Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Life Sci Alliance ; 7(10)2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39103227

RESUMO

The complex multistep activation cascade of Ire1 involves changes in the Ire1 conformation and oligomeric state. Ire1 activation enhances ER folding capacity, in part by overexpressing the ER Hsp70 molecular chaperone BiP; in turn, BiP provides tight negative control of Ire1 activation. This study demonstrates that BiP regulates Ire1 activation through a direct interaction with Ire1 oligomers. Particularly, we demonstrated that the binding of Ire1 luminal domain (LD) to unfolded protein substrates not only trigger conformational changes in Ire1-LD that favour the formation of Ire1-LD oligomers but also exposes BiP binding motifs, enabling the molecular chaperone BiP to directly bind to Ire1-LD in an ATP-dependent manner. These transient interactions between BiP and two short motifs in the disordered region of Ire1-LD are reminiscent of interactions between clathrin and another Hsp70, cytoplasmic Hsc70. BiP binding to substrate-bound Ire1-LD oligomers enables unfolded protein substrates and BiP to synergistically and dynamically control Ire1-LD oligomerisation, helping to return Ire1 to its deactivated state when an ER stress response is no longer required.


Assuntos
Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático , Endorribonucleases , Proteínas de Choque Térmico , Ligação Proteica , Proteínas Serina-Treonina Quinases , Proteínas Serina-Treonina Quinases/metabolismo , Chaperona BiP do Retículo Endoplasmático/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/química , Endorribonucleases/metabolismo , Endorribonucleases/química , Humanos , Retículo Endoplasmático/metabolismo , Resposta a Proteínas não Dobradas , Multimerização Proteica , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP70/química , Dobramento de Proteína , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/química , Domínios Proteicos
2.
J Am Chem Soc ; 146(31): 21525-21534, 2024 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-39047145

RESUMO

Various approaches have been developed to target RNA and modulate its function with modes of action including binding and cleavage. Herein, we explored how small molecule binding is correlated with cleavage induced by heterobifunctional ribonuclease targeting chimeras (RiboTACs), where RNase L is recruited to cleave the bound RNA target, in a transcriptome-wide, unbiased fashion. Only a fraction of bound targets was cleaved by RNase L, induced by RiboTAC binding. Global analysis suggested that (i) cleaved targets generally form a region of stable structure that encompasses the small molecule binding site; (ii) cleaved targets have preferred RNase L cleavage sites nearby small molecule binding sites; (iii) RiboTACs facilitate a cellular interaction between cleaved targets and RNase L; and (iv) the expression level of the target influences the extent of cleavage observed. In one example, we converted a binder of LGALS1 (galectin-1) mRNA into a RiboTAC. In MDA-MB-231 cells, the binder had no effect on galectin-1 protein levels, while the RiboTAC cleaved LGALS1 mRNA, reduced galectin-1 protein abundance, and affected galectin-1-associated oncogenic cellular phenotypes. Using LGALS1, we further assessed additional factors including the length of the linker that tethers the two components of the RiboTAC, cellular uptake, and the RNase L-recruiting module on RiboTAC potency. Collectively, these studies may facilitate triangulation of factors to enable the design of RiboTACs.


Assuntos
Transcriptoma , Humanos , Linhagem Celular Tumoral , Endorribonucleases/metabolismo , Endorribonucleases/química , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Ribonucleases/metabolismo , Ribonucleases/química
3.
Nucleic Acids Res ; 52(13): 7809-7824, 2024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-38874491

RESUMO

RNA helicases-central enzymes in RNA metabolism-often feature intrinsically disordered regions (IDRs) that enable phase separation and complex molecular interactions. In the bacterial pathogen Pseudomonas aeruginosa, the non-redundant RhlE1 and RhlE2 RNA helicases share a conserved REC catalytic core but differ in C-terminal IDRs. Here, we show how the IDR diversity defines RhlE RNA helicase specificity of function. Both IDRs facilitate RNA binding and phase separation, localizing proteins in cytoplasmic clusters. However, RhlE2 IDR is more efficient in enhancing REC core RNA unwinding, exhibits a greater tendency for phase separation, and interacts with the RNase E endonuclease, a crucial player in mRNA degradation. Swapping IDRs results in chimeric proteins that are biochemically active but functionally distinct as compared to their native counterparts. The RECRhlE1-IDRRhlE2 chimera improves cold growth of a rhlE1 mutant, gains interaction with RNase E and affects a subset of both RhlE1 and RhlE2 RNA targets. The RECRhlE2-IDRRhlE1 chimera instead hampers bacterial growth at low temperatures in the absence of RhlE1, with its detrimental effect linked to aberrant RNA droplets. By showing that IDRs modulate both protein core activities and subcellular localization, our study defines the impact of IDR diversity on the functional differentiation of RNA helicases.


Assuntos
Proteínas de Bactérias , Endorribonucleases , Proteínas Intrinsicamente Desordenadas , Pseudomonas aeruginosa , RNA Helicases , RNA Helicases/metabolismo , RNA Helicases/genética , RNA Helicases/química , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/química , Endorribonucleases/metabolismo , Endorribonucleases/química , Endorribonucleases/genética , Proteínas Intrinsicamente Desordenadas/metabolismo , Proteínas Intrinsicamente Desordenadas/química , Proteínas Intrinsicamente Desordenadas/genética , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/genética , Ligação Proteica
4.
Sci Signal ; 17(837): eadi9844, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38771918

RESUMO

Oligoadenylate synthetase 3 (OAS3) and ribonuclease L (RNase L) are components of a pathway that combats viral infection in mammals. Upon detection of viral double-stranded RNA (dsRNA), OAS3 synthesizes 2'-5'-oligo(A), which activates the RNase domain of RNase L by promoting the homodimerization and oligomerization of RNase L monomers. Activated RNase L rapidly degrades all cellular mRNAs, shutting off several cellular processes. We sought to understand the molecular mechanisms underlying the rapid activation of RNase L in response to viral infection. Through superresolution microscopy and live-cell imaging, we showed that OAS3 and RNase L concentrated into higher-order cytoplasmic complexes known as dsRNA-induced foci (dRIF) in response to dsRNA or infection with dengue virus, Zika virus, or West Nile virus. The concentration of OAS3 and RNase L at dRIF corresponded with the activation of RNase L-mediated RNA decay. We showed that dimerized/oligomerized RNase L concentrated in a liquid-like shell surrounding a core OAS3-dRIF structure and dynamically exchanged with the cytosol. These data establish that the condensation of dsRNA, OAS3, and RNase L into dRIF is a molecular switch that promotes the rapid activation of RNase L upon detection of dsRNA in mammalian cells.


Assuntos
2',5'-Oligoadenilato Sintetase , Endorribonucleases , RNA de Cadeia Dupla , Zika virus , Endorribonucleases/metabolismo , Endorribonucleases/genética , Endorribonucleases/química , Humanos , 2',5'-Oligoadenilato Sintetase/metabolismo , 2',5'-Oligoadenilato Sintetase/genética , 2',5'-Oligoadenilato Sintetase/química , RNA de Cadeia Dupla/metabolismo , RNA de Cadeia Dupla/química , RNA de Cadeia Dupla/genética , Zika virus/metabolismo , Animais , Vírus da Dengue/metabolismo , RNA Viral/metabolismo , RNA Viral/genética , Estabilidade de RNA , Vírus do Nilo Ocidental/metabolismo , Vírus do Nilo Ocidental/genética , Infecção por Zika virus/metabolismo , Infecção por Zika virus/virologia , Ativação Enzimática , Células HeLa , Células HEK293
5.
Nucleic Acids Res ; 52(10): 5438-5450, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38716860

RESUMO

In recent years, several noncanonical RNA caps derived from cofactors and metabolites have been identified. Purine-containing RNA caps have been extensively studied, with multiple decapping enzymes identified and efficient capture and sequencing protocols developed for nicotinamide adenine dinucleotide (NAD)-RNA, which allowed for a stepwise elucidation of capping functions. Despite being identified as an abundant noncanonical RNA-cap, UDP-sugar-capped RNA remains poorly understood, which is partly due to its complex in vitro preparation. Here, we describe a scalable synthesis of sugar-capped uridine-guanosine dinucleotides from readily available protected building blocks and their enzymatic conversion into several cell wall precursor-capped dinucleotides. We employed these capped dinucleotides in T7 RNA polymerase-catalyzed in vitro transcription reactions to efficiently generate RNAs capped with uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), its N-azidoacetyl derivative UDP-GlcNAz, and various cell wall precursors. We furthermore identified four enzymes capable of processing UDP-GlcNAc-capped RNA in vitro: MurA, MurB and MurC from Escherichia coli can sequentially modify the sugar-cap structure and were used to introduce a bioorthogonal, clickable moiety, and the human Nudix hydrolase Nudt5 was shown to efficiently decap UDP-GlcNAc-RNA. Our findings underscore the importance of efficient synthetic methods for capped model RNAs. Additionally, we provide useful enzymatic tools that could be utilized in the development and application of UDP-GlcNAc capture and sequencing protocols. Such protocols are essential for deepening our understanding of the widespread yet enigmatic GlcNAc modification of RNA and its physiological significance.


Assuntos
Capuzes de RNA , Uridina Difosfato N-Acetilglicosamina , Uridina Difosfato N-Acetilglicosamina/metabolismo , Capuzes de RNA/metabolismo , Endorribonucleases/metabolismo , Endorribonucleases/química , RNA Polimerases Dirigidas por DNA/metabolismo , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/química , Humanos , Escherichia coli/genética , Escherichia coli/enzimologia , Escherichia coli/metabolismo , Proteínas Virais
6.
Nucleic Acids Res ; 52(11): 6674-6686, 2024 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-38647084

RESUMO

The ability of obstacles in cellular transcripts to protect downstream but not upstream sites en masse from attack by RNase E has prompted the hypothesis that this mRNA-degrading endonuclease may scan 5'-monophosphorylated RNA linearly for cleavage sites, starting at the 5' end. However, despite its proposed regulatory importance, the migration of RNase E on RNA has never been directly observed. We have now used single-molecule FRET to monitor the dynamics of this homotetrameric enzyme on RNA. Our findings reveal that RNase E slides along unpaired regions of RNA without consuming a molecular source of energy such as ATP and that its forward progress can be impeded when it encounters a large structural discontinuity. This movement, which is bidirectional, occurs in discrete steps of variable length and requires an RNA ligand much longer than needed to occupy a single RNase E subunit. These results indicate that RNase E scans for cleavage sites by one-dimensional diffusion and suggest a possible molecular mechanism.


Assuntos
Endorribonucleases , Transferência Ressonante de Energia de Fluorescência , RNA , Endorribonucleases/metabolismo , Endorribonucleases/química , RNA/metabolismo , RNA/química , Difusão , Imagem Individual de Molécula/métodos , Trifosfato de Adenosina/metabolismo , Conformação de Ácido Nucleico
7.
J Biol Chem ; 300(3): 105729, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38336296

RESUMO

RNase P and RNase mitochondrial RNA processing (MRP) are ribonucleoproteins (RNPs) that consist of a catalytic RNA and a varying number of protein cofactors. RNase P is responsible for precursor tRNA maturation in all three domains of life, while RNase MRP, exclusive to eukaryotes, primarily functions in rRNA biogenesis. While eukaryotic RNase P is associated with more protein cofactors and has an RNA subunit with fewer auxiliary structural elements compared to its bacterial cousin, the double-anchor precursor tRNA recognition mechanism has remarkably been preserved during evolution. RNase MRP shares evolutionary and structural similarities with RNase P, preserving the catalytic core within the RNA moiety inherited from their common ancestor. By incorporating new protein cofactors and RNA elements, RNase MRP has established itself as a distinct RNP capable of processing ssRNA substrates. The structural information on RNase P and MRP helps build an evolutionary trajectory, depicting how emerging protein cofactors harmonize with the evolution of RNA to shape different functions for RNase P and MRP. Here, we outline the structural and functional relationship between RNase P and MRP to illustrate the coevolution of RNA and protein cofactors, a key driver for the extant, diverse RNP world.


Assuntos
Endorribonucleases , Evolução Molecular , Subunidades Proteicas , RNA Catalítico , Ribonuclease P , Coenzimas , Endorribonucleases/química , Endorribonucleases/metabolismo , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Ribonuclease P/química , Ribonuclease P/metabolismo , Processamento Pós-Transcricional do RNA , RNA Catalítico/genética , RNA Catalítico/metabolismo , RNA de Transferência/genética , RNA de Transferência/metabolismo , Especificidade por Substrato , Eucariotos/enzimologia
8.
Nucleic Acids Res ; 52(3): 1298-1312, 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38117986

RESUMO

Bacteria harbor diverse mechanisms to defend themselves against their viral predators, bacteriophages. In response, phages can evolve counter-defense systems, most of which are poorly understood. In T4-like phages, the gene tifA prevents bacterial defense by the type III toxin-antitoxin (TA) system toxIN, but the mechanism by which TifA inhibits ToxIN remains unclear. Here, we show that TifA directly binds both the endoribonuclease ToxN and RNA, leading to the formation of a high molecular weight ribonucleoprotein complex in which ToxN is inhibited. The RNA binding activity of TifA is necessary for its interaction with and inhibition of ToxN. Thus, we propose that TifA inhibits ToxN during phage infection by trapping ToxN on cellular RNA, particularly the abundant 16S rRNA, thereby preventing cleavage of phage transcripts. Taken together, our results reveal a novel mechanism underlying inhibition of a phage-defensive RNase toxin by a small, phage-encoded protein.


Assuntos
Bacteriófagos , Sistemas Toxina-Antitoxina , Antitoxinas/genética , Bacteriófagos/metabolismo , Endorribonucleases/genética , Endorribonucleases/química , RNA Ribossômico 16S
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...