Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Intervalo de año de publicación
1.
Nucleic Acids Res ; 52(10): 6036-6048, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38709891

RESUMEN

Nonsense-mediated mRNA decay (NMD) is a conserved co-translational mRNA surveillance and turnover pathway across eukaryotes. NMD has a central role in degrading defective mRNAs and also regulates the stability of a significant portion of the transcriptome. The pathway is organized around UPF1, an RNA helicase that can interact with several NMD-specific factors. In human cells, degradation of the targeted mRNAs begins with a cleavage event that requires the recruitment of the SMG6 endonuclease to UPF1. Previous studies have identified functional links between SMG6 and UPF1, but the underlying molecular mechanisms have remained elusive. Here, we used mass spectrometry, structural biology and biochemical approaches to identify and characterize a conserved short linear motif in SMG6 that interacts with the cysteine/histidine-rich (CH) domain of UPF1. Unexpectedly, we found that the UPF1-SMG6 interaction is precluded when the UPF1 CH domain is engaged with another NMD factor, UPF2. Based on cryo-EM data, we propose that the formation of distinct SMG6-containing and UPF2-containing NMD complexes may be dictated by different conformational states connected to the RNA-binding status of UPF1. Our findings rationalize a key event in metazoan NMD and advance our understanding of mechanisms regulating activity and guiding substrate recognition by the SMG6 endonuclease.


Asunto(s)
Endonucleasas , Degradación de ARNm Mediada por Codón sin Sentido , ARN Helicasas , Proteínas de Unión al ARN , Transactivadores , Humanos , Microscopía por Crioelectrón , Endonucleasas/metabolismo , Endonucleasas/genética , Endorribonucleasas , Modelos Moleculares , Unión Proteica , ARN Helicasas/metabolismo , ARN Helicasas/genética , ARN Helicasas/química , ARN Mensajero/metabolismo , ARN Mensajero/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/química , Transactivadores/metabolismo , Transactivadores/genética , Transactivadores/química , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Motivos de Unión al ARN
2.
Elife ; 122024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38517938

RESUMEN

Cyclic nucleotide binding domains (CNB) confer allosteric regulation by cAMP or cGMP to many signaling proteins, including PKA and PKG. PKA of phylogenetically distant Trypanosoma is the first exception as it is cyclic nucleotide-independent and responsive to nucleoside analogues (Bachmaier et al., 2019). Here, we show that natural nucleosides inosine, guanosine and adenosine are nanomolar affinity CNB ligands and activators of PKA orthologs of the important tropical pathogens Trypanosoma brucei, Trypanosoma cruzi, and Leishmania. The sequence and structural determinants of binding affinity, -specificity and kinase activation of PKAR were established by structure-activity relationship (SAR) analysis, co-crystal structures and mutagenesis. Substitution of two to three amino acids in the binding sites is sufficient for conversion of CNB domains from nucleoside to cyclic nucleotide specificity. In addition, a trypanosomatid-specific C-terminal helix (αD) is required for high affinity binding to CNB-B. The αD helix functions as a lid of the binding site that shields ligands from solvent. Selectivity of guanosine for CNB-B and of adenosine for CNB-A results in synergistic kinase activation at low nanomolar concentration. PKA pulldown from rapid lysis establishes guanosine as the predominant ligand in vivo in T. brucei bloodstream forms, whereas guanosine and adenosine seem to synergize in the procyclic developmental stage in the insect vector. We discuss the versatile use of CNB domains in evolution and recruitment of PKA for novel nucleoside-mediated signaling.


Asunto(s)
AMP Cíclico , Nucleósidos de Purina , AMP Cíclico/metabolismo , Nucleósidos/farmacología , Regulación Alostérica , Nucleótidos Cíclicos , Guanosina , Adenosina
3.
Life Sci Alliance ; 6(9)2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37468166

RESUMEN

Fun30 is the prototype of the Fun30-SMARCAD1-ETL subfamily of nucleosome remodelers involved in DNA repair and gene silencing. These proteins appear to act as single-subunit nucleosome remodelers, but their molecular mechanisms are, at this point, poorly understood. Using multiple sequence alignment and structure prediction, we identify an evolutionarily conserved domain that is modeled to contain a SAM-like fold with one long, protruding helix, which we term SAM-key. Deletion of the SAM-key within budding yeast Fun30 leads to a defect in DNA repair and gene silencing similar to that of the fun30Δ mutant. In vitro, Fun30 protein lacking the SAM-key is able to bind nucleosomes but is deficient in DNA-stimulated ATPase activity and nucleosome sliding and eviction. A structural model based on AlphaFold2 prediction and verified by crosslinking-MS indicates an interaction of the long SAM-key helix with protrusion I, a subdomain located between the two ATPase lobes that is critical for control of enzymatic activity. Mutation of the interaction interface phenocopies the domain deletion with a lack of DNA-stimulated ATPase activation and a nucleosome-remodeling defect, thereby confirming a role of the SAM-key helix in regulating ATPase activity. Our data thereby demonstrate a central role of the SAM-key domain in mediating the activation of Fun30 catalytic activity, thus highlighting the importance of allosteric activation for this class of enzymes.


Asunto(s)
Nucleosomas , Proteínas de Saccharomyces cerevisiae , Nucleosomas/genética , Nucleosomas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , ADN/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo
4.
Cell Rep ; 42(1): 111902, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36586408

RESUMEN

The evolutionary conserved CCR4-NOT complex functions in the cytoplasm as the main mRNA deadenylase in both constitutive mRNA turnover and regulated mRNA decay pathways. The versatility of this complex is underpinned by its modular multi-subunit organization, with distinct structural modules actuating different functions. The structure and function of all modules are known, except for that of the N-terminal module. Using different structural approaches, we obtained high-resolution data revealing the architecture of the human N-terminal module composed of CNOT1, CNOT10, and CNOT11. The structure shows how two helical domains of CNOT1 sandwich CNOT10 and CNOT11, leaving the most conserved domain of CNOT11 protruding into solvent as an antenna. We discovered that GGNBP2, a protein identified as a tumor suppressor and spermatogenic factor, is a conserved interacting partner of the CNOT11 antenna domain. Structural and biochemical analyses thus pinpoint the N-terminal CNOT1-CNOT10-CNOT11 module as a conserved protein-protein interaction platform.


Asunto(s)
Factores de Transcripción , Humanos , Factores de Transcripción/metabolismo , Unión Proteica
5.
EMBO J ; 40(15): e107807, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34191293

RESUMEN

Eukaryotic cells employ three SMC (structural maintenance of chromosomes) complexes to control DNA folding and topology. The Smc5/6 complex plays roles in DNA repair and in preventing the accumulation of deleterious DNA junctions. To elucidate how specific features of Smc5/6 govern these functions, we reconstituted the yeast holo-complex. We found that the Nse5/6 sub-complex strongly inhibited the Smc5/6 ATPase by preventing productive ATP binding. This inhibition was relieved by plasmid DNA binding but not by short linear DNA, while opposing effects were observed without Nse5/6. We uncovered two binding sites for Nse5/6 on Smc5/6, based on an Nse5/6 crystal structure and cross-linking mass spectrometry data. One binding site is located at the Smc5/6 arms and one at the heads, the latter likely exerting inhibitory effects on ATP hydrolysis. Cysteine cross-linking demonstrated that the interaction with Nse5/6 anchored the ATPase domains in a non-productive state, which was destabilized by ATP and DNA. Under similar conditions, the Nse4/3/1 module detached from the ATPase. Altogether, we show how DNA substrate selection is modulated by direct inhibition of the Smc5/6 ATPase by Nse5/6.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Adenosina Trifosfato/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Microscopía por Crioelectrón , Cristalografía por Rayos X , ADN de Hongos/metabolismo , Hidrólisis , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Conformación Proteica , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
6.
Elife ; 92020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-33191913

RESUMEN

The yeast THO complex is recruited to active genes and interacts with the RNA-dependent ATPase Sub2 to facilitate the formation of mature export-competent messenger ribonucleoprotein particles and to prevent the co-transcriptional formation of RNA:DNA-hybrid-containing structures. How THO-containing complexes function at the mechanistic level is unclear. Here, we elucidated a 3.4 Å resolution structure of Saccharomyces cerevisiae THO-Sub2 by cryo-electron microscopy. THO subunits Tho2 and Hpr1 intertwine to form a platform that is bound by Mft1, Thp2, and Tex1. The resulting complex homodimerizes in an asymmetric fashion, with a Sub2 molecule attached to each protomer. The homodimerization interfaces serve as a fulcrum for a seesaw-like movement concomitant with conformational changes of the Sub2 ATPase. The overall structural architecture and topology suggest the molecular mechanisms of nucleic acid remodeling during mRNA biogenesis.


Asunto(s)
Adenosina Trifosfatasas/química , Proteínas de Saccharomyces cerevisiae/química , Factores de Transcripción/química , Adenosina Trifosfatasas/metabolismo , Microscopía por Crioelectrón , Ácidos Nucleicos/química , Ácidos Nucleicos/metabolismo , Conformación Proteica , Ribonucleoproteínas/química , Ribonucleoproteínas/metabolismo , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/metabolismo
7.
Protein Sci ; 29(6): 1502-1510, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32239748

RESUMEN

Motile cilia protrude from cell surfaces and are necessary to create movement of cells and fluids in the body. At the molecular level, cilia contain several dynein molecular motor complexes including outer dynein arms (ODAs) that are attached periodically to the ciliary axoneme, where they hydrolyse ATP to create the force required for bending and motility of the cilium. ODAs are preassembled in the cytoplasm and subsequently trafficked into the cilium by the intraflagellar transport (IFT) system. In the case of the green alga Chlamydomonas reinhardtii, the adaptor protein ODA16 binds to ODAs and directly to the IFT complex component IFT46 to facilitate the ciliary import of ODAs. Here, we purified recombinant human IFT46 and ODA16, determined the high-resolution crystal structure of the ODA16 protein, and carried out direct interaction studies of IFT46 and ODA16. The human ODA16 C-terminal 320 residues adopt the fold of an eight-bladed ß-propeller with high overall structural similarity to the Chlamydomonas ODA16. However, the small 80 residue N-terminal domain, which in Chlamydomonas ODA16 is located on top of the ß-propeller and is required to form the binding cleft for IFT46, has no visible electron density in case of the human ODA16 structure. Furthermore, size exclusion chromatography and pull-down experiments failed to detect a direct interaction between human ODA16 and IFT46. These data suggest that additional factors may be required for the ciliary import of ODAs in human cells with motile cilia.


Asunto(s)
Cilios/metabolismo , Dineínas/metabolismo , Proteínas Recombinantes/metabolismo , Chlamydomonas reinhardtii/química , Chlamydomonas reinhardtii/metabolismo , Cilios/química , Cristalografía por Rayos X , Dineínas/química , Dineínas/aislamiento & purificación , Humanos , Modelos Moleculares , Conformación Proteica , Transporte de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación
8.
Nat Commun ; 10(1): 1421, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30926779

RESUMEN

Protein kinase A (PKA), the main effector of cAMP in eukaryotes, is a paradigm for the mechanisms of ligand-dependent and allosteric regulation in signalling. Here we report the orthologous but cAMP-independent PKA of the protozoan Trypanosoma and identify 7-deaza-nucleosides as potent activators (EC50 ≥ 6.5 nM) and high affinity ligands (KD ≥ 8 nM). A co-crystal structure of trypanosome PKA with 7-cyano-7-deazainosine and molecular docking show how substitution of key amino acids in both CNB domains of the regulatory subunit and its unique C-terminal αD helix account for this ligand swap between trypanosome PKA and canonical cAMP-dependent PKAs. We propose nucleoside-related endogenous activators of Trypanosoma brucei PKA (TbPKA). The existence of eukaryotic CNB domains not associated with binding of cyclic nucleotides suggests that orphan CNB domains in other eukaryotes may bind undiscovered signalling molecules. Phosphoproteome analysis validates 7-cyano-7-deazainosine as powerful cell-permeable inducer to explore cAMP-independent PKA signalling in medically important neglected pathogens.


Asunto(s)
Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Activadores de Enzimas/farmacología , Nucleósidos/análogos & derivados , Trypanosoma brucei brucei/enzimología , Secuencia de Aminoácidos , Cristalografía por Rayos X , AMP Cíclico/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/química , Dipiridamol/farmacología , Evaluación Preclínica de Medicamentos , Activadores de Enzimas/química , Holoenzimas/metabolismo , Leishmania/efectos de los fármacos , Simulación del Acoplamiento Molecular , Fosforilación/efectos de los fármacos , Transducción de Señal , Trypanosoma brucei brucei/efectos de los fármacos , Tubercidina/farmacología
9.
Nature ; 557(7707): 734-738, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29795347

RESUMEN

Conventional ubiquitination regulates key cellular processes by catalysing the ATP-dependent formation of an isopeptide bond between ubiquitin (Ub) and primary amines in substrate proteins 1 . Recently, the SidE family of bacterial effector proteins (SdeA, SdeB, SdeC and SidE) from pathogenic Legionella pneumophila were shown to use NAD+ to mediate phosphoribosyl-linked ubiquitination of serine residues in host proteins2, 3. However, the molecular architecture of the catalytic platform that enables this complex multistep process remains unknown. Here we describe the structure of the catalytic core of SdeA, comprising mono-ADP-ribosyltransferase (mART) and phosphodiesterase (PDE) domains, and shed light on the activity of two distinct catalytic sites for serine ubiquitination. The mART catalytic site is composed of an α-helical lobe (AHL) that, together with the mART core, creates a chamber for NAD+ binding and ADP-ribosylation of ubiquitin. The catalytic site in the PDE domain cleaves ADP-ribosylated ubiquitin to phosphoribosyl ubiquitin (PR-Ub) and mediates a two-step PR-Ub transfer reaction: first to a catalytic histidine 277 (forming a transient SdeA H277-PR-Ub intermediate) and subsequently to a serine residue in host proteins. Structural analysis revealed a substrate binding cleft in the PDE domain, juxtaposed with the catalytic site, that is essential for positioning serines for ubiquitination. Using degenerate substrate peptides and newly identified ubiquitination sites in RTN4B, we show that disordered polypeptides with hydrophobic residues surrounding the target serine residues are preferred substrates for SdeA ubiquitination. Infection studies with L. pneumophila expressing substrate-binding mutants of SdeA revealed that substrate ubiquitination, rather than modification of the cellular ubiquitin pool, determines the pathophysiological effect of SdeA during acute bacterial infection.


Asunto(s)
Biocatálisis , Legionella pneumophila/enzimología , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Serina/metabolismo , Ubiquitinación , ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/metabolismo , Adenosina Difosfato/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Legionella pneumophila/genética , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/microbiología , Proteínas de la Membrana/genética , Modelos Moleculares , Hidrolasas Diéster Fosfóricas/química , Hidrolasas Diéster Fosfóricas/metabolismo , Estructura Secundaria de Proteína , Especificidad por Sustrato , Ubiquitina/metabolismo
10.
Mol Cell ; 67(2): 334-347.e5, 2017 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-28689660

RESUMEN

Multi-subunit SMC complexes control chromosome superstructure and promote chromosome disjunction, conceivably by actively translocating along DNA double helices. SMC subunits comprise an ABC ATPase "head" and a "hinge" dimerization domain connected by a 49 nm coiled-coil "arm." The heads undergo ATP-dependent engagement and disengagement to drive SMC action on the chromosome. Here, we elucidate the architecture of prokaryotic Smc dimers by high-throughput cysteine cross-linking and crystallography. Co-alignment of the Smc arms tightly closes the interarm space and misaligns the Smc head domains at the end of the rod by close apposition of their ABC signature motifs. Sandwiching of ATP molecules between Smc heads requires them to substantially tilt and translate relative to each other, thereby opening up the Smc arms. We show that this mechanochemical gating reaction regulates chromosome targeting and propose a mechanism for DNA translocation based on the merging of DNA loops upon closure of Smc arms.


Asunto(s)
Bacillus subtilis/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Segregación Cromosómica , Cromosomas Bacterianos , Adenosina Trifosfato/metabolismo , Bacillus subtilis/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Cristalografía por Rayos X , Cisteína , Ensayos Analíticos de Alto Rendimiento , Modelos Moleculares , Mutación , Conformación de Ácido Nucleico , Conformación Proteica , Multimerización de Proteína , Estabilidad Proteica , Relación Estructura-Actividad
11.
J Biol Chem ; 292(18): 7462-7473, 2017 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28298440

RESUMEN

Motile cilia are found on unicellular organisms such as the green alga Chlamydomonas reinhardtii, on sperm cells, and on cells that line the trachea and fallopian tubes in mammals. The motility of cilia relies on a number of large protein complexes including the force-generating outer dynein arms (ODAs). The transport of ODAs into cilia has been previously shown to require the transport adaptor ODA16, as well as the intraflagellar transport (IFT) protein IFT46, but the molecular mechanism by which ODAs are recognized and transported into motile cilia is still unclear. Here, we determined the high-resolution crystal structure of C. reinhardtii ODA16 (CrODA16) and mapped the binding to IFT46 and ODAs. The CrODA16 structure revealed a small 80-residue N-terminal domain and a C-terminal 8-bladed ß-propeller domain that are both required for the association with the N-terminal 147 residues of IFT46. The dissociation constant of the IFT46-ODA16 complex was 200 nm, demonstrating that CrODA16 associates with the IFT complex with an affinity comparable with that of the individual IFT subunits. Furthermore, we show, using ODAs extracted from the axonemes of C. reinhardtii, that the C-terminal ß-propeller but not the N-terminal domain of CrODA16 is required for the interaction with ODAs. These data allowed us to present an architectural model for ODA16-mediated IFT of ODAs.


Asunto(s)
Proteínas Portadoras , Chlamydomonas reinhardtii , Dineínas , Flagelos , Proteínas de Plantas , Axonema/química , Axonema/genética , Axonema/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Chlamydomonas reinhardtii/química , Chlamydomonas reinhardtii/genética , Chlamydomonas reinhardtii/metabolismo , Cristalografía por Rayos X , Dineínas/química , Dineínas/genética , Dineínas/metabolismo , Flagelos/química , Flagelos/genética , Flagelos/metabolismo , Proteínas de Plantas/química , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Dominios Proteicos , Transporte de Proteínas/fisiología
12.
Cell Rep ; 13(4): 703-711, 2015 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-26489469

RESUMEN

The DEAD-box protein DDX6 is a central component of translational repression mechanisms in maternal mRNA storage in oocytes and microRNA-mediated silencing in somatic cells. DDX6 interacts with the CCR4-NOT complex and functions in concert with several post-transcriptional regulators, including Edc3, Pat1, and 4E-T. We show that the conserved CUP-homology domain (CHD) of human 4E-T interacts directly with DDX6 in both the presence and absence of the central MIF4G domain of CNOT1. The 2.1-Å resolution structure of the corresponding ternary complex reveals how 4E-T CHD wraps around the RecA2 domain of DDX6 and contacts CNOT1. Although 4E-T CHD lacks recognizable sequence similarity with Edc3 or Pat1, it shares the same DDX6-binding surface. In contrast to 4E-T, however, the Edc3 and Pat1 FDF motifs dissociate from DDX6 upon CNOT1 MIF4G binding in vitro. The results underscore the presence of a complex network of simultaneous and/or mutually exclusive interactions in DDX6-mediated repression.


Asunto(s)
Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , ARN Helicasas DEAD-box/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/química , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores CCR4/metabolismo , Factores de Transcripción/metabolismo , Sitios de Unión/genética , Sitios de Unión/fisiología , Proteínas Portadoras/genética , ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/genética , Humanos , Modelos Biológicos , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Proteínas de Transporte Nucleocitoplasmático/química , Proteínas de Transporte Nucleocitoplasmático/genética , Unión Proteica/genética , Unión Proteica/fisiología , Estructura Secundaria de Proteína , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética , Receptores CCR4/química , Receptores CCR4/genética , Factores de Transcripción/química , Factores de Transcripción/genética
13.
Mol Cell ; 54(5): 751-65, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24768538

RESUMEN

MicroRNAs (miRNAs) control gene expression by regulating mRNA translation and stability. The CCR4-NOT complex is a key effector of miRNA function acting downstream of GW182/TNRC6 proteins. We show that miRNA-mediated repression requires the central region of CNOT1, the scaffold protein of CCR4-NOT. A CNOT1 domain interacts with CNOT9, which in turn interacts with the silencing domain of TNRC6 in a tryptophan motif-dependent manner. These interactions are direct, as shown by the structure of a CNOT9-CNOT1 complex with bound tryptophan. Another domain of CNOT1 with an MIF4G fold recruits the DEAD-box ATPase DDX6, a known translational inhibitor. Structural and biochemical approaches revealed that CNOT1 modulates the conformation of DDX6 and stimulates ATPase activity. Structure-based mutations showed that the CNOT1 MIF4G-DDX6 interaction is important for miRNA-mediated repression. These findings provide insights into the repressive steps downstream of the GW182/TNRC6 proteins and the role of the CCR4-NOT complex in posttranscriptional regulation in general.


Asunto(s)
ARN Helicasas DEAD-box/química , MicroARNs/genética , Proteínas Proto-Oncogénicas/química , Interferencia de ARN , Factores de Transcripción/química , Sustitución de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Complejos Multiproteicos/química , Mutagénesis Sitio-Dirigida , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
Nat Struct Mol Biol ; 20(3): 371-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23353789

RESUMEN

Eukaryotic structural maintenance of chromosomes (SMC)-kleisin complexes form large, ring-shaped assemblies that promote accurate chromosome segregation. Their asymmetric structural core comprises SMC heterodimers that associate with both ends of a kleisin subunit. However, prokaryotic condensin Smc-ScpAB is composed of symmetric Smc homodimers associated with the kleisin ScpA in a postulated symmetrical manner. Here, we demonstrate that Smc molecules have two distinct binding sites for ScpA. The N terminus of ScpA binds the Smc coiled coil, whereas the C terminus binds the Smc ATPase domain. We show that in Bacillus subtilis cells, an Smc dimer is bridged by a single ScpAB to generate asymmetric tripartite rings analogous to eukaryotic SMC complexes. We define a molecular mechanism that ensures asymmetric assembly, and we conclude that the basic architecture of SMC-kleisin rings evolved before the emergence of eukaryotes.


Asunto(s)
Adenosina Trifosfatasas/química , Proteínas Bacterianas/química , Proteínas de Ciclo Celular/química , Proteínas de Unión al ADN/química , Complejos Multiproteicos/química , Adenosina Trifosfatasas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Reactivos de Enlaces Cruzados , Cristalografía por Rayos X , Proteínas de Unión al ADN/metabolismo , Modelos Moleculares , Complejos Multiproteicos/metabolismo , Mutación , Conformación Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Streptococcus pneumoniae/química
15.
Nat Struct Mol Biol ; 14(7): 620-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17589525

RESUMEN

The U2AF-homology motif (UHM) mediates protein-protein interactions between factors involved in constitutive RNA splicing. Here we report that the splicing factor SPF45 regulates alternative splicing of the apoptosis regulatory gene FAS (also called CD95). The SPF45 UHM is necessary for this activity and binds UHM-ligand motifs (ULMs) present in the 3' splice site-recognizing factors U2AF65, SF1 and SF3b155. We describe a 2.1-A crystal structure of SPF45-UHM in complex with a ULM peptide from SF3b155. Features distinct from those of previously described UHM-ULM structures allowed the design of mutations in the SPF45 UHM that selectively impair binding to individual ULMs. Splicing assays using the ULM-selective SPF45 variants demonstrate that individual UHM-ULM interactions are required for FAS splicing regulation by SPF45 in vivo. Our data suggest that networks of UHM-ULM interactions are involved in regulating alternative splicing.


Asunto(s)
Empalme Alternativo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Receptor fas/genética , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Cristalografía por Rayos X , Exones , Humanos , Datos de Secuencia Molecular , Mutación , Proteínas Nucleares/química , Péptidos/química , Fosfoproteínas/química , Conformación Proteica , Mapeo de Interacción de Proteínas , Factores de Empalme de ARN , Proteínas de Unión al ARN/genética , Ribonucleoproteína Nuclear Pequeña U2/química , Ribonucleoproteínas/química , Factor de Empalme U2AF
16.
Nucleic Acids Res ; 31(14): 4091-8, 2003 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12853626

RESUMEN

The Hfq protein was discovered in Escherichia coli in the early seventies as a host factor for the Qbeta phage RNA replication. During the last decade, it was shown to be involved in many RNA processing events and remote sequence homology indicated a link to spliceosomal Sm proteins. We report the crystal structure of the E.coli Hfq protein showing that its monomer displays a characteristic Sm-fold and forms a homo-hexamer, in agreement with former biochemical data. Overall, the structure of the E.coli Hfq ring is similar to the one recently described for Staphylococcus aureus. This confirms that bacteria contain a hexameric Sm-like protein which is likely to be an ancient and less specialized form characterized by a relaxed RNA binding specificity. In addition, we identified an Hfq ortholog in the archaeon Methanococcus jannaschii which lacks a classical Sm/Lsm gene. Finally, a detailed structural comparison shows that the Sm-fold is remarkably well conserved in bacteria, Archaea and Eukarya, and represents a universal and modular building unit for oligomeric RNA binding proteins.


Asunto(s)
Bacterias/genética , Proteínas de Escherichia coli/genética , Proteína de Factor 1 del Huésped/genética , Ribonucleoproteínas Nucleares Pequeñas/genética , Secuencia de Aminoácidos , Autoantígenos , Cristalografía por Rayos X , Dimerización , Proteínas de Escherichia coli/química , Evolución Molecular , Proteína de Factor 1 del Huésped/química , Datos de Secuencia Molecular , Conformación Proteica , Estructura Secundaria de Proteína , Ribonucleoproteínas Nucleares Pequeñas/química , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Proteínas Nucleares snRNP
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA