Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 626(8001): 1125-1132, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38355796

RESUMO

To conserve energy during starvation and stress, many organisms use hibernation factor proteins to inhibit protein synthesis and protect their ribosomes from damage1,2. In bacteria, two families of hibernation factors have been described, but the low conservation of these proteins and the huge diversity of species, habitats and environmental stressors have confounded their discovery3-6. Here, by combining cryogenic electron microscopy, genetics and biochemistry, we identify Balon, a new hibernation factor in the cold-adapted bacterium Psychrobacter urativorans. We show that Balon is a distant homologue of the archaeo-eukaryotic translation factor aeRF1 and is found in 20% of representative bacteria. During cold shock or stationary phase, Balon occupies the ribosomal A site in both vacant and actively translating ribosomes in complex with EF-Tu, highlighting an unexpected role for EF-Tu in the cellular stress response. Unlike typical A-site substrates, Balon binds to ribosomes in an mRNA-independent manner, initiating a new mode of ribosome hibernation that can commence while ribosomes are still engaged in protein synthesis. Our work suggests that Balon-EF-Tu-regulated ribosome hibernation is a ubiquitous bacterial stress-response mechanism, and we demonstrate that putative Balon homologues in Mycobacteria bind to ribosomes in a similar fashion. This finding calls for a revision of the current model of ribosome hibernation inferred from common model organisms and holds numerous implications for how we understand and study ribosome hibernation.


Assuntos
Proteínas de Bactérias , Resposta ao Choque Frio , Fatores de Terminação de Peptídeos , Biossíntese de Proteínas , Psychrobacter , Proteínas Ribossômicas , Ribossomos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/ultraestrutura , Fator Tu de Elongação de Peptídeos/química , Fator Tu de Elongação de Peptídeos/metabolismo , Fator Tu de Elongação de Peptídeos/ultraestrutura , Proteínas Ribossômicas/química , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Proteínas Ribossômicas/ultraestrutura , Ribossomos/química , Ribossomos/metabolismo , Ribossomos/ultraestrutura , Psychrobacter/química , Psychrobacter/genética , Psychrobacter/metabolismo , Psychrobacter/ultraestrutura , Microscopia Crioeletrônica , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/genética , Fatores de Terminação de Peptídeos/metabolismo , Fatores de Terminação de Peptídeos/ultraestrutura
2.
Nucleic Acids Res ; 49(16): 9539-9547, 2021 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34403461

RESUMO

In Escherichia coli, elevated levels of free l-tryptophan (l-Trp) promote translational arrest of the TnaC peptide by inhibiting its termination. However, the mechanism by which translation-termination by the UGA-specific decoding release factor 2 (RF2) is inhibited at the UGA stop codon of stalled TnaC-ribosome-nascent chain complexes has so far been ambiguous. This study presents cryo-EM structures for ribosomes stalled by TnaC in the absence and presence of RF2 at average resolutions of 2.9 and 3.5 Å, respectively. Stalled TnaC assumes a distinct conformation composed of two small α-helices that act together with residues in the peptide exit tunnel (PET) to coordinate a single L-Trp molecule. In addition, while the peptidyl-transferase center (PTC) is locked in a conformation that allows RF2 to adopt its canonical position in the ribosome, it prevents the conserved and catalytically essential GGQ motif of RF2 from adopting its active conformation in the PTC. This explains how translation of the TnaC peptide effectively allows the ribosome to function as a L-Trp-specific small-molecule sensor that regulates the tnaCAB operon.


Assuntos
Proteínas de Escherichia coli/ultraestrutura , Fatores de Terminação de Peptídeos/ultraestrutura , Biossíntese de Proteínas , Ribossomos/ultraestrutura , Códon de Terminação/genética , Microscopia Crioeletrônica , Escherichia coli/genética , Escherichia coli/ultraestrutura , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/genética , Conformação Proteica , Conformação Proteica em alfa-Hélice , Ribossomos/genética , Triptofano/genética
3.
Biomolecules ; 10(10)2020 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-32987720

RESUMO

The precise kinetic pathways of peptide clustering and fibril formation are not fully understood. Here we study the initial clustering kinetics and transient cluster morphologies during aggregation of the heptapeptide fragment GNNQQNY from the yeast prion protein Sup35. We use a mid-resolution coarse-grained molecular dynamics model of Bereau and Deserno to explore the aggregation pathways from the initial random distribution of free monomers to the formation of large clusters. By increasing the system size to 72 peptides we could follow directly the molecular events leading to the formation of stable fibril-like structures. To quantify those structures we developed a new cluster helicity parameter. We found that the formation of fibril-like structures is a cooperative processes that requires a critical number of monomers, M⋆≈25, in a cluster. The terminal tyrosine residue is the structural determinant in the formation of helical fibril-like structures. This work supports and quantifies the two-step aggregation model where the initially formed amorphous clusters grow and, when they are large enough, rearrange into mature twisted structures. However, in addition to the nucleated fibrillation, growing aggregates undergo further internal reorganization, which leads to more compact structures of large aggregates.


Assuntos
Amiloide/ultraestrutura , Fatores de Terminação de Peptídeos/ultraestrutura , Peptídeos/química , Proteínas Priônicas/ultraestrutura , Proteínas de Saccharomyces cerevisiae/ultraestrutura , Amiloide/genética , Humanos , Cinética , Simulação de Dinâmica Molecular , Fatores de Terminação de Peptídeos/genética , Peptídeos/genética , Proteínas Priônicas/genética , Agregados Proteicos/genética , Agregação Patológica de Proteínas/genética , Conformação Proteica , Conformação Proteica em alfa-Hélice/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/ultraestrutura , Proteínas de Saccharomyces cerevisiae/genética
4.
Proc Natl Acad Sci U S A ; 117(14): 7831-7836, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32213585

RESUMO

The yeast prion protein Sup35, which contains intrinsically disordered regions, forms amyloid fibrils responsible for a prion phenotype [PSI+]. Using high-speed atomic force microscopy (HS-AFM), we directly visualized the prion determinant domain (Sup35NM) and the formation of its oligomers and fibrils at subsecond and submolecular resolutions. Monomers with freely moving tail-like regions initially appeared in the images, and subsequently oligomers with distinct sizes of ∼1.7 and 3 to 4 nm progressively accumulated. Nevertheless, these oligomers did not form fibrils, even after an incubation for 2 h in the presence of monomers. Fibrils appeared after much longer monomer incubation. The fibril elongation occurred smoothly without discrete steps, suggesting gradual conversions of the incorporated monomers into cross-ß structures. The individual oligomers were separated from each other and also from the fibrils by respective, identical lengths on the mica surface, probably due to repulsion caused by the freely moving disordered regions. Based on these HS-AFM observations, we propose that the freely moving tails of the monomers are incorporated into the fibril ends, and then the structural conversions to cross-ß structures gradually occur.


Assuntos
Amiloide/ultraestrutura , Microscopia de Força Atômica , Fatores de Terminação de Peptídeos/ultraestrutura , Proteínas Priônicas/ultraestrutura , Proteínas de Saccharomyces cerevisiae/ultraestrutura , Saccharomyces cerevisiae/ultraestrutura , Amiloide/genética , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/genética , Proteínas Priônicas/genética , Conformação Proteica em Folha beta/genética , Domínios Proteicos/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética
5.
Nat Commun ; 10(1): 2579, 2019 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-31189921

RESUMO

When the ribosome encounters a stop codon, it recruits a release factor (RF) to hydrolyze the ester bond between the peptide chain and tRNA. RFs have structural motifs that recognize stop codons in the decoding center and a GGQ motif for induction of hydrolysis in the peptidyl transfer center 70 Å away. Surprisingly, free RF2 is compact, with only 20 Å between its codon-reading and GGQ motifs. Cryo-EM showed that ribosome-bound RFs have extended structures, suggesting that RFs are compact when entering the ribosome and then extend their structures upon stop codon recognition. Here we use time-resolved cryo-EM to visualize transient compact forms of RF1 and RF2 at 3.5 and 4 Å resolution, respectively, in the codon-recognizing ribosome complex on the native pathway. About 25% of complexes have RFs in the compact state at 24 ms reaction time, and within 60 ms virtually all ribosome-bound RFs are transformed to their extended forms.


Assuntos
Proteínas de Escherichia coli/ultraestrutura , Modelos Moleculares , Terminação Traducional da Cadeia Peptídica/fisiologia , Fatores de Terminação de Peptídeos/ultraestrutura , Domínios Proteicos/fisiologia , Sítios de Ligação/fisiologia , Códon de Terminação/metabolismo , Microscopia Crioeletrônica , Proteínas de Escherichia coli/metabolismo , Fatores de Terminação de Peptídeos/metabolismo , RNA de Transferência/metabolismo , Subunidades Ribossômicas Maiores de Bactérias/metabolismo , Subunidades Ribossômicas Menores de Bactérias/metabolismo , Fatores de Tempo
6.
J Struct Biol ; 201(1): 5-14, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29078994

RESUMO

Prion and some other incurable human neurodegenerative diseases are associated with misfolding of specific proteins, followed by the formation of amyloids. Despite the widespread usage of the transmission electron and of the atomic force microscopy for studing such amyloids, many related methodological issues still have not been studied until now. Here, we consider one of the first amyloids found in Saccharomyces cerevisiae yeast, i.e. Sup35NMp, to study the adsorption of monomeric protein and its fibrils on the surface of mica, silica, gold and on formvar film. Comparison of linear characteristics of these units calculated by processing of images obtained by the atomic force, transmission and scanning electron microscopy was carried out. The minimal number of measurements of fibril diameters to obtain the values in a given confidence interval were determined. We investigated the film formed by monomeric protein on mica surface, which veiled some morphology features of fibrils. Besides, we revealed that parts of the Sup35NMp excluded from the fibril core can form a wide "coat". The length of the protein forming the core of the fibrils was estimated.


Assuntos
Amiloide/química , Fatores de Terminação de Peptídeos/química , Príons/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/química , Adsorção , Silicatos de Alumínio/química , Amiloide/ultraestrutura , Ouro/química , Microscopia de Força Atômica/métodos , Microscopia Eletrônica de Varredura/métodos , Microscopia Eletrônica de Transmissão/métodos , Fatores de Terminação de Peptídeos/ultraestrutura , Príons/ultraestrutura , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/ultraestrutura , Proteínas de Saccharomyces cerevisiae/ultraestrutura , Dióxido de Silício/química , Propriedades de Superfície
7.
Nat Struct Mol Biol ; 24(9): 752-757, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28741611

RESUMO

Many antibiotics stop bacterial growth by inhibiting different steps of protein synthesis. However, no specific inhibitors of translation termination are known. Proline-rich antimicrobial peptides, a component of the antibacterial defense system of multicellular organisms, interfere with bacterial growth by inhibiting translation. Here we show that Api137, a derivative of the insect-produced antimicrobial peptide apidaecin, arrests terminating ribosomes using a unique mechanism of action. Api137 binds to the Escherichia coli ribosome and traps release factor (RF) RF1 or RF2 subsequent to the release of the nascent polypeptide chain. A high-resolution cryo-EM structure of the ribosome complexed with RF1 and Api137 reveals the molecular interactions that lead to RF trapping. Api137-mediated depletion of the cellular pool of free release factors causes the majority of ribosomes to stall at stop codons before polypeptide release, thereby resulting in a global shutdown of translation termination.


Assuntos
Anti-Infecciosos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Proteínas de Escherichia coli/metabolismo , Fatores de Terminação de Peptídeos/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Ribossomos/efeitos dos fármacos , Microscopia Crioeletrônica , Escherichia coli/efeitos dos fármacos , Proteínas de Escherichia coli/ultraestrutura , Modelos Biológicos , Modelos Moleculares , Fatores de Terminação de Peptídeos/ultraestrutura , Ribossomos/ultraestrutura
8.
Elife ; 62017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28300532

RESUMO

ArfA rescues ribosomes stalled on truncated mRNAs by recruiting release factor RF2, which normally binds stop codons to catalyze peptide release. We report two 3.2 Šresolution cryo-EM structures - determined from a single sample - of the 70S ribosome with ArfA•RF2 in the A site. In both states, the ArfA C-terminus occupies the mRNA tunnel downstream of the A site. One state contains a compact inactive RF2 conformation. Ordering of the ArfA N-terminus in the second state rearranges RF2 into an extended conformation that docks the catalytic GGQ motif into the peptidyl-transferase center. Our work thus reveals the structural dynamics of ribosome rescue. The structures demonstrate how ArfA 'senses' the vacant mRNA tunnel and activates RF2 to mediate peptide release without a stop codon, allowing stalled ribosomes to be recycled.


Assuntos
Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/ultraestrutura , Fatores de Terminação de Peptídeos/metabolismo , Fatores de Terminação de Peptídeos/ultraestrutura , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/ultraestrutura , Ribossomos/metabolismo , Ribossomos/ultraestrutura , Microscopia Crioeletrônica , Ligação Proteica
9.
Nature ; 541(7638): 554-557, 2017 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-28077875

RESUMO

Quality control mechanisms intervene appropriately when defective translation events occur, in order to preserve the integrity of protein synthesis. Rescue of ribosomes translating on messenger RNAs that lack stop codons is one of the co-translational quality control pathways. In many bacteria, ArfA recognizes stalled ribosomes and recruits the release factor RF2, which catalyses the termination of protein synthesis. Although an induced-fit mechanism of nonstop mRNA surveillance mediated by ArfA and RF2 has been reported, the molecular interaction between ArfA and RF2 in the ribosome that is responsible for the mechanism is unknown. Here we report an electron cryo-microscopy structure of ArfA and RF2 in complex with the 70S ribosome bound to a nonstop mRNA. The structure, which is consistent with our kinetic and biochemical data, reveals the molecular interactions that enable ArfA to specifically recruit RF2, not RF1, into the ribosome and to enable RF2 to release the truncated protein product in this co-translational quality control pathway. The positively charged C-terminal domain of ArfA anchors in the mRNA entry channel of the ribosome. Furthermore, binding of ArfA and RF2 induces conformational changes in the ribosomal decoding centre that are similar to those seen in other protein-involved decoding processes. Specific interactions between residues in the N-terminal domain of ArfA and RF2 help RF2 to adopt a catalytically competent conformation for peptide release. Our findings provide a framework for understanding recognition of the translational state of the ribosome by new proteins, and expand our knowledge of the decoding potential of the ribosome.


Assuntos
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Terminação Traducional da Cadeia Peptídica , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Ribossomos/metabolismo , Biocatálise , Códon de Terminação , Microscopia Crioeletrônica , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/ultraestrutura , Proteínas de Escherichia coli/ultraestrutura , Modelos Moleculares , Fatores de Terminação de Peptídeos/ultraestrutura , Ligação Proteica , Domínios Proteicos , RNA Mensageiro/química , RNA Mensageiro/genética , Proteínas de Ligação a RNA/ultraestrutura , Subunidades Ribossômicas Menores de Bactérias/química , Subunidades Ribossômicas Menores de Bactérias/metabolismo , Subunidades Ribossômicas Menores de Bactérias/ultraestrutura , Ribossomos/química , Ribossomos/ultraestrutura
10.
Nature ; 541(7638): 550-553, 2017 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-27906160

RESUMO

During cellular translation of messenger RNAs by ribosomes, the translation apparatus sometimes pauses or stalls at the elongation and termination steps. With the exception of programmed stalling, which is usually used by cells for regulatory purposes, ribosomes stalled on mRNAs need to be terminated and recycled to maintain adequate translation capacity. Much ribosome stalling originates in aberrant mRNAs that lack a stop codon. Transcriptional errors, misprocessing of primary transcripts, and undesired mRNA cleavage all contribute to the formation of non-stop mRNAs. Ribosomes stalled at the 3' end of non-stop mRNAs do not undergo normal termination owing to the lack of specific stop-codon recognition by canonical peptide release factors at the A-site decoding centre. In bacteria, the transfer-messenger RNA (tmRNA)-SmpB-mediated trans-translation rescue system reroutes stalled ribosomes to the normal elongation cycle and translation termination. Two additional rescue systems, ArfA-RF2 (refs 13, 14, 15, 16) and ArfB (formerly known as YaeJ), are also present in many bacterial species, but their mechanisms are not fully understood. Here, using cryo-electron microscopy, we characterize the structure of the Escherichia coli 70S ribosome bound with ArfA, the release factor RF2, a short non-stop mRNA and a cognate P-site tRNA. The C-terminal loop of ArfA occupies the mRNA entry channel on the 30S subunit, whereas its N terminus is sandwiched between the decoding centre and the switch loop of RF2, leading to marked conformational changes in both the decoding centre and RF2. Despite the distinct conformation of RF2, its conserved catalytic GGQ motif is precisely positioned next to the CCA-end of the P-site tRNA. These data illustrate a stop-codon surrogate mechanism for ArfA in facilitating the termination of non-stop ribosomal complexes by RF2.


Assuntos
Microscopia Crioeletrônica , Proteínas de Escherichia coli/metabolismo , Terminação Traducional da Cadeia Peptídica , Fatores de Terminação de Peptídeos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribossomos/metabolismo , Códon de Terminação , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/ultraestrutura , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/ultraestrutura , Modelos Moleculares , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/ultraestrutura , Ligação Proteica , Conformação Proteica , RNA Mensageiro/química , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/ultraestrutura , Subunidades Ribossômicas Menores de Bactérias/química , Subunidades Ribossômicas Menores de Bactérias/metabolismo , Subunidades Ribossômicas Menores de Bactérias/ultraestrutura , Ribossomos/química , Ribossomos/ultraestrutura
11.
Nature ; 541(7638): 546-549, 2017 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-27906161

RESUMO

In bacteria, ribosomes stalled on truncated mRNAs that lack a stop codon are rescued by the transfer-messenger RNA (tmRNA), alternative rescue factor A (ArfA) or ArfB systems. Although tmRNA-ribosome and ArfB-ribosome structures have been determined, how ArfA recognizes the presence of truncated mRNAs and recruits the canonical termination release factor RF2 to rescue the stalled ribosomes is unclear. Here we present a cryo-electron microscopy reconstruction of the Escherichia coli 70S ribosome stalled on a truncated mRNA in the presence of ArfA and RF2. The structure shows that the C terminus of ArfA binds within the mRNA entry channel on the small ribosomal subunit, and explains how ArfA distinguishes between ribosomes that bear truncated or full-length mRNAs. The N terminus of ArfA establishes several interactions with the decoding domain of RF2, and this finding illustrates how ArfA recruits RF2 to the stalled ribosome. Furthermore, ArfA is shown to stabilize a unique conformation of the switch loop of RF2, which mimics the canonical translation termination state by directing the catalytically important GGQ motif within domain 3 of RF2 towards the peptidyl-transferase centre of the ribosome. Thus, our structure reveals not only how ArfA recruits RF2 to the ribosome but also how it promotes an active conformation of RF2 to enable translation termination in the absence of a stop codon.


Assuntos
Códon de Terminação , Microscopia Crioeletrônica , Proteínas de Escherichia coli/química , Terminação Traducional da Cadeia Peptídica , Fatores de Terminação de Peptídeos/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/química , Ribossomos/metabolismo , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/ultraestrutura , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/ultraestrutura , Modelos Moleculares , Fatores de Terminação de Peptídeos/metabolismo , Fatores de Terminação de Peptídeos/ultraestrutura , Ligação Proteica , Conformação Proteica , Estabilidade Proteica , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/ultraestrutura , Ribossomos/química , Ribossomos/ultraestrutura
12.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 4): 882-95, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25849399

RESUMO

Structural analysis of protein fibrillation is inherently challenging. Given the crucial role of fibrils in amyloid diseases, method advancement is urgently needed. A hybrid modelling approach is presented enabling detailed analysis of a highly ordered and hierarchically organized fibril of the GNNQQNY peptide fragment of a yeast prion protein. Data from small-angle X-ray solution scattering, fibre diffraction and electron microscopy are combined with existing high-resolution X-ray crystallographic structures to investigate the fibrillation process and the hierarchical fibril structure of the peptide fragment. The elongation of these fibrils proceeds without the accumulation of any detectable amount of intermediate oligomeric species, as is otherwise reported for, for example, glucagon, insulin and α-synuclein. Ribbons constituted of linearly arranged protofilaments are formed. An additional hierarchical layer is generated via the pairing of ribbons during fibril maturation. Based on the complementary data, a quasi-atomic resolution model of the protofilament peptide arrangement is suggested. The peptide structure appears in a ß-sheet arrangement reminiscent of the ß-zipper structures evident from high-resolution crystal structures, with specific differences in the relative peptide orientation. The complexity of protein fibrillation and structure emphasizes the need to use multiple complementary methods.


Assuntos
Amiloide/química , Fatores de Terminação de Peptídeos/química , Príons/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/química , Sequência de Aminoácidos , Amiloide/ultraestrutura , Microscopia Eletrônica , Modelos Moleculares , Fatores de Terminação de Peptídeos/ultraestrutura , Príons/ultraestrutura , Estrutura Secundária de Proteína , Proteínas de Saccharomyces cerevisiae/ultraestrutura , Espalhamento a Baixo Ângulo , Difração de Raios X
13.
Biochim Biophys Acta ; 1854(3): 218-28, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25542374

RESUMO

Assembly of amyloid proteins into aggregates requires the ordering of the monomers in oligomers and especially in such highly organized structures as fibrils. This ordering is accompanied by structural transitions leading to the formation of ordered ß-structural motifs in proteins and peptides lacking secondary structures. To characterize the effect of the monomer arrangements on the aggregation process at various stages, we performed comparative studies of the yeast prion protein Sup35 heptapeptide (GNNQQNY) along with its dimeric form CGNNQQNY-(d-Pro)-G-GNNQQNY. The (d-Pro)-G linker in this construct is capable of adopting a ß-turn, facilitating the assembly of the dimer into the dimeric antiparallel hairpin structure (AP-hairpin). We applied Atomic Force Microscopy (AFM) techniques to follow peptide-peptide interactions at the single molecule level, to visualize the morphology of aggregates formed by both constructs, thioflavin T (ThT) fluorescence to follow the aggregation kinetics, and circular dichroism (CD) spectroscopy to characterize the secondary structure of the constructs. The ThT fluorescence data showed that the AP-hairpin aggregation kinetics is insensitive to the external environment such as ionic strength and pH contrary to the monomers the kinetics of which depends dramatically on the ionic strength and pH. The AFM topographic imaging revealed that AP-hairpins primarily assemble into globular aggregates, whereas linear fibrils are primary assemblies of the monomers suggesting that both constructs follow different aggregation pathways during the self-assembly. These morphological differences are in line with the AFM force spectroscopy experiments and CD spectroscopy measurements, suggesting that the AP-hairpin is structurally rigid regardless of changes of environmental factors.


Assuntos
Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/ultraestrutura , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/ultraestrutura , Sítios de Ligação , Cristalização/métodos , Dimerização , Ligação Proteica , Conformação Proteica
15.
Elife ; 2: e00411, 2013 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-23755360

RESUMO

Termination of messenger RNA translation in Bacteria and Archaea is initiated by release factors (RFs) 1 or 2 recognizing a stop codon in the ribosomal A site and releasing the peptide from the P-site transfer RNA. After release, RF-dissociation is facilitated by the G-protein RF3. Structures of ribosomal complexes with RF1 or RF2 alone or with RF3 alone-RF3 bound to a non-hydrolyzable GTP-analog-have been reported. Here, we present the cryo-EM structure of a post-termination ribosome containing both apo-RF3 and RF1. The conformation of RF3 is distinct from those of free RF3•GDP and ribosome-bound RF3•GDP(C/N)P. Furthermore, the conformation of RF1 differs from those observed in RF3-lacking ribosomal complexes. Our study provides structural keys to the mechanism of guanine nucleotide exchange on RF3 and to an L12-mediated ribosomal recruitment of RF3. In conjunction with previous observations, our data provide the foundation to structurally characterize the complete action cycle of the G-protein RF3. DOI:http://dx.doi.org/10.7554/eLife.00411.001.


Assuntos
Microscopia Crioeletrônica , Proteínas de Escherichia coli/metabolismo , Fatores de Terminação de Peptídeos/metabolismo , RNA Bacteriano/biossíntese , RNA Mensageiro/biossíntese , Ribossomos/metabolismo , Terminação da Transcrição Genética , Sítios de Ligação , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/ultraestrutura , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Simulação de Dinâmica Molecular , Mutação , Fatores de Terminação de Peptídeos/genética , Fatores de Terminação de Peptídeos/ultraestrutura , Ligação Proteica , Conformação Proteica , Proteínas Ribossômicas/metabolismo , Ribossomos/ultraestrutura , Fatores de Tempo
16.
Proc Natl Acad Sci U S A ; 109(45): 18413-8, 2012 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-23091004

RESUMO

Eukaryotic translation termination results from the complex functional interplay between two eukaryotic release factors, eRF1 and eRF3, and the ribosome, in which GTP hydrolysis by eRF3 couples codon recognition with peptidyl-tRNA hydrolysis by eRF1. Here, using cryo-electron microscopy (cryo-EM) and flexible fitting, we determined the structure of eRF1-eRF3-guanosine 5'-[ß,γ-imido]triphosphate (GMPPNP)-bound ribosomal pretermination complex (pre-TC), which corresponds to the initial, pre-GTP hydrolysis stage of factor attachment. Our results show that eukaryotic translation termination involves a network of interactions between the two release factors and the ribosome. Our structure provides mechanistic insight into the coordination between GTP hydrolysis by eRF3 and subsequent peptide release by eRF1.


Assuntos
Microscopia Crioeletrônica , Mamíferos/metabolismo , Terminação Traducional da Cadeia Peptídica , Fatores de Terminação de Peptídeos/ultraestrutura , Animais , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Humanos , Modelos Moleculares , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica , Aminoacil-RNA de Transferência/química , Aminoacil-RNA de Transferência/metabolismo , Coelhos , Ribossomos/metabolismo , Ribossomos/ultraestrutura , Saccharomyces cerevisiae
17.
Trends Biochem Sci ; 36(5): 282-92, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21420300

RESUMO

When the ribosome machinery reaches a stop codon in the mRNA, protein synthesis stops, and nascent polypeptide release is catalysed by class-I release factors (RFs); class-II RFs then promote the release of class-I RFs. Cryo electron microscopy structures of termination complexes and crystal structures of isolated factors have provided insights into key concepts such as bridging of active sites on the ribosome, and conformational changes that regulate the termination process. Recent crystal structures of the four possible functional ribosome complexes that contain the class-I RFs and the three stop codons have uncovered the molecular mechanisms by which RF1/RF2 (i) both recognise UAA, but discriminate specifically between UAG and UGA, and (ii) catalyse peptide release. Moreover, ongoing research also promises to reveal the structure-function relations of class-II RFs.


Assuntos
Códon de Terminação/genética , Fatores de Terminação de Peptídeos/metabolismo , Biossíntese de Proteínas/genética , Ribossomos/metabolismo , Animais , Sequência de Bases , Catálise , Microscopia Crioeletrônica , Humanos , Modelos Genéticos , Fatores de Terminação de Peptídeos/ultraestrutura , Ribossomos/genética , Ribossomos/ultraestrutura
18.
Mol Microbiol ; 79(2): 523-32, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21219467

RESUMO

In yeast cells infected with the [PSI+] prion, Sup35p forms aggregates and its activity in translation termination is downregulated. Transfection experiments have shown that Sup35p filaments assembled in vitro are infectious, suggesting that they reproduce or closely resemble the prion. We have used several EM techniques to study the molecular architecture of filaments, seeking clues as to the mechanism of downregulation. Sup35p has an N-terminal 'prion' domain; a highly charged middle (M-)domain; and a C-terminal domain with the translation termination activity. By negative staining, cryo-EM and scanning transmission EM (STEM), filaments of full-length Sup35p show a thin backbone fibril surrounded by a diffuse 65-nm-wide cloud of globular C-domains. In diameter (∼8 nm) and appearance, the backbones resemble amyloid fibrils of N-domains alone. STEM mass-per-unit-length data yield ∼1 subunit per 0.47 nm for N-fibrils, NM-filaments and Sup35p filaments, further supporting the fibril backbone model. The 30 nm radial span of decorating C-domains indicates that the M-domains assume highly extended conformations, offering an explanation for the residual Sup35p activity in infected cells, whereby the C-domains remain free enough to interact with ribosomes.


Assuntos
Amiloide/química , Amiloide/metabolismo , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/metabolismo , Príons/química , Príons/metabolismo , Multimerização Proteica , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Amiloide/ultraestrutura , Microscopia Eletrônica , Fatores de Terminação de Peptídeos/ultraestrutura , Príons/ultraestrutura , Estrutura Terciária de Proteína , Proteínas de Saccharomyces cerevisiae/ultraestrutura
19.
Nat Struct Mol Biol ; 17(12): 1422-30, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21113168

RESUMO

Amyloid fibrils are important in diverse cellular functions, feature in many human diseases and have potential applications in nanotechnology. Here we describe methods that combine optical trapping and fluorescent imaging to characterize the forces that govern the integrity of amyloid fibrils formed by a yeast prion protein. A crucial advance was to use the self-templating properties of amyloidogenic proteins to tether prion fibrils, enabling their manipulation in the optical trap. At normal pulling forces the fibrils were impervious to disruption. At much higher forces (up to 250 pN), discontinuities occurred in force-extension traces before fibril rupture. Experiments with selective amyloid-disrupting agents and mutations demonstrated that such discontinuities were caused by the unfolding of individual subdomains. Thus, our results reveal unusually strong noncovalent intermolecular contacts that maintain fibril integrity even when individual monomers partially unfold and extend fibril length.


Assuntos
Amiloide/fisiologia , Pinças Ópticas , Fatores de Terminação de Peptídeos/fisiologia , Príons/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Amiloide/ultraestrutura , Fenômenos Biomecânicos , Microscopia de Fluorescência , Fatores de Terminação de Peptídeos/metabolismo , Fatores de Terminação de Peptídeos/ultraestrutura , Príons/metabolismo , Príons/ultraestrutura , Desdobramento de Proteína , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/ultraestrutura , Estresse Mecânico
20.
Biosens Bioelectron ; 26(4): 1137-41, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20970983

RESUMO

In order to obtain an ultra-sensitive molecular biosensor, we designed an auto-biotinylated bifunctional protein nanowire (bFPNw) based on the self-assembly of a yeast amyloid protein, Sup35, to which protein G and a biotin acceptor peptide (BAP) were genetically fused. These auto-biotinylated bFPNws can transfer hundreds of commercially available diagnostic enzymes to an antigen-antibody complex via the biotin-avidin system, greatly enhancing the sensitivity of immune-biosensing. Compared to our previously reported seeding-induced bFPNws (Men et al., 2009), these auto-biotinylated bFPNws gave greater signal amplification, reduced non-specific binding and improved stability. The auto-biotinylated self-assembled bFPNw molecular biosensors were applied to detect Yersinia pestis (Y. pestis) F1 antigen and showed a 2000- to 4000-fold increase in sensitivity compared to traditional immunoassays, demonstrating the potential use of these self-assembling protein nanowires in biosensing.


Assuntos
Técnicas Biossensoriais/instrumentação , Imunoensaio/instrumentação , Nanofios/química , Antígenos de Bactérias/análise , Distinções e Prêmios , Proteínas de Bactérias/análise , Proteínas de Bactérias/química , Técnicas Biossensoriais/métodos , Técnicas Biossensoriais/estatística & dados numéricos , Biotinilação , Reagentes de Ligações Cruzadas/química , Imunoensaio/métodos , Imunoensaio/estatística & dados numéricos , Nanofios/ultraestrutura , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/ultraestrutura , Multimerização Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/ultraestrutura , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/ultraestrutura , Yersinia pestis/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...