Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 3888, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38719828

RESUMO

PRPF40A plays an important role in the regulation of pre-mRNA splicing by mediating protein-protein interactions in the early steps of spliceosome assembly. By binding to proteins at the 5´ and 3´ splice sites, PRPF40A promotes spliceosome assembly by bridging the recognition of the splices. The PRPF40A WW domains are expected to recognize proline-rich sequences in SF1 and SF3A1 in the early spliceosome complexes E and A, respectively. Here, we combine NMR, SAXS and ITC to determine the structure of the PRPF40A tandem WW domains in solution and characterize the binding specificity and mechanism for proline-rich motifs recognition. Our structure of the PRPF40A WW tandem in complex with a high-affinity SF1 peptide reveals contributions of both WW domains, which also enables tryptophan sandwiching by two proline residues in the ligand. Unexpectedly, a proline-rich motif in the N-terminal region of PRPF40A mediates intramolecular interactions with the WW tandem. Using NMR, ITC, mutational analysis in vitro, and immunoprecipitation experiments in cells, we show that the intramolecular interaction acts as an autoinhibitory filter for proof-reading of high-affinity proline-rich motifs in bona fide PRPF40A binding partners. We propose that similar autoinhibitory mechanisms are present in most WW tandem-containing proteins to enhance binding selectivity and regulation of WW/proline-rich peptide interaction networks.


Assuntos
Prolina , Ligação Proteica , Domínios WW , Humanos , Motivos de Aminoácidos , Modelos Moleculares , Prolina/metabolismo , Prolina/química , Splicing de RNA , Fatores de Processamento de RNA/metabolismo , Fatores de Processamento de RNA/química , Fatores de Processamento de RNA/genética , Espalhamento a Baixo Ângulo , Spliceossomos/metabolismo , Difração de Raios X
2.
Genes Dev ; 37(21-24): 968-983, 2023 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-37977822

RESUMO

The spliceosomal gene SF3B1 is frequently mutated in cancer. While it is known that SF3B1 hotspot mutations lead to loss of splicing factor SUGP1 from spliceosomes, the cancer-relevant SF3B1-SUGP1 interaction has not been characterized. To address this issue, we show by structural modeling that two regions flanking the SUGP1 G-patch make numerous contacts with the region of SF3B1 harboring hotspot mutations. Experiments confirmed that all the cancer-associated mutations in these regions, as well as mutations affecting other residues in the SF3B1-SUGP1 interface, not only weaken or disrupt the interaction but also alter splicing similarly to SF3B1 cancer mutations. Finally, structural modeling of a trimeric protein complex reveals that the SF3B1-SUGP1 interaction "loops out" the G-patch for interaction with the helicase DHX15. Our study thus provides an unprecedented molecular view of a protein complex essential for accurate splicing and also reveals that numerous cancer-associated mutations disrupt the critical SF3B1-SUGP1 interaction.


Assuntos
Neoplasias , Spliceossomos , Humanos , RNA Mensageiro/metabolismo , Spliceossomos/genética , Spliceossomos/metabolismo , Fatores de Processamento de RNA/química , Splicing de RNA/genética , Neoplasias/genética , Neoplasias/metabolismo , Mutação , Fosfoproteínas/metabolismo
3.
J Biol Chem ; 298(8): 102224, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35780835

RESUMO

During spliceosome assembly, the 3' splice site is recognized by sequential U2AF2 complexes, first with Splicing Factor 1 (SF1) and second by the SF3B1 subunit of the U2 small nuclear ribonuclear protein particle. The U2AF2-SF1 interface is well characterized, comprising a U2AF homology motif (UHM) of U2AF2 bound to a U2AF ligand motif (ULM) of SF1. However, the structure of the U2AF2-SF3B1 interface and its importance for pre-mRNA splicing are unknown. To address this knowledge gap, we determined the crystal structure of the U2AF2 UHM bound to a SF3B1 ULM site at 1.8-Å resolution. We discovered a distinctive trajectory of the SF3B1 ULM across the U2AF2 UHM surface, which differs from prior UHM/ULM structures and is expected to modulate the orientations of the full-length proteins. We established that the binding affinity of the U2AF2 UHM for the cocrystallized SF3B1 ULM rivals that of a nearly full-length U2AF2 protein for an N-terminal SF3B1 region. An additional SF3B6 subunit had no detectable effect on the U2AF2-SF3B1 binding affinities. We further showed that key residues at the U2AF2 UHM-SF3B1 ULM interface contribute to coimmunoprecipitation of the splicing factors. Moreover, disrupting the U2AF2-SF3B1 interface changed splicing of representative human transcripts. From analysis of genome-wide data, we found that many of the splice sites coregulated by U2AF2 and SF3B1 differ from those coregulated by U2AF2 and SF1. Taken together, these findings support distinct structural and functional roles for the U2AF2-SF1 and U2AF2-SF3B1 complexes during the pre-mRNA splicing process.


Assuntos
Precursores de RNA , Fatores de Processamento de RNA/química , Splicing de RNA , Fator de Processamento U2AF/química , Humanos , Ligantes , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Ligação Proteica , Precursores de RNA/metabolismo , Fatores de Processamento de RNA/metabolismo , Fator de Processamento U2AF/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35101980

RESUMO

In mammals, the structural basis for the interaction between U1 and U2 small nuclear ribonucleoproteins (snRNPs) during the early steps of splicing is still elusive. The binding of the ubiquitin-like (UBL) domain of SF3A1 to the stem-loop 4 of U1 snRNP (U1-SL4) contributes to this interaction. Here, we determined the 3D structure of the complex between the UBL of SF3A1 and U1-SL4 RNA. Our crystallography, NMR spectroscopy, and cross-linking mass spectrometry data show that SF3A1-UBL recognizes, sequence specifically, the GCG/CGC RNA stem and the apical UUCG tetraloop of U1-SL4. In vitro and in vivo mutational analyses support the observed intermolecular contacts and demonstrate that the carboxyl-terminal arginine-glycine-glycine-arginine (RGGR) motif of SF3A1-UBL binds sequence specifically by inserting into the RNA major groove. Thus, the characterization of the SF3A1-UBL/U1-SL4 complex expands the repertoire of RNA binding domains and reveals the capacity of RGG/RG motifs to bind RNA in a sequence-specific manner.


Assuntos
Fatores de Processamento de RNA/química , Ribonucleoproteína Nuclear Pequena U1/química , Ribonucleoproteína Nuclear Pequena U2/química , Cristalografia por Raios X , Humanos , Ressonância Magnética Nuclear Biomolecular , Motivos de Nucleotídeos , Fatores de Processamento de RNA/genética , Ribonucleoproteína Nuclear Pequena U1/genética , Ribonucleoproteína Nuclear Pequena U2/genética
5.
Science ; 375(6576): 50-57, 2022 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-34822310

RESUMO

Recognition of the intron branch site (BS) by the U2 small nuclear ribonucleoprotein (snRNP) is a critical event during spliceosome assembly. In mammals, BS sequences are poorly conserved, and unambiguous intron recognition cannot be achieved solely through a base-pairing mechanism. We isolated human 17S U2 snRNP and reconstituted in vitro its adenosine 5´-triphosphate (ATP)­dependent remodeling and binding to the pre­messenger RNA substrate. We determined a series of high-resolution (2.0 to 2.2 angstrom) structures providing snapshots of the BS selection process. The substrate-bound U2 snRNP shows that SF3B6 stabilizes the BS:U2 snRNA duplex, which could aid binding of introns with poor sequence complementarity. ATP-dependent remodeling uncoupled from substrate binding captures U2 snRNA in a conformation that competes with BS recognition, providing a selection mechanism based on branch helix stability.


Assuntos
Íntrons , Precursores de RNA/química , Ribonucleoproteína Nuclear Pequena U2/química , Spliceossomos/química , Microscopia Crioeletrônica , Humanos , Modelos Moleculares , Conformação de Ácido Nucleico , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Ligação Proteica , Conformação Proteica , Precursores de RNA/metabolismo , Splicing de RNA , Fatores de Processamento de RNA/química , Fatores de Processamento de RNA/metabolismo , RNA Nuclear Pequeno/química , RNA Nuclear Pequeno/metabolismo , Ribonucleoproteína Nuclear Pequena U2/metabolismo , Spliceossomos/metabolismo , Transativadores/química , Transativadores/metabolismo
6.
Int J Mol Sci ; 22(20)2021 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-34681880

RESUMO

The SF3B1 protein, part of the SF3b complex, recognizes the intron branch point sequence of precursor messenger RNA (pre-mRNA), thus contributing to splicing fidelity. SF3B1 is frequently mutated in cancer and is the target of distinct families of splicing modulators (SMs). Among these, H3B-8800 is of particular interest, as it induces preferential lethality in cancer cells bearing the frequent and highly pathogenic K700E SF3B1 mutation. Despite the potential of H3B-8800 to treat myeloid leukemia and other cancer types hallmarked by SF3B1 mutations, the molecular mechanism underlying its preferential lethality towards spliceosome-mutant cancer cells remains elusive. Here, microsecond-long all-atom simulations addressed the binding/dissociation mechanism of H3B-8800 to wild type and K700E SF3B1-containing SF3b (K700ESB3b) complexes at the atomic level, unlocking that the K700E mutation little affects the thermodynamics and kinetic traits of H3B-8800 binding. This supports the hypothesis that the selectivity of H3B-8800 towards mutant cancer cells is unrelated to its preferential targeting of K700ESB3b. Nevertheless, this set of simulations discloses that the K700E mutation and H3B-8800 binding affect the overall SF3b internal motion, which in turn may influence the way SF3b interacts with other spliceosome components. Finally, we unveil the existence of a putative druggable SF3b pocket in the vicinity of K700E that could be harnessed in future rational drug-discovery efforts to specifically target mutant SF3b.


Assuntos
Mutação , Neoplasias/patologia , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Piperazinas/metabolismo , Piridinas/metabolismo , Fatores de Processamento de RNA/química , Fatores de Processamento de RNA/metabolismo , Splicing de RNA , Humanos , Simulação de Dinâmica Molecular , Neoplasias/genética , Neoplasias/metabolismo , Fenótipo , Fosfoproteínas/genética , Piperazinas/química , Conformação Proteica , Piridinas/química , Fatores de Processamento de RNA/genética
7.
Science ; 373(6560): eabj5338, 2021 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-34516797

RESUMO

The human small subunit processome mediates early maturation of the small ribosomal subunit by coupling RNA folding to subsequent RNA cleavage and processing steps. We report the high-resolution cryo­electron microscopy structures of maturing human small subunit (SSU) processomes at resolutions of 2.7 to 3.9 angstroms. These structures reveal the molecular mechanisms that enable crucial progressions during SSU processome maturation. RNA folding states within these particles are communicated to and coordinated with key enzymes that drive irreversible steps such as targeted exosome-mediated RNA degradation, protein-guided site-specific endonucleolytic RNA cleavage, and tightly controlled RNA unwinding. These conserved mechanisms highlight the SSU processome's impressive structural plasticity, which endows this 4.5-megadalton nucleolar assembly with the distinctive ability to mature the small ribosomal subunit from within.


Assuntos
Nucléolo Celular/ultraestrutura , Dobramento de RNA , Estabilidade de RNA , RNA Nucleolar Pequeno/química , Nucléolo Celular/metabolismo , Microscopia Crioeletrônica , RNA Helicases DEAD-box/química , Humanos , Clivagem do RNA , Fatores de Processamento de RNA/química
8.
Elife ; 102021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34263725

RESUMO

Correct 3'end processing of mRNAs is one of the regulatory cornerstones of gene expression. In a parasite that must adapt to the regulatory requirements of its multi-host life style, there is a need to adopt additional means to partition the distinct transcriptional signatures of the closely and tandemly arranged stage-specific genes. In this study, we report our findings in T. gondii of an m6A-dependent 3'end polyadenylation serving as a transcriptional barrier at these loci. We identify the core polyadenylation complex within T. gondii and establish CPSF4 as a reader for m6A-modified mRNAs, via a YTH domain within its C-terminus, a feature which is shared with plants. We bring evidence of the specificity of this interaction both biochemically, and by determining the crystal structure at high resolution of the T. gondii CPSF4-YTH in complex with an m6A-modified RNA. We show that the loss of m6A, both at the level of its deposition or its recognition is associated with an increase in aberrantly elongated chimeric mRNAs emanating from impaired transcriptional termination, a phenotype previously noticed in the plant model Arabidopsis thaliana. Nanopore direct RNA sequencing shows the occurrence of transcriptional read-through breaching into downstream repressed stage-specific genes, in the absence of either CPSF4 or the m6A RNA methylase components in both T. gondii and A. thaliana. Taken together, our results shed light on an essential regulatory mechanism coupling the pathways of m6A metabolism directly to the cleavage and polyadenylation processes, one that interestingly seem to serve, in both T. gondii and A. thaliana, as a guardian against aberrant transcriptional read-throughs.


Assuntos
Genes Controladores do Desenvolvimento , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Poliadenilação , Toxoplasma/metabolismo , Transcriptoma , Arabidopsis/genética , Sítios de Ligação , Fator de Especificidade de Clivagem e Poliadenilação/metabolismo , Regulação da Expressão Gênica , Humanos , Glicoproteínas de Membrana/química , Metiltransferases/metabolismo , Modelos Moleculares , Proteínas do Tecido Nervoso/química , Fatores de Processamento de RNA/química , RNA Mensageiro/metabolismo , Leitura , Análise de Sequência de RNA , Dedos de Zinco
9.
Cell Chem Biol ; 28(9): 1356-1365.e4, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-33784500

RESUMO

RNA splicing, a highly conserved process in eukaryotic gene expression, is seen as a promising target for anticancer agents. Splicing is associated with other RNA processing steps, such as transcription and nuclear export; however, our understanding of the interaction between splicing and other RNA regulatory mechanisms remains incomplete. Moreover, the impact of chemical splicing inhibition on long non-coding RNAs (lncRNAs) has been poorly understood. Here, we demonstrate that spliceostatin A (SSA), a chemical splicing modulator that binds to the SF3B subcomplex of the U2 small nuclear ribonucleoprotein particle (snRNP), limits U1 snRNP availability in splicing, resulting in premature cleavage and polyadenylation of MALAT1, a nuclear lncRNA, as well as protein-coding mRNAs. Therefore, truncated transcripts are exported into the cytoplasm and translated, resulting in aberrant protein products. Our work demonstrates that active recycling of the splicing machinery maintains homeostasis of RNA processing beyond intron excision.


Assuntos
Fosfoproteínas/antagonistas & inibidores , Piranos/farmacologia , Fatores de Processamento de RNA/antagonistas & inibidores , RNA Longo não Codificante/metabolismo , Ribonucleoproteína Nuclear Pequena U1/antagonistas & inibidores , Compostos de Espiro/farmacologia , Feminino , Células HeLa , Humanos , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Poliadenilação/efeitos dos fármacos , Piranos/química , Splicing de RNA/efeitos dos fármacos , Fatores de Processamento de RNA/química , Fatores de Processamento de RNA/metabolismo , Ribonucleoproteína Nuclear Pequena U1/química , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Compostos de Espiro/química , Células Tumorais Cultivadas
10.
Biochem Biophys Res Commun ; 546: 21-28, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33561744

RESUMO

SF3B1, an essential component of the U2 snRNP, is frequently mutated in cancers. Cancer-associated SF3B1 mutation causes aberrant RNA splicing, mostly at 3' splice sites (3'ss). RNA splicing of DVL2, a regulator of Notch signaling, is affected by SF3B1 mutation. Here, we report that the mutated SF3B1 use an alternative branchpoint sequence (BPS) for the aberrant splicing of DVL2, which has a higher affinity to U2 snRNA than the BPS for the canonical splicing of DVL2. Swapping the position of the alternative BPS with the position of the canonical BPS decreased the aberrant splicing of DVL2, suggesting that the mutated SF3B1 prefers to use BPS with high affinity to U2 snRNA for splicing. Additionally, swapping the positions of two BPSs associated with the canonical splicing of DVL2 demonstrated that both the affinity to the U2 snRNA and the distance to the 3'ss are important to the selection of BPS. Importantly, the aberrant splicing of DVL2 does not require the canonical 3'ss and the canonical polypyrimidine tract, which reveals a novel type of aberrant splicing induced by SF3B1 mutation. These findings provide a more comprehensive understanding of the mechanisms underlying aberrant splicing induced by SF3B1 mutation in cancer.


Assuntos
Processamento Alternativo , Proteínas Desgrenhadas/genética , Mutação , Neoplasias/genética , Fosfoproteínas/genética , Fatores de Processamento de RNA/genética , Sequência de Bases , Proteínas Desgrenhadas/química , Humanos , Fosfoproteínas/química , Sítios de Splice de RNA/genética , Fatores de Processamento de RNA/química , RNA Nuclear Pequeno/genética
11.
Int J Biochem Cell Biol ; 132: 105919, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33422691

RESUMO

The AAA-ATPase NVL2 associates with an RNA helicase MTR4 and the nuclear RNA exosome in the course of ribosome biogenesis. In our proteomic screen, we had identified a ribosome biogenesis factor WDR74 as a MTR4-interacting partner, whose dissociation is stimulated by the ATP hydrolysis of NVL2. In this study, we report the identification of splicing factor 30 (SPF30), another MTR4-interacting protein with a similar regulatory mechanism. SPF30 is a pre-mRNA splicing factor harboring a Tudor domain in its central region, which regulates various cellular events by binding to dimethylarginine-modified proteins. The interaction between SPF30 and the exosome core is mediated by MTR4 and RRP6, a catalytic component of the nuclear exosome. The N- and C-terminal regions, but not the Tudor domain, of SPF30 are involved in the association with MTR4 and the exosome. The knockdown of SPF30 caused subtle delay in the 12S pre-rRNA processing to mature 5.8S rRNA, even though no obvious effect was observed on the ribosome subunit profile in the cells. Shotgun proteomic analysis to search for SPF30-interacting proteins indicated its role in ribosome biogenesis, pre-mRNA splicing, and box C/D snoRNA biogenesis. These results suggest that SPF30 collaborates with the MTR4-exosome machinery to play a functional role in multiple RNA metabolic pathways, some of which may be regulated by the ATP hydrolysis of NVL2.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , Exossomos/genética , RNA Helicases/metabolismo , Fatores de Processamento de RNA/química , Fatores de Processamento de RNA/metabolismo , Estabilidade de RNA , Proteínas do Complexo SMN/química , Proteínas do Complexo SMN/metabolismo , Humanos , Ligação Proteica , Domínios Proteicos
12.
Science ; 371(6535)2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33509932

RESUMO

The minor spliceosome mediates splicing of the rare but essential U12-type precursor messenger RNA. Here, we report the atomic features of the activated human minor spliceosome determined by cryo-electron microscopy at 2.9-angstrom resolution. The 5' splice site and branch point sequence of the U12-type intron are recognized by the U6atac and U12 small nuclear RNAs (snRNAs), respectively. Five newly identified proteins stabilize the conformation of the catalytic center: The zinc finger protein SCNM1 functionally mimics the SF3a complex of the major spliceosome, the RBM48-ARMC7 complex binds the γ-monomethyl phosphate cap at the 5' end of U6atac snRNA, the U-box protein PPIL2 coordinates loop I of U5 snRNA and stabilizes U5 small nuclear ribonucleoprotein (snRNP), and CRIPT stabilizes U12 snRNP. Our study provides a framework for the mechanistic understanding of the function of the human minor spliceosome.


Assuntos
Spliceossomos/química , Spliceossomos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas do Domínio Armadillo/química , Proteínas do Domínio Armadillo/metabolismo , Microscopia Crioeletrônica , Ciclofilinas/química , Ciclofilinas/metabolismo , Humanos , Íntrons , Modelos Moleculares , Conformação de Ácido Nucleico , Conformação Proteica , Domínios Proteicos , Precursores de RNA/química , Precursores de RNA/metabolismo , Splicing de RNA , Fatores de Processamento de RNA/química , Fatores de Processamento de RNA/metabolismo , RNA Nuclear Pequeno/química , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteína Nuclear Pequena U5/química , Ribonucleoproteína Nuclear Pequena U5/metabolismo , Ribonucleoproteínas Nucleares Pequenas/metabolismo
13.
J Chem Theory Comput ; 17(2): 1240-1249, 2021 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-33472367

RESUMO

N6-Methyladenosine (m6A) is the most frequent modification in eukaryotic messenger RNA (mRNA) and its cellular processing and functions are regulated by the reader proteins YTHDCs and YTHDFs. However, the mechanism of m6A recognition by the reader proteins is still elusive. Here, we investigate this recognition process by combining atomistic simulations, site-directed mutagenesis, and biophysical experiments using YTHDC1 as a model. We find that the N6 methyl group of m6A contributes to the binding through its specific interactions with an aromatic cage (formed by Trp377 and Trp428) and also by favoring the association-prone conformation of m6A-containing RNA in solution. The m6A binding site dynamically equilibrates between multiple metastable conformations with four residues being involved in the regulation of m6A binding (Trp428, Met438, Ser378, and Thr379). Trp428 switches between two conformational states to build and dismantle the aromatic cage. Interestingly, mutating Met438 and Ser378 to alanine does not alter m6A binding to the protein but significantly redistributes the binding enthalpy and entropy terms, i.e., enthalpy-entropy compensation. Such compensation is reasoned by different entropy-enthalpy transduction associated with both conformational changes of the wild-type and mutant proteins and the redistribution of water molecules. In contrast, the point mutant Thr379Val significantly changes the thermal stability and binding capability of YTHDC1 to its natural ligand. Additionally, thermodynamic analysis and free energy calculations shed light on the role of a structural water molecule that synergistically binds to YTHDC1 with m6A and acts as the hub of a hydrogen-bond network. Taken together, the experimental data and simulation results may accelerate the discovery of chemical probes, m6A-editing tools, and drug candidates against reader proteins.


Assuntos
Adenosina/análogos & derivados , Proteínas do Tecido Nervoso/química , Fatores de Processamento de RNA/química , Termodinâmica , Adenosina/química , Calorimetria/métodos , Cristalografia por Raios X , Metilação , Conformação Molecular , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Domínios Proteicos , Água/química
14.
Chembiochem ; 22(3): 565-570, 2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-32975902

RESUMO

The 55-residue OCRE domains of the splicing factors RBM5 and RBM10 contain 15 tyrosines in compact, globular folds. At 25 °C, all 15 tyrosines show symmetric 1 H NMR spectra, with averaged signals for the pairs of δ- and ϵ-ring hydrogens. At 4 °C, two tyrosines were identified as showing 1 H NMR line-broadening due to lowered frequency of the ring-flipping. For the other 13 tyrosine rings, it was not evident, from the 1 H NMR data alone, whether they were either all flipping at high frequencies, or whether slowed flipping went undetected due to small chemical-shift differences between pairs of exchanging ring hydrogen atoms. Here, we integrate 1 H NMR spectroscopy and molecular dynamics (MD) simulations to determine the tyrosine ring-flip frequencies. In the RBM10-OCRE domain, we found that, for 11 of the 15 tyrosines, these frequencies are in the range 2.0×106 to 1.3×108  s-1 , and we established an upper limit of <1.0×106  s-1 for the remaining four residues. The experimental data and the MD simulation are mutually supportive, and their combined use extends the analysis of aromatic ring-flip events beyond the limitations of routine 1 H NMR line-shape analysis into the nanosecond frequency range.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Ligação a DNA/química , Simulação de Dinâmica Molecular , Fatores de Processamento de RNA/química , Proteínas de Ligação a RNA/química , Proteínas Supressoras de Tumor/química , Tirosina/química , Motivos de Aminoácidos , Humanos , Espectroscopia de Prótons por Ressonância Magnética
15.
Dev Cell ; 56(1): 111-124.e6, 2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33238149

RESUMO

To date, the effects of specific modification types and sites on protein lifetime have not been systematically illustrated. Here, we describe a proteomic method, DeltaSILAC, to quantitatively assess the impact of site-specific phosphorylation on the turnover of thousands of proteins in live cells. Based on the accurate and reproducible mass spectrometry-based method, a pulse labeling approach using stable isotope-labeled amino acids in cells (pSILAC), phosphoproteomics, and a unique peptide-level matching strategy, our DeltaSILAC profiling revealed a global, unexpected delaying effect of many phosphosites on protein turnover. We further found that phosphorylated sites accelerating protein turnover are functionally selected for cell fitness, enriched in Cyclin-dependent kinase substrates, and evolutionarily conserved, whereas the glutamic acids surrounding phosphosites significantly delay protein turnover. Our method represents a generalizable approach and provides a rich resource for prioritizing the effects of phosphorylation sites on protein lifetime in the context of cell signaling and disease biology.


Assuntos
Marcação por Isótopo/métodos , Espectrometria de Massas/métodos , Fosfoproteínas/metabolismo , Proteólise , Proteoma/metabolismo , Proteômica/métodos , Sequência de Aminoácidos , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Glutamatos/metabolismo , Humanos , Peptídeos/metabolismo , Peroxirredoxina VI/química , Peroxirredoxina VI/metabolismo , Fosfoproteínas/química , Fosforilação , Proteoma/genética , Fatores de Processamento de RNA/química , Fatores de Processamento de RNA/metabolismo , Transdução de Sinais/genética
16.
Angew Chem Int Ed Engl ; 60(6): 3163-3169, 2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33108679

RESUMO

Defects in the functions of RNA binding proteins (RBPs) are at the origin of many diseases; however, targeting RBPs with conventional drugs has proven difficult. PROTACs are a new class of drugs that mediate selective degradation of a target protein through a cell's ubiquitination machinery. PROTACs comprise a moiety that binds the selected protein, conjugated to a ligand of an E3 ligase. Herein, we introduce RNA-PROTACs as a new concept in the targeting of RBPs. These chimeric structures employ small RNA mimics as targeting groups that dock the RNA-binding site of the RBP, whereupon a conjugated E3-recruiting peptide derived from the HIF-1α protein directs the RBP for proteasomal degradation. We performed a proof-of-concept demonstration with the degradation of two RBPs-a stem cell factor LIN28 and a splicing factor RBFOX1-and showed their use in cancer cell lines. The RNA-PROTAC approach opens the way to rapid, selective targeting of RBPs in a rational and general fashion.


Assuntos
Proteínas de Ligação a RNA/metabolismo , RNA/metabolismo , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Humanos , Oligonucleotídeos/química , Oligonucleotídeos/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Proteólise , RNA/química , Fatores de Processamento de RNA/química , Fatores de Processamento de RNA/metabolismo , Proteínas de Ligação a RNA/química , Ubiquitina-Proteína Ligases/química
17.
Neuron ; 109(2): 241-256.e9, 2021 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-33220177

RESUMO

Autosomal-recessive cerebellar hypoplasia and ataxia constitute a group of heterogeneous brain disorders caused by disruption of several fundamental cellular processes. Here, we identified 10 families showing a neurodegenerative condition involving pontocerebellar hypoplasia with microcephaly (PCHM). Patients harbored biallelic mutations in genes encoding the spliceosome components Peptidyl-Prolyl Isomerase Like-1 (PPIL1) or Pre-RNA Processing-17 (PRP17). Mouse knockouts of either gene were lethal in early embryogenesis, whereas PPIL1 patient mutation knockin mice showed neuron-specific apoptosis. Loss of either protein affected splicing integrity, predominantly affecting short and high GC-content introns and genes involved in brain disorders. PPIL1 and PRP17 form an active isomerase-substrate interaction, but we found that isomerase activity is not critical for function. Thus, we establish disrupted splicing integrity and "major spliceosome-opathies" as a new mechanism underlying PCHM and neurodegeneration and uncover a non-enzymatic function of a spliceosomal proline isomerase.


Assuntos
Proteínas de Ciclo Celular/genética , Doenças Cerebelares/genética , Microcefalia/genética , Mutação/genética , Peptidilprolil Isomerase/genética , Fatores de Processamento de RNA/genética , Spliceossomos/genética , Sequência de Aminoácidos , Animais , Proteínas de Ciclo Celular/química , Doenças Cerebelares/complicações , Doenças Cerebelares/diagnóstico por imagem , Estudos de Coortes , Feminino , Técnicas de Inativação de Genes/métodos , Células HEK293 , Transtornos Heredodegenerativos do Sistema Nervoso/complicações , Transtornos Heredodegenerativos do Sistema Nervoso/diagnóstico por imagem , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microcefalia/complicações , Microcefalia/diagnóstico por imagem , Linhagem , Peptidilprolil Isomerase/química , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Fatores de Processamento de RNA/química
18.
Int J Mol Sci ; 21(24)2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33333932

RESUMO

Uveal melanoma (UM) is the most common primary intraocular malignancy of the eye. It has a high metastatic potential and mainly spreads to the liver. Genetics play a vital role in tumor classification and prognostication of UM metastatic disease. One of the driver genes mutated in metastasized UM is subunit 1 of splicing factor 3b (SF3B1), a component of the spliceosome complex. Recurrent mutations in components of the spliceosome complex are observed in UM and other malignancies, suggesting an important role in tumorigenesis. SF3B1 is the most common mutated spliceosome gene and in UM it is associated with late-onset metastasis. This review summarizes the genetic and epigenetic insights of spliceosome mutations in UM. They form a distinct subgroup of UM and have similarities with other spliceosome mutated malignancies.


Assuntos
Melanoma/genética , Mutação , Fatores de Processamento de RNA/genética , Neoplasias Uveais/genética , Substituição de Aminoácidos , Éxons , Frequência do Gene , Humanos , Melanoma/metabolismo , Melanoma/mortalidade , Melanoma/patologia , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Splicing de RNA , Fatores de Processamento de RNA/química , Fatores de Processamento de RNA/metabolismo , Spliceossomos , Telômero/genética , Neoplasias Uveais/metabolismo , Neoplasias Uveais/mortalidade , Neoplasias Uveais/patologia
19.
Nat Commun ; 11(1): 5621, 2020 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-33159082

RESUMO

Interactions between U2AF homology motifs (UHMs) and U2AF ligand motifs (ULMs) play a crucial role in early spliceosome assembly in eukaryotic gene regulation. UHM-ULM interactions mediate heterodimerization of the constitutive splicing factors U2AF65 and U2AF35 and between other splicing factors that regulate spliceosome assembly at the 3' splice site, where UHM domains of alternative splicing factors, such as SPF45 and PUF60, contribute to alternative splicing regulation. Here, we performed high-throughput screening using fluorescence polarization assays with hit validation by NMR and identified phenothiazines as general inhibitors of UHM-ULM interactions. NMR studies show that these compounds occupy the tryptophan binding pocket of UHM domains. Co-crystal structures of the inhibitors with the PUF60 UHM domain and medicinal chemistry provide structure-activity-relationships and reveal functional groups important for binding. These inhibitors inhibit early spliceosome assembly on pre-mRNA substrates in vitro. Our data show that spliceosome assembly can be inhibited by targeting UHM-ULM interactions by small molecules, thus extending the toolkit of splicing modulators for structural and biochemical studies of the spliceosome and splicing regulation.


Assuntos
Fenotiazinas/química , Fenotiazinas/farmacologia , Spliceossomos/efeitos dos fármacos , Spliceossomos/metabolismo , Processamento Alternativo , Humanos , Ligação Proteica/efeitos dos fármacos , Domínios Proteicos , Precursores de RNA/genética , Precursores de RNA/metabolismo , Fatores de Processamento de RNA/química , Fatores de Processamento de RNA/genética , Fatores de Processamento de RNA/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Spliceossomos/genética , Fator de Processamento U2AF/química , Fator de Processamento U2AF/genética , Fator de Processamento U2AF/metabolismo
20.
PLoS One ; 15(11): e0242725, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33253191

RESUMO

Pre-mRNA splicing is critical for achieving required amounts of a transcript at a given time and for regulating production of encoded protein. A given pre-mRNA may be spliced in many ways, or not at all, giving rise to multiple gene products. Numerous splicing factors are recruited to pre-mRNA splice sites to ensure proper splicing. One such factor, the 60 kDa poly(U)-binding splicing factor (PUF60), is recruited to sites that are not always spliced, but rather function as alternative splice sites. In this study, we characterized the interaction of PUF60 with a splice site from the adenovirus major late promoter (the AdML 3' splice site, AdML3'). We found that the PUF60-AdML3' dissociation constants are in the micromolar range, with the binding affinity predominantly provided by PUF60's two central RNA recognition motifs (RRMs). A 1.95 Å crystal structure of the two PUF60 RRMs in complex with AdML3' revealed a dimeric organization placing two stretches of nucleic acid tracts in opposing directionalities, which can cause looping of nucleic acid and explain how PUF60 affects pre-mRNA geometry to effect splicing. Solution characterization of this complex by light-scattering and UV/Vis spectroscopy suggested a potential 2:1 (PUF602:AdML3') stoichiometry, consistent with the crystal structure. This work defines the sequence specificity of the alternative splicing factor PUF60 at the pre-mRNA 3' splice site. Our observations suggest that control of pre-mRNA directionality is important in the early stage of spliceosome assembly, and advance our understanding of the molecular mechanism by which alternative and constitutive splicing factors differentiate among 3' splice sites.


Assuntos
Adenoviridae/química , Íntrons , Regiões Promotoras Genéticas , Sítios de Splice de RNA , Fatores de Processamento de RNA/química , RNA Viral/química , Proteínas Repressoras/química , Adenoviridae/metabolismo , Cristalografia por Raios X , Humanos , Fatores de Processamento de RNA/metabolismo , RNA Viral/metabolismo , Proteínas Repressoras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...