binding interface on the large ribosomal subunit is buried by the small subunit during protein synthesis, whereas that on the small subunit remains solvent-accessible. The RNA polymerase binding site on the ribosome includes that of the isolated small ribosomal subunit. This direct interaction between RNA polymerase and ribosomes may contribute to the coupling of transcription to translation.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Escherichia coli Proteins/metabolism , Protein Biosynthesis , Ribosome Subunits/metabolism , Transcription, Genetic , DNA-Directed RNA Polymerases/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Kinetics , Models, Molecular , Protein Binding , Protein Domains , Ribosome Subunits/chemistry , Ribosome Subunits/genetics
12.
Q Rev Biophys ; 49: e11, 2016 Jan.
Article in English | MEDLINE | ID: mdl-27658712

ABSTRACT

Translation of proteins by the ribosome regulates gene expression, with recent results underscoring the importance of translational control. Misregulation of translation underlies many diseases, including cancer and many genetic diseases. Decades of biochemical and structural studies have delineated many of the mechanistic details in prokaryotic translation, and sketched the outlines of eukaryotic translation. However, translation may not proceed linearly through a single mechanistic pathway, but likely involves multiple pathways and branchpoints. The stochastic nature of biological processes would allow different pathways to occur during translation that are biased by the interaction of the ribosome with other translation factors, with many of the steps kinetically controlled. These multiple pathways and branchpoints are potential regulatory nexus, allowing gene expression to be tuned at the translational level. As research focus shifts toward eukaryotic translation, certain themes will be echoed from studies on prokaryotic translation. This review provides a general overview of the dynamic data related to prokaryotic and eukaryotic translation, in particular recent findings with single-molecule methods, complemented by biochemical, kinetic, and structural findings. We will underscore the importance of viewing the process through the viewpoints of regulation, translational control, and heterogeneous pathways.

13.
Mol Cell ; 62(1): 92-103, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27058789

ABSTRACT

The complexity of eukaryotic translation allows fine-tuned regulation of protein synthesis. Viruses use internal ribosome entry sites (IRESs) to minimize or, like the CrPV IRES, eliminate the need for initiation factors. Here, by exploiting the CrPV IRES, we observed the entire process of initiation and transition to elongation in real time. We directly tracked the CrPV IRES, 40S and 60S ribosomal subunits, and tRNA using single-molecule fluorescence spectroscopy and identified multiple parallel initiation pathways within the system. Our results distinguished two pathways of 80S:CrPV IRES complex assembly that produce elongation-competent complexes. Following 80S assembly, the requisite eEF2-mediated translocation results in an unstable intermediate that is captured by binding of the elongator tRNA. Whereas initiation can occur in the 0 and +1 frames, the arrival of the first tRNA defines the reading frame and strongly favors 0 frame initiation. Overall, even in the simplest system, an intricate reaction network regulates translation initiation.


Subject(s)
Dicistroviridae/genetics , Internal Ribosome Entry Sites , Protein Biosynthesis , RNA, Viral/genetics , Dicistroviridae/metabolism , RNA, Transfer/metabolism , RNA, Viral/metabolism , Ribosomal Proteins/metabolism
14.
Nat Struct Mol Biol ; 23(2): 110-5, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26751643

ABSTRACT

N(6)-methylation of adenosine (forming m(6)A) is the most abundant post-transcriptional modification within the coding region of mRNA, but its role during translation remains unknown. Here, we used bulk kinetic and single-molecule methods to probe the effect of m(6)A in mRNA decoding. Although m(6)A base-pairs with uridine during decoding, as shown by X-ray crystallographic analyses of Thermus thermophilus ribosomal complexes, our measurements in an Escherichia coli translation system revealed that m(6)A modification of mRNA acts as a barrier to tRNA accommodation and translation elongation. The interaction between an m(6)A-modified codon and cognate tRNA echoes the interaction between a near-cognate codon and tRNA, because delay in tRNA accommodation depends on the position and context of m(6)A within codons and on the accuracy level of translation. Overall, our results demonstrate that chemical modification of mRNA can change translational dynamics.


Subject(s)
Adenosine/analogs & derivatives , Escherichia coli/genetics , Protein Biosynthesis , RNA, Bacterial/genetics , RNA, Messenger/genetics , RNA, Transfer/genetics , Thermus thermophilus/genetics , Adenosine/analysis , Adenosine/genetics , Codon , Crystallography, X-Ray , Escherichia coli/chemistry , RNA, Bacterial/chemistry , RNA, Messenger/chemistry , RNA, Transfer/chemistry , Thermus thermophilus/chemistry
15.
Cell ; 163(5): 1267-1280, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26590426

ABSTRACT

Nearly half of the ribosomes translating a particular bacteriophage T4 mRNA bypass a region of 50 nt, resuming translation 3' of this gap. How this large-scale, specific hop occurs and what determines whether a ribosome bypasses remain unclear. We apply single-molecule fluorescence with zero-mode waveguides to track individual Escherichia coli ribosomes during translation of T4's gene 60 mRNA. Ribosomes that bypass are characterized by a 10- to 20-fold longer pause in a non-canonical rotated state at the take-off codon. During the pause, mRNA secondary structure rearrangements are coupled to ribosome forward movement, facilitated by nascent peptide interactions that disengage the ribosome anticodon-codon interactions for slippage. Close to the landing site, the ribosome then scans mRNA in search of optimal base-pairing interactions. Our results provide a mechanistic and conformational framework for bypassing, highlighting a non-canonical ribosomal state to allow for mRNA structure refolding to drive large-scale ribosome movements.


Subject(s)
Escherichia coli/metabolism , Protein Biosynthesis , RNA, Messenger/chemistry , Ribosomes/metabolism , 5' Flanking Region , Anticodon , Base Sequence , Codon , Fluorescence Resonance Energy Transfer , Inverted Repeat Sequences , Molecular Sequence Data , Nucleic Acid Conformation , Peptide Elongation Factor G/metabolism , RNA, Messenger/metabolism , Ribosomes/chemistry
16.
Proc Natl Acad Sci U S A ; 112(2): 319-25, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25516984

ABSTRACT

Translation initiation can occur by multiple pathways. To delineate these pathways by single-molecule methods, fluorescently labeled ribosomal subunits are required. Here, we labeled human 40S ribosomal subunits with a fluorescent SNAP-tag at ribosomal protein eS25 (RPS25). The resulting ribosomal subunits could be specifically labeled in living cells and in vitro. Using single-molecule Förster resonance energy transfer (FRET) between RPS25 and domain II of the hepatitis C virus (HCV) internal ribosome entry site (IRES), we measured the rates of 40S subunit arrival to the HCV IRES. Our data support a single-step model of HCV IRES recruitment to 40S subunits, irreversible on the initiation time scale. We furthermore demonstrated that after binding, the 40S:HCV IRES complex is conformationally dynamic, undergoing slow large-scale rearrangements. Addition of translation extracts suppresses these fluctuations, funneling the complex into a single conformation on the 80S assembly pathway. These findings show that 40S:HCV IRES complex formation is accompanied by dynamic conformational rearrangements that may be modulated by initiation factors.


Subject(s)
Hepacivirus/genetics , Hepacivirus/metabolism , Ribosome Subunits, Small, Eukaryotic/metabolism , Amino Acid Sequence , Base Sequence , Binding Sites/genetics , CRISPR-Cas Systems , Fluorescence Resonance Energy Transfer , Gene Knockout Techniques , HeLa Cells , Hepacivirus/pathogenicity , Host-Pathogen Interactions , Humans , Kinetics , Molecular Sequence Data , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Peptide Chain Initiation, Translational , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism
17.
Nature ; 512(7514): 328-32, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-24919156

ABSTRACT

Spontaneous changes in the reading frame of translation are rare (frequency of 10(-3) to 10(-4) per codon), but can be induced by specific features in the messenger RNA (mRNA). In the presence of mRNA secondary structures, a heptanucleotide 'slippery sequence' usually defined by the motif X XXY YYZ, and (in some prokaryotic cases) mRNA sequences that base pair with the 3' end of the 16S ribosomal rRNA (internal Shine-Dalgarno sequences), there is an increased probability that a specific programmed change of frame occurs, wherein the ribosome shifts one nucleotide backwards into an overlapping reading frame (-1 frame) and continues by translating a new sequence of amino acids. Despite extensive biochemical and genetic studies, there is no clear mechanistic description for frameshifting. Here we apply single-molecule fluorescence to track the compositional and conformational dynamics of individual ribosomes at each codon during translation of a frameshift-inducing mRNA from the dnaX gene in Escherichia coli. Ribosomes that frameshift into the -1 frame are characterized by a tenfold longer pause in elongation compared to non-frameshifted ribosomes, which translate through unperturbed. During the pause, interactions of the ribosome with the mRNA stimulatory elements uncouple EF-G catalysed translocation from normal ribosomal subunit reverse-rotation, leaving the ribosome in a non-canonical intersubunit rotated state with an exposed codon in the aminoacyl-tRNA site (A site). tRNA(Lys) sampling and accommodation to the empty A site and EF-G action either leads to the slippage of the tRNAs into the -1 frame or maintains the ribosome into the 0 frame. Our results provide a general mechanistic and conformational framework for -1 frameshifting, highlighting multiple kinetic branchpoints during elongation.


Subject(s)
Frameshifting, Ribosomal , Peptide Chain Elongation, Translational , Ribosomes/metabolism , Bacterial Proteins/genetics , Codon/genetics , DNA Polymerase III/genetics , Escherichia coli , Kinetics , Peptide Elongation Factor G/metabolism , RNA, Messenger/genetics , RNA, Transfer, Amino Acyl/metabolism , Reading Frames/genetics , Ribosome Subunits/chemistry , Ribosome Subunits/metabolism , Ribosomes/chemistry , Rotation , Time Factors
18.
Biochemistry ; 53(14): 2215-7, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24654570

ABSTRACT

Bacimethrin-derived 2'-methoxythiamin pyrophosphate inhibits microbial growth by disrupting metabolic pathways dependent on thiamin-utilizing enzymes. This study describes the discovery of the bacimethrin biosynthetic gene cluster of Clostridium botulinum A ATCC 19397 and in vitro reconstitution of bacimethrin biosynthesis from cytidine 5'-monophosphate.


Subject(s)
Clostridium botulinum/metabolism , Thiamine/antagonists & inhibitors , Clostridium botulinum/genetics , Multigene Family
19.
Proc Natl Acad Sci U S A ; 111(2): 664-9, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24379388

ABSTRACT

Zero-mode waveguides provide a powerful technology for studying single-molecule real-time dynamics of biological systems at physiological ligand concentrations. We customized a commercial zero-mode waveguide-based DNA sequencer for use as a versatile instrument for single-molecule fluorescence detection and showed that the system provides long fluorophore lifetimes with good signal to noise and low spectral cross-talk. We then used a ribosomal translation assay to show real-time fluidic delivery during data acquisition, showing it is possible to follow the conformation and composition of thousands of single biomolecules simultaneously through four spectral channels. This instrument allows high-throughput multiplexed dynamics of single-molecule biological processes over long timescales. The instrumentation presented here has broad applications to single-molecule studies of biological systems and is easily accessible to the biophysical community.


Subject(s)
Biophysics/methods , Fluorescence , High-Throughput Screening Assays/methods , Monitoring, Physiologic/methods , Software , Algorithms , Biophysics/instrumentation , Computer Systems , High-Throughput Screening Assays/instrumentation , Monitoring, Physiologic/instrumentation
20.
Structure ; 21(12): 2197-207, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24183571

ABSTRACT

Recognition of the mRNA 5' m7G(5')ppp(5')N cap is key to translation initiation for most eukaryotic mRNAs. The cap is bound by the eIF4F complex, consisting of a cap-binding protein (eIF4E), a "scaffold" protein (eIF4G), and an RNA helicase (eIF4A). As a central early step in initiation, regulation of eIF4F is crucial for cellular viability. Although the structure and function of eIF4E have been defined, a dynamic mechanistic picture of its activity at the molecular level in the eIF4F·mRNA complex is still unavailable. Here, using single-molecule fluorescence, we measured the effects of Saccharomyces cerevisiae eIF4F factors, mRNA secondary structure, and the poly(A)-binding protein Pab1p on eIF4E-mRNA binding dynamics. Our data provide an integrated picture of how eIF4G and mRNA structure modulate eIF4E-mRNA interaction, and uncover an eIF4G- and poly(A)-independent activity of poly(A)-binding protein that prolongs the eIF4E·mRNA complex lifetime.


Subject(s)
Eukaryotic Initiation Factor-4E/chemistry , RNA Caps/chemistry , RNA, Fungal/chemistry , Saccharomyces cerevisiae/metabolism , Carbocyanines , Eukaryotic Initiation Factor-4G/chemistry , Eukaryotic Initiation Factor-4G/metabolism , Exoribonucleases/chemistry , Fluorescence Resonance Energy Transfer , Fluorescent Dyes , Nucleic Acid Conformation , Peptide Chain Initiation, Translational , Poly A/chemistry , Poly(A)-Binding Proteins/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...