Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomol NMR Assign ; 18(1): 71-78, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38551798

RESUMEN

The intraflagellar transport (IFT) machinery plays a crucial role in the bidirectional trafficking of components necessary for ciliary signaling, such as the Hedgehog, Wnt/PCR, and cAMP/PKA systems. Defects in some components of the IFT machinery cause dysfunction, leading to a wide range of human diseases and developmental disorders termed ciliopathies, such as nephronophthisis. The IFT machinery comprises three sub-complexes: BBsome, IFT-A, and IFT-B. The IFT protein 54 (IFT54) is an important component of the IFT-B sub-complex. In anterograde movement, IFT54 binds to active kinesin-II, walking along the cilia microtubule axoneme and carrying the dynein-2 complex in an inactive state, which works for retrograde movement. Several mutations in IFT54 are known to cause Senior-Loken syndrome, a ciliopathy. IFT54 possesses a divergent Calponin Homology (CH) domain termed as NN-CH domain at its N-terminus. However, several aspects of the function of the NN-CH domain of IFT54 are still obscure. Here, we report the 1H, 15N, and 13C resonance assignments of the NN-CH domain of human IFT54 and its solution structure. The NN-CH domain of human IFT54 adopts essentially the α1-α2-α3-α4-α5 topology as that of mouse IFT54, whose structure was determined by X-ray crystallographic study. The structural information and assignments obtained in this study shed light on the molecular function of the NN-CH domain in IFT54.


Asunto(s)
Resonancia Magnética Nuclear Biomolecular , Dominios Proteicos , Humanos , Calponinas , Proteínas de Microfilamentos/química , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/metabolismo , Soluciones , Isótopos de Nitrógeno
2.
Nat Chem Biol ; 20(5): 605-614, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38267667

RESUMEN

In response to environmental changes, cells flexibly and rapidly alter gene expression through translational controls. In plants, the translation of NIP5;1, a boric acid diffusion facilitator, is downregulated in response to an excess amount of boric acid in the environment through upstream open reading frames (uORFs) that consist of only AUG and stop codons. However, the molecular details of how this minimum uORF controls translation of the downstream main ORF in a boric acid-dependent manner have remained unclear. Here, by combining ribosome profiling, translation complex profile sequencing, structural analysis with cryo-electron microscopy and biochemical assays, we show that the 80S ribosome assembled at AUG-stop migrates into the subsequent RNA segment, followed by downstream translation initiation, and that boric acid impedes this process by the stable confinement of eukaryotic release factor 1 on the 80S ribosome on AUG-stop. Our results provide molecular insight into translation regulation by a minimum and environment-responsive uORF.


Asunto(s)
Ácidos Bóricos , Biosíntesis de Proteínas , Ribosomas , Ribosomas/metabolismo , Ácidos Bóricos/química , Factores de Terminación de Péptidos/metabolismo , Factores de Terminación de Péptidos/química , Factores de Terminación de Péptidos/genética , Microscopía por Crioelectrón , Sistemas de Lectura Abierta , Codón de Terminación , Arabidopsis/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética
3.
Commun Biol ; 6(1): 284, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36932164

RESUMEN

The control of cell movement through manipulation of cytoskeletal structure has therapeutic prospects notably in the development of novel anti-metastatic drugs. In this study, we determine the structure of Ras-binding domain (RBD) of ELMO1, a protein involved in cytoskeletal regulation, both alone and in complex with the activator RhoG and verify its targetability through computational nanobody design. Using our dock-and-design approach optimized with native-like initial pose selection, we obtain Nb01, a detectable binder from scratch in the first-round design. An affinity maturation step guided by structure-activity relationship at the interface generates 23 Nb01 sequence variants and 17 of them show enhanced binding to ELMO1-RBD and are modeled to form major spatial overlaps with RhoG. The best binder, Nb29, inhibited ELMO1-RBD/RhoG interaction. Molecular dynamics simulation of the flexibility of CDR2 and CDR3 of Nb29 reveal the design of stabilizing mutations at the CDR-framework junctions potentially confers the affinity enhancement.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Simulación de Dinámica Molecular , Proteínas de Unión al GTP rho , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
4.
Biomol NMR Assign ; 16(2): 297-303, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35666428

RESUMEN

Ribosome biogenesis is a complicated, multistage process coordinated by ribosome assembly factors. Ribosome binding factor A (RbfA) is a bacterial one, which possesses a single structural type-II KH domain. By this domain, RbfA binds to a 16S rRNA precursor in small ribosomal subunits to promote its 5'-end processing. The human RbfA homolog, mtRbfA, binds to 12S rRNAs in the mitoribosomal small subunits and promotes its critical maturation process, the dimethylation of two highly conserved consecutive adenines, which differs from that of RbfA. However, the structural basis of the mtRbfA-mediated maturation process is poorly understood. Herein, we report the 1H, 15N, and 13C resonance assignments of the KH domain of mtRbfA and its solution structure. The mtRbfA domain adopts essentially the same α1-ß1-ß2-α2(kinked)-ß3 topology as the type-II KH domain. Comparison with the RbfA counterpart showed structural differences in specific regions that function as a putative RNA-binding site. Particularly, the α2 helix of mtRbfA forms a single helix with a moderate kink at the Ser-Ala-Ala sequence, whereas the corresponding α2 helix of RbfA is interrupted by a distinct kink at the Ala-x-Gly sequence, characteristic of bacterial RbfA proteins, to adopt an α2-kink-α3 conformation. Additionally, the region linking α1 and ß1 differs considerably in the sequence and structure between RbfA and mtRbfA. These findings suggest some variations of the RNA-binding mode between them and provide a structural basis for mtRbfA function in mitoribosome biogenesis.


Asunto(s)
Proteínas de Escherichia coli , Proteínas Mitocondriales/química , Ribosomas Mitocondriales , Proteínas de Unión al ARN/química , Proteínas Bacterianas/química , Proteínas de Escherichia coli/química , Humanos , Ribosomas Mitocondriales/metabolismo , Resonancia Magnética Nuclear Biomolecular , Precursores del ARN/metabolismo , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo , Proteínas Ribosómicas/química , Ribosomas/metabolismo , Vitamina B 12/análogos & derivados
5.
Biomol NMR Assign ; 16(1): 41-49, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34783967

RESUMEN

Matrin-3 is a multifunctional protein that binds to both DNA and RNA. Its DNA-binding activity is linked to the formation of the nuclear matrix and transcriptional regulation, while its RNA-binding activity is linked to mRNA metabolism including splicing, transport, stabilization, and degradation. Correspondingly, Matrin-3 has two zinc finger domains for DNA binding and two consecutive RNA recognition motif (RRM) domains for RNA binding. Matrin-3 has been reported to cause amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) when its disordered region contains pathogenic mutations. Simultaneously, it has been shown that the RNA-binding activity of Matrin-3 mediated by its RRM domains, affects the formation of insoluble cytoplasmic granules, which are related to the pathogenic mechanism of ALS/FTD. Thus, the effect of the RRM domains on the phase separation of condensed protein/RNA mixtures has to be clarified for a comprehensive understanding of ALS/FTD. Here, we report the 1H, 15N, and 13C resonance assignments of the two RNA binding domains and their solution structures. The resonance assignments and the solution structures obtained in this work will contribute to the elucidation of the molecular basis of Matrin-3 in the pathogenic mechanism of ALS and/or FTD.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Demencia Frontotemporal/genética , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Humanos , Resonancia Magnética Nuclear Biomolecular , ARN/metabolismo , Motivo de Reconocimiento de ARN
6.
Biomol NMR Assign ; 15(1): 1-7, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32930954

RESUMEN

In humans, YTH (YT521-B homology) domain containing protein 2 (YTHDC2) plays a crucial role in the phase-shift from mitosis to meiosis. YTH domains bind to methylated adenosine nucleotides such as m6A. In a phylogenic tree, the YTH domain of YTHDC2 (YTH2) and that of the YTH containing protein YTHDC1 (YTH1) belong to the same sub-group. However, the binding affinity of m6A differs between these proteins. Here, we report 1H, 13C and 15N resonance assignment of YTH2 and its solution structure to examine the difference of the structural architecture and the dynamic properties of YTH1 and YTH2. YTH2 adopts a ß1-α1-ß2-α2-ß3-ß4-ß5-α3-ß6-α4 topology, which was also observed in YTH1. However, the ß4-ß5 loops of YTH1 and YTH2 are distinct in length and amino acid composition. Our data revealed that, unlike in YTH1, the structure of m6A-binding pocket of YTH2 formed by the ß4-ß5 loop is stabilized by electrostatic interaction. This assignment and the structural information for YTH2 will provide the insight on the further functional research of YTHDC2.


Asunto(s)
Resonancia Magnética Nuclear Biomolecular , Adenosina , ARN
7.
Sci Rep ; 10(1): 20069, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33208800

RESUMEN

Gaussia luciferase (GLuc) is a small luciferase (18.2 kDa; 168 residues) and is thus attracting much attention as a reporter protein, but the lack of structural information is hampering further application. Here, we report the first solution structure of a fully active, recombinant GLuc determined by heteronuclear multidimensional NMR. We obtained a natively folded GLuc by bacterial expression and efficient refolding using a Solubility Enhancement Petide (SEP) tag. Almost perfect assignments of GLuc's 1H, 13C and 15N backbone signals were obtained. GLuc structure was determined using CYANA, which automatically identified over 2500 NOEs of which > 570 were long-range. GLuc is an all-alpha-helix protein made of nine helices. The region spanning residues 10-18, 36-81, 96-145 and containing eight out of the nine helices was determined with a Cα-atom RMSD of 1.39 Å ± 0.39 Å. The structure of GLuc is novel and unique. Two homologous sequential repeats form two anti-parallel bundles made by 4 helices and tied together by three disulfide bonds. The N-terminal helix 1 is grabbed by these 4 helices. Further, we found a hydrophobic cavity where several residues responsible for bioluminescence were identified in previous mutational studies, and we thus hypothesize that this is a catalytic cavity, where the hydrophobic coelenterazine binds and the bioluminescence reaction takes place.


Asunto(s)
Copépodos/enzimología , Disulfuros/química , Imidazoles/metabolismo , Luciferasas/química , Luciferasas/metabolismo , Resonancia Magnética Nuclear Biomolecular/métodos , Pirazinas/metabolismo , Secuencia de Aminoácidos , Animales , Conformación Proteica , Dominios Proteicos , Pliegue de Proteína
8.
Eur Biophys J ; 48(4): 361-369, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30937482

RESUMEN

Transcriptional repressor Bach1 plays an important role in antioxidant response. Bach1 function is regulated by heme binding to the four cysteine-proline (CP) motifs in Bach1, which leads to inhibition of its activity. Three of these CP motifs are located N-terminal to the bZip (basic leucine zipper) domain that is responsible for DNA binding. Based on sequence analysis, the region surrounding these CP motifs was expected to be intrinsically disordered. Bach1 is one of few known intrinsically disordered proteins that accept multiple heme molecules for functional regulation, but the molecular mechanisms of heme binding and functional regulation remain unclear. Uncovering these mechanisms is important for understanding Bach1-mediated antioxidant response. Biophysical characterization revealed that 5-coordinated heme binding was unique to the CP motifs within the heme-binding region of Bach1, whereas 6-coordinated binding occurred nonspecifically. Comparison of the wild-type protein and a CP motif mutant indicated that the level of 6-coordinated heme binding was reduced in the absence of 5-coordinated heme binding. Analytical ultracentrifugation showed that the CP motif mutant protein had a more elongated conformation than the wild-type protein, suggesting that cysteines within the CP motifs contribute to intramolecular interactions in Bach1. Thus, heme binding at the CP motifs induces a global conformational change in the Bach1 heme-binding region, and this conformational change, in turn, regulates the biological activity of Bach1.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/química , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Fenómenos Biofísicos , Hemo/metabolismo , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Células HEK293 , Humanos , Ratones , Unión Proteica
9.
Structure ; 27(5): 741-748.e3, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30853411

RESUMEN

The Dedicator Of CytoKinesis (DOCK) family of atypical guanine nucleotide exchange factors activates the Rho family GTPases Rac and/or Cdc42 through DOCK homology region 2 (DHR-2). Previous structural analyses of the DHR-2 domains of DOCK2 and DOCK9 have shown that they preferentially bind Rac1 and Cdc42, respectively; however, the molecular mechanism by which DHR-2 distinguishes between these GTPases is unclear. Here we report the crystal structure of the Cdc42-bound form of the DOCK7 DHR-2 domain showing dual specificity for Rac1 and Cdc42. The structure revealed increased substrate tolerance of DOCK7 at the interfaces with switch 1 and residue 56 of Cdc42. Furthermore, molecular dynamics simulations showed a closed-to-open conformational change in the DOCK7 DHR-2 domain between the Cdc42- and Rac1-bound states by lobe B displacement. Our results suggest that lobe B acts as a sensor for identifying different switch 1 conformations and explain how DOCK7 recognizes both Rac1 and Cdc42.


Asunto(s)
Proteínas Activadoras de GTPasa/química , Factores de Intercambio de Guanina Nucleótido/química , Especificidad por Sustrato , Proteína de Unión al GTP cdc42/química , Proteína de Unión al GTP rac1/química , Cristalización , Cristalografía por Rayos X , Humanos , Conformación Molecular , Simulación de Dinámica Molecular , Mutagénesis
10.
Biomol NMR Assign ; 11(2): 265-268, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28808919

RESUMEN

Musashi1 (Msi1) is an RNA-binding protein that is involved in cell fate determination. Here, we report the 1H, 15N, and 13C resonance assignments of Msi1 second RNA-binding domain in free form and in complex with RNA. The assignments can be utilized for NMR structure and dynamics analyses of the Msi1:RNA complex, and moreover, for chemical shift perturbation analyses to evaluate the binding of potential small molecule inhibitors against Msi1:RNA interaction.


Asunto(s)
Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Resonancia Magnética Nuclear Biomolecular , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , ARN/química , ARN/metabolismo , Animales , Ratones , Unión Proteica , Dominios Proteicos
11.
Molecules ; 22(7)2017 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-28753936

RESUMEN

Musashi-1 (Msi1) controls the maintenance of stem cells and tumorigenesis through binding to its target mRNAs and subsequent translational regulation. Msi1 has two RNA-binding domains (RBDs), RBD1 and RBD2, which recognize r(GUAG) and r(UAG), respectively. These minimal recognition sequences are connected by variable linkers in the Msi1 target mRNAs, however, the molecular mechanism by which Msi1 recognizes its targets is not yet understood. We previously determined the solution structure of the Msi1 RBD1:r(GUAGU) complex. Here, we determined the first structure of the RBD2:r(GUAGU) complex. The structure revealed that the central trinucleotide, r(UAG), is specifically recognized by the intermolecular hydrogen-bonding and aromatic stacking interactions. Importantly, the C-terminal region, which is disordered in the free form, took a certain conformation, resembling a helix. The observation of chemical shift perturbation and intermolecular NOEs, together with increases in the heteronuclear steady-state {¹H}-15N NOE values on complex formation, indicated the involvement of the C-terminal region in RNA binding. On the basis of the two complex structures, we built a structural model of consecutive RBDs with r(UAGGUAG) containing both minimal recognition sequences, which resulted in no steric hindrance. The model suggests recognition of variable lengths (n) of the linker up to n = 50 may be possible.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Enlace de Hidrógeno , Ratones , Unión Proteica , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo
12.
Protein Sci ; 26(2): 280-291, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27862552

RESUMEN

The spliceosomal protein SF3b49, a component of the splicing factor 3b (SF3b) protein complex in the U2 small nuclear ribonucleoprotein, contains two RNA recognition motif (RRM) domains. In yeast, the first RRM domain (RRM1) of Hsh49 protein (yeast orthologue of human SF3b49) reportedly interacts with another component, Cus1 protein (orthologue of human SF3b145). Here, we solved the solution structure of the RRM1 of human SF3b49 and examined its mode of interaction with a fragment of human SF3b145 using NMR methods. Chemical shift mapping showed that the SF3b145 fragment spanning residues 598-631 interacts with SF3b49 RRM1, which adopts a canonical RRM fold with a topology of ß1-α1-ß2-ß3-α2-ß4. Furthermore, a docking model based on NOESY measurements suggests that residues 607-616 of the SF3b145 fragment adopt a helical structure that binds to RRM1 predominantly via α1, consequently exhibiting a helix-helix interaction in almost antiparallel. This mode of interaction was confirmed by a mutational analysis using GST pull-down assays. Comparison with structures of all RRM domains when complexed with a peptide found that this helix-helix interaction is unique to SF3b49 RRM1. Additionally, all amino acid residues involved in the interaction are well conserved among eukaryotes, suggesting evolutionary conservation of this interaction mode between SF3b49 RRM1 and SF3b145.


Asunto(s)
Simulación del Acoplamiento Molecular , Pliegue de Proteína , Factores de Empalme de ARN/química , Secuencias de Aminoácidos , Humanos , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Dominios Proteicos , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo
13.
Genes Dev ; 29(15): 1649-60, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26215567

RESUMEN

The pre-mRNA splicing reaction of eukaryotic cells has to be carried out extremely accurately, as failure to recognize the splice sites correctly causes serious disease. The small subunit of the U2AF heterodimer is essential for the determination of 3' splice sites in pre-mRNA splicing, and several single-residue mutations of the U2AF small subunit cause severe disorders such as myelodysplastic syndromes. However, the mechanism of RNA recognition is poorly understood. Here we solved the crystal structure of the U2AF small subunit (U2AF23) from fission yeast, consisting of an RNA recognition motif (RRM) domain flanked by two conserved CCCH-type zinc fingers (ZFs). The two ZFs are positioned side by side on the ß sheet of the RRM domain. Further mutational analysis revealed that the ZFs bind cooperatively to the target RNA sequence, but the RRM domain acts simply as a scaffold to organize the ZFs and does not itself contact the RNA directly. This completely novel and unexpected mode of RNA-binding mechanism by the U2AF small subunit sheds light on splicing errors caused by mutations of this highly conserved protein.


Asunto(s)
Modelos Moleculares , Proteínas Nucleares/química , Sitios de Empalme de ARN , Ribonucleoproteínas/química , Schizosaccharomyces/fisiología , Dedos de Zinc/fisiología , Secuencias de Aminoácidos , Sitios de Unión , Análisis Mutacional de ADN , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Schizosaccharomyces/química , Factor de Empalme U2AF
14.
J Struct Funct Genomics ; 16(2): 55-65, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25801860

RESUMEN

ZFAT is a transcriptional regulator, containing eighteen C2H2-type zinc-fingers and one AT-hook, involved in autoimmune thyroid disease, apoptosis, and immune-related cell survival. We determined the solution structures of the thirteen individual ZFAT zinc-fingers (ZF) and the tandemly arrayed zinc-fingers in the regions from ZF2 to ZF5, by NMR spectroscopy. ZFAT has eight uncommon bulged-out helix-containing zinc-fingers, and six of their structures (ZF4, ZF5, ZF6, ZF10, ZF11, and ZF13) were determined. The distribution patterns of the putative DNA-binding surface residues are different among the ZFAT zinc-fingers, suggesting the distinct DNA sequence preferences of the N-terminal and C-terminal zinc-fingers. Since ZFAT has three to five consecutive tandem zinc-fingers, which may cooperatively function as a unit, we also determined two tandemly arrayed zinc-finger structures, between ZF2 to ZF4 and ZF3 to ZF5. Our NMR spectroscopic analysis detected the interaction between ZF4 and ZF5, which are connected by an uncommon linker sequence, KKIK. The ZF4-ZF5 linker restrained the relative structural space between the two zinc-fingers in solution, unlike the other linker regions with determined structures, suggesting the involvement of the ZF4-ZF5 interfinger linker in the regulation of ZFAT function.


Asunto(s)
Proteínas de Unión al ADN/química , Conformación Proteica , Factores de Transcripción/química , Dedos de Zinc/genética , Secuencia de Aminoácidos/genética , Animales , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Humanos , Ratones , Resonancia Magnética Nuclear Biomolecular , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Tiroiditis Autoinmune/genética , Factores de Transcripción/metabolismo , Transcripción Genética
15.
Chembiochem ; 16(1): 167-76, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25469677

RESUMEN

We developed fluorescent turn-on probes containing a fluorescent nucleoside, 5-(benzofuran-2-yl)deoxyuridine (dU(BF)) or 5-(3-methylbenzofuran-2-yl)deoxyuridine (dU(MBF)), for the detection of single-stranded DNA or RNA by utilizing DNA triplex formation. Fluorescence measurements revealed that the probe containing dU(MBF) achieved superior fluorescence enhancement than that containing dU(BF). NMR and fluorescence analyses indicated that the fluorescence intensity increased upon triplex formation partly as a consequence of a conformational change at the bond between the 3-methylbenzofuran and uracil rings. In addition, it is suggested that the microenvironment around the 3-methylbenzofuran ring contributed to the fluorescence enhancement. Further, we developed a method for detecting RNA by rolling circular amplification in combination with triplex-induced fluorescence enhancement of the oligonucleotide probe containing dU(MBF).


Asunto(s)
Benzofuranos/química , ADN/química , Desoxiuridina/química , Oligonucleótidos/química , Uracilo/química , Secuencia de Bases , ADN de Cadena Simple/química , Desoxiuridina/análogos & derivados , Colorantes Fluorescentes/química , Modelos Moleculares , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , ARN/química , Espectrometría de Fluorescencia
16.
FEBS Open Bio ; 4: 689-703, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25161877

RESUMEN

Anaphase-promoting complex or cyclosome (APC/C) is a multisubunit ubiquitin ligase E3 that targets cell-cycle regulators. Cdc20 is required for full activation of APC/C in M phase, and mediates substrate recognition. In vertebrates, Emi2/Erp1/FBXO43 inhibits APC/C-Cdc20, and functions as a cytostatic factor that causes long-term M phase arrest of mature oocytes. In this study, we found that a fragment corresponding to the zinc-binding region (ZBR) domain of Emi2 inhibits cell-cycle progression, and impairs the association of Cdc20 with the APC/C core complex in HEK293T cells. Furthermore, we revealed that the ZBR fragment of Emi2 inhibits in vitro ubiquitin chain elongation catalyzed by the APC/C cullin-RING ligase module, the ANAPC2-ANAPC11 subcomplex, in combination with the ubiquitin chain-initiating E2, E2C/UBE2C/UbcH10. Structural analyses revealed that the Emi2 ZBR domain uses different faces for the two mechanisms. Thus, the double-faced ZBR domain of Emi2 antagonizes the APC/C function by inhibiting both the binding with the coactivator Cdc20 and ubiquitylation mediated by the cullin-RING ligase module and E2C. In addition, the tail region between the ZBR domain and the C-terminal RL residues [the post-ZBR (PZ) region] interacts with the cullin subunit, ANAPC2. In the case of the ZBR fragment of the somatic paralogue of Emi2, Emi1/FBXO5, these inhibitory activities against cell division and ubiquitylation were not observed. Finally, we identified two sets of key residues in the Emi2 ZBR domain that selectively exert each of the dual Emi2-specific modes of APC/C inhibition, by their mutation in the Emi2 ZBR domain and their transplantation into the Emi1 ZBR domain.

17.
Nat Struct Mol Biol ; 21(9): 778-86, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25132178

RESUMEN

Tissue-specific alternative pre-mRNA splicing is often cooperatively regulated by multiple splicing factors, but the structural basis of cooperative RNA recognition is poorly understood. In Caenorhabditis elegans, ligand binding specificity of fibroblast growth factor receptors (FGFRs) is determined by mutually exclusive alternative splicing of the sole FGFR gene, egl-15. Here we determined the solution structure of a ternary complex of the RNA-recognition motif (RRM) domains from the RBFOX protein ASD-1, SUP-12 and their target RNA from egl-15. The two RRM domains cooperatively interact with the RNA by sandwiching a G base to form the stable complex. Multichromatic fluorescence splicing reporters confirmed the requirement of the G and the juxtaposition of the respective cis elements for effective splicing regulation in vivo. Moreover, we identified a new target for the heterologous complex through an element search, confirming the functional significance of the intermolecular coordination.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Empalme Alternativo , Animales , Secuencia de Bases , Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/química , Cristalografía por Rayos X , Modelos Moleculares , Estructura Terciaria de Proteína , ARN/química , Proteínas de Unión al ARN/química
18.
Proteins ; 82(10): 2879-86, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25066254

RESUMEN

The family of cytoplasmic polyadenylation element binding proteins CPEB1, CPEB2, CPEB3, and CPEB4 binds to the 3'-untranslated region (3'-UTR) of mRNA, and plays significant roles in mRNA metabolism and translation regulation. They have a common domain organization, involving two consecutive RNA recognition motif (RRM) domains followed by a zinc finger domain in the C-terminal region. We solved the solution structure of the first RRM domain (RRM1) of human CPEB3, which revealed that CPEB3 RRM1 exhibits structural features distinct from those of the canonical RRM domain. Our structural data provide important information about the RNA binding ability of CPEB3 RRM1.


Asunto(s)
Modelos Moleculares , Fragmentos de Péptidos/química , Proteínas de Unión al ARN/química , Regiones no Traducidas 3' , Secuencia de Aminoácidos , Sitios de Unión , Secuencia Conservada , Bases de Datos de Proteínas , Humanos , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estabilidad Proteica , Estructura Secundaria de Proteína , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Solubilidad
19.
Biomol NMR Assign ; 7(1): 69-72, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22446849

RESUMEN

RNA helicase A (RHA) is a multifunctional protein that regulates gene expression. RHA has two double-stranded RNA-binding domains (dsRBDs) that serve as modules for highly structured RNA binding and protein-protein interactions. Using the dsRBDs, RHA binds to cellular and viral mRNAs, exports them from the nucleus, and regulates splicing as well as translational initiation. The RHA dsRBDs also reportedly mediate interactions with small RNAs and other dsRBD-containing proteins, and altogether form a processing complex involved in RNA silencing pathways. In addition, the RHA dsRBDs bridge RNA polymerase II with several transcription factors. Here we report the (1)H, (13)C, and (15)N chemical shift assignments of the dsRBDs of RHA. The resonance assignments obtained in this work will contribute to the elucidation of the interactions between RHA and transcriptional or post-transcriptional gene regulators.


Asunto(s)
Resonancia Magnética Nuclear Biomolecular , ARN Helicasas/química , ARN Helicasas/metabolismo , ARN Bicatenario/metabolismo , Animales , Ratones , Estructura Terciaria de Proteína
20.
FEBS Lett ; 586(21): 3858-64, 2012 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-23010590

RESUMEN

The WWE domain is often identified in proteins associated with ubiquitination or poly-ADP-ribosylation. Structural information about WWE domains has been obtained for the ubiquitination-related proteins, such as Deltex and RNF146, but not yet for the poly-ADP-ribose polymerases (PARPs). Here we determined the solution structures of the WWE domains from PARP11 and PARP14, and compared them with that of the RNF146 WWE domain. NMR perturbation experiments revealed the specific differences in their ADP-ribose recognition modes that correlated with their individual biological activities. The present structural information sheds light on the ADP-ribose recognition modes by the PARP WWE domains.


Asunto(s)
Adenosina Difosfato Ribosa/química , Poli(ADP-Ribosa) Polimerasas/síntesis química , Ubiquitina-Proteína Ligasas/síntesis química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Simulación por Computador , Humanos , Espectroscopía de Resonancia Magnética , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Poli(ADP-Ribosa) Polimerasas/química , Unión Proteica , Estructura Terciaria de Proteína , Alineación de Secuencia , Ubiquitina-Proteína Ligasas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...