Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Elife ; 122024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38415718

RESUMEN

Sirtuin 6 (SIRT6) is an NAD+-dependent histone H3 deacetylase that is prominently found associated with chromatin, attenuates transcriptionally active promoters and regulates DNA repair, metabolic homeostasis and lifespan. Unlike other sirtuins, it has low affinity to free histone tails but demonstrates strong binding to nucleosomes. It is poorly understood how SIRT6 docking on nucleosomes stimulates its histone deacetylation activity. Here, we present the structure of human SIRT6 bound to a nucleosome determined by cryogenic electron microscopy. The zinc finger domain of SIRT6 associates tightly with the acidic patch of the nucleosome through multiple arginine anchors. The Rossmann fold domain binds to the terminus of the looser DNA half of the nucleosome, detaching two turns of the DNA from the histone octamer and placing the NAD+ binding pocket close to the DNA exit site. This domain shows flexibility with respect to the fixed zinc finger and moves with, but also relative to, the unwrapped DNA terminus. We apply molecular dynamics simulations of the histone tails in the nucleosome to show that in this mode of interaction, the active site of SIRT6 is perfectly poised to catalyze deacetylation of the H3 histone tail and that the partial unwrapping of the DNA allows even lysines close to the H3 core to reach the enzyme.


Asunto(s)
Nucleosomas , Sirtuinas , Humanos , Histonas , NAD , Cromatina , Glicosiltransferasas , Histona Desacetilasas , ADN
2.
Nat Struct Mol Biol ; 30(9): 1337-1345, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37550452

RESUMEN

Histone acetylation regulates most DNA transactions and is dynamically controlled by highly conserved enzymes. The only essential histone acetyltransferase (HAT) in yeast, Esa1, is part of the 1-MDa NuA4 complex, which plays pivotal roles in both transcription and DNA-damage repair. NuA4 has the unique capacity to acetylate histone targets located several nucleosomes away from its recruitment site. Neither the molecular mechanism of this activity nor its physiological importance are known. Here we report the structure of the Pichia pastoris NuA4 complex, with its core resolved at 3.4-Å resolution. Three subunits, Epl1, Eaf1 and Swc4, intertwine to form a stable platform that coordinates all other modules. The HAT module is firmly anchored into the core while retaining the ability to stretch out over a long distance. We provide structural, biochemical and genetic evidence that an unfolded linker region of the Epl1 subunit is critical for this long-range activity. Specifically, shortening the Epl1 linker causes severe growth defects and reduced H4 acetylation levels over broad chromatin regions in fission yeast. Our work lays the foundations for a mechanistic understanding of NuA4's regulatory role and elucidates how its essential long-range activity is attained.


Asunto(s)
Histonas , Proteínas de Saccharomyces cerevisiae , Histonas/genética , Histonas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Cromatina , Nucleosomas , Saccharomyces cerevisiae/metabolismo , Histona Acetiltransferasas/metabolismo , ADN , Acetilación
3.
Nat Commun ; 13(1): 1546, 2022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-35318334

RESUMEN

RNA polymerase (RNAP) frequently pauses during the transcription of DNA to RNA to regulate gene expression. Transcription factors NusA and NusG modulate pausing, have opposing roles, but can bind RNAP simultaneously. Here we report cryo-EM reconstructions of Escherichia coli RNAP bound to NusG, or NusA, or both. RNAP conformational changes, referred to as swivelling, correlate with transcriptional pausing. NusA facilitates RNAP swivelling to further increase pausing, while NusG counteracts this role. Their structural effects are consistent with biochemical results on two categories of transcriptional pauses. In addition, the structures suggest a cooperative mechanism of NusA and NusG during Rho-mediated transcription termination. Our results provide a structural rationale for the stochastic nature of pausing and termination and how NusA and NusG can modulate it.


Asunto(s)
Proteínas de Escherichia coli , Factores de Transcripción , Proteínas Bacterianas/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Conformación de Ácido Nucleico , Factores de Elongación de Péptidos/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Factores de Elongación Transcripcional/metabolismo
4.
C R Biol ; 343(3): 247-255, 2021 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-33621454

RESUMEN

The transcription of eukaryotic protein genes is controlled by a plethora of proteins which act together in multi-component complexes to facilitate the DNA dependent RNA polymerase II (Pol II) enzyme to bind to the transcription start site and to generate messenger RNA from the gene's coding sequence. The protein that guides the transcription machinery to the exact transcription start site is called TATA-box Binding Protein, or TBP. TBP is part of two large protein complexes involved in Pol II transcription, TFIID and SAGA. The two complexes share several subunits implicated in the interaction with TBP and contain proteins with structural elements highly homologous to nucleosomal histones. Despite the intensive study of transcription initiation, the mode of interaction of TBP with these complexes and its release upon DNA binding was elusive. In this study we demonstrate the quasi-atomic model of SAGA in complex with TBP. The structure reveals the intricate network of interactions that coordinate the different functional domains of SAGA and resolves a deformed octamer of histone-fold domains at the core of SAGA. This deformed octamer is precisely tuned to establish a peripheral site for TBP binding, where it is protected by steric hindrance against the binding of spurious DNA. Complementary biochemical analysis points to a mechanism for TBP delivery and release from SAGA that requires the general transcription factor TFIIA and whose efficiency correlates with the affinity of DNA to TBP.As the TBP binding machinery is highly similar in TFIID and SAGA, we demonstrated a universal mechanism of how TBP is delivered to gene promoters during transcription initiation.


La transcription des gènes des protéines eucaryotes est contrôlée par une pléthore de protéines agissant de concert sous forme de complexes multi-composants pour faciliter la liaison de l'enzyme ARN polymérase II ADN-dépendante (Pol II) au site d'initiation de la transcription et pour générer un ARN messager à partir de la séquence codante du gène. La protéine qui guide la machinerie de transcription vers le site d'initiation de la transcription est appelée protéine de liaison à la boîte TATA, ou TBP. TBP fait partie de deux complexes protéiques impliqués dans la transcription par la Pol II, TFIID et SAGA. Les deux complexes partagent plusieurs sous-unités impliquées dans l'interaction avec TBP et comportent des protéines présentant des éléments structuraux hautement homologues aux histones nucléosomiques. Malgré l'étude intensive de l'initiation de la transcription, le mode d'interaction de TBP avec ces complexes ainsi que sa libération lors de sa liaison de l'ADN étaient évasifs. Dans cette étude, nous avons déterminé un modèle quasi-atomique de SAGA en complexe avec TBP. La structure révèle le réseau d'interactions qui coordonnent les différents domaines fonctionnels de SAGA et résout un octamère déformé des domaines homologues aux histones au cœur de SAGA. Cet octamère déformé est précisément adapté pour établir un site périphérique de liaison à TBP, où ce dernier est protégé par une inhibition stérique contre la fixation d'un ADN parasite. L'analyse biochimique complémentaire a mis en évidence un mécanisme de libération de TBP de SAGA qui nécessite le facteur de transcription général TFIIA et dont l'efficacité corrèle avec l'affinité de l'ADN pour TBP.Comme le mécanisme de liaison de TBP est très similaire dans TFIID et SAGA, nous avons mis en évidence un mécanisme universel décrivant la manière dont TBP est délivré aux promoteurs de gènes lors de l'initiation de la transcription.


Asunto(s)
Transactivadores , Factores de Transcripción , Regiones Promotoras Genéticas , Proteína de Unión a TATA-Box/genética , Proteína de Unión a TATA-Box/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/genética , Transcripción Genética
5.
FEBS J ; 288(10): 3135-3147, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32946670

RESUMEN

In eukaryotes, transcription of protein encoding genes is initiated by the controlled deposition of the TATA-box binding protein TBP onto gene promoters, followed by the ordered assembly of a pre-initiation complex. The SAGA co-activator is a 19-subunit complex that stimulates transcription by the action of two chromatin-modifying enzymatic modules, a transcription activator binding module, and by delivering TBP. Recent cryo electron microscopy structures of yeast SAGA with bound nucleosome or TBP reveal the architecture of the different functional domains of the co-activator. An octamer of histone fold domains is found at the core of SAGA. This octamer, which deviates considerably from the symmetrical analogue forming the nucleosome, establishes a peripheral site for TBP binding where steric hindrance represses interaction with spurious DNA. The structures point to a mechanism for TBP delivery and release from SAGA that requires TFIIA and whose efficiency correlates with the affinity of DNA to TBP. These results provide a structural basis for understanding specific TBP delivery onto gene promoters and the role played by SAGA in regulating gene expression. The properties of the TBP delivery machine harboured by SAGA are compared with the TBP loading device present in the TFIID complex and show multiple similitudes.


Asunto(s)
Regulación de la Expresión Génica , Nucleosomas , Saccharomyces cerevisiae/genética , TATA Box , Factor de Transcripción TFIID/genética , Transcripción Genética , Sitios de Unión , Secuencia Conservada , Histonas/genética , Histonas/metabolismo , Humanos , Regiones Promotoras Genéticas , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Saccharomyces cerevisiae/metabolismo , Transducción de Señal , Proteína de Unión a TATA-Box/genética , Proteína de Unión a TATA-Box/metabolismo , Transactivadores/metabolismo , Factor de Transcripción TFIID/metabolismo
6.
Biochim Biophys Acta Gene Regul Mech ; 1864(2): 194614, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32739556

RESUMEN

Transcription initiation is a major regulatory step in eukaryotic gene expression. It involves the assembly of general transcription factors and RNA polymerase II into a functional pre-initiation complex at core promoters. The degree of chromatin compaction controls the accessibility of the transcription machinery to template DNA. Co-activators have critical roles in this process by actively regulating chromatin accessibility. Many transcriptional coactivators are multisubunit complexes, organized into distinct structural and functional modules and carrying multiple regulatory activities. The first nuclear histone acetyltransferase (HAT) characterized was General Control Non-derepressible 5 (Gcn5). Gcn5 was subsequently identified as a subunit of the HAT module of the Spt-Ada-Gcn5-acetyltransferase (SAGA) complex, which is an experimental paradigm for multifunctional co-activators. We know today that Gcn5 is the catalytic subunit of multiple distinct co-activator complexes with specific functions. In this review, we summarize recent advances in the structure of Gcn5-containing co-activator complexes, most notably SAGA, and discuss how these new structural insights contribute to better understand their functions.


Asunto(s)
Regulación de la Expresión Génica , Complejos Multienzimáticos/metabolismo , Estructura Cuaternaria de Proteína/fisiología , Transactivadores/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Secuencia de Aminoácidos/genética , Animales , Arabidopsis/enzimología , Arabidopsis/genética , Secuencia Conservada , Microscopía por Crioelectrón , Cristalografía , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Evolución Molecular , Histonas/metabolismo , Humanos , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/ultraestructura , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Relación Estructura-Actividad , Transactivadores/genética , Transactivadores/ultraestructura , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/ultraestructura
7.
Nature ; 577(7792): 711-716, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31969704

RESUMEN

SAGA (Spt-Ada-Gcn5-acetyltransferase) is a 19-subunit complex that stimulates transcription via two chromatin-modifying enzymatic modules and by delivering the TATA box binding protein (TBP) to nucleate the pre-initiation complex on DNA, a pivotal event in the expression of protein-encoding genes1. Here we present the structure of yeast SAGA with bound TBP. The core of the complex is resolved at 3.5 Å resolution (0.143 Fourier shell correlation). The structure reveals the intricate network of interactions that coordinate the different functional domains of SAGA and resolves an octamer of histone-fold domains at the core of SAGA. This deformed octamer deviates considerably from the symmetrical analogue in the nucleosome and is precisely tuned to establish a peripheral site for TBP, where steric hindrance represses binding of spurious DNA. Complementary biochemical analysis points to a mechanism for TBP delivery and release from SAGA that requires transcription factor IIA and whose efficiency correlates with the affinity of DNA to TBP. We provide the foundations for understanding the specific delivery of TBP to gene promoters and the multiple roles of SAGA in regulating gene expression.


Asunto(s)
Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Pichia , Regiones Promotoras Genéticas/genética , Proteína de Unión a TATA-Box/metabolismo , Transactivadores/química , Transactivadores/metabolismo , Sitios de Unión , ADN de Hongos/química , ADN de Hongos/metabolismo , Regulación Fúngica de la Expresión Génica , Histona Acetiltransferasas/química , Histona Acetiltransferasas/metabolismo , Histonas/química , Histonas/metabolismo , Modelos Moleculares , Pichia/química , Pichia/genética , Unión Proteica , Conformación Proteica , Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores Asociados con la Proteína de Unión a TATA/química , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Proteína de Unión a TATA-Box/química , Factor de Transcripción TFIIA/química , Factor de Transcripción TFIIA/metabolismo , Factor de Transcripción TFIID/química , Factor de Transcripción TFIID/metabolismo
8.
Mol Cell ; 75(2): 298-309.e4, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31103420

RESUMEN

Regulatory sequences or erroneous incorporations during DNA transcription cause RNA polymerase backtracking and inactivation in all kingdoms of life. Reactivation requires RNA transcript cleavage. Essential transcription factors (GreA and GreB, or TFIIS) accelerate this reaction. We report four cryo-EM reconstructions of Escherichia coli RNA polymerase representing the entire reaction pathway: (1) a backtracked complex; a backtracked complex with GreB (2) before and (3) after RNA cleavage; and (4) a reactivated, substrate-bound complex with GreB before RNA extension. Compared with eukaryotes, the backtracked RNA adopts a different conformation. RNA polymerase conformational changes cause distinct GreB states: a fully engaged GreB before cleavage; a disengaged GreB after cleavage; and a dislodged, loosely bound GreB removed from the active site to allow RNA extension. These reconstructions provide insight into the catalytic mechanism and dynamics of RNA cleavage and extension and suggest how GreB targets backtracked complexes without interfering with canonical transcription.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/química , Proteínas de Escherichia coli/química , Complejos Multiproteicos/química , ARN/química , Transcripción Genética , Factores de Elongación Transcripcional/química , Secuencia de Aminoácidos/genética , Dominio Catalítico/genética , Microscopía por Crioelectrón , ARN Polimerasas Dirigidas por ADN/genética , Escherichia coli/química , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Complejos Multiproteicos/genética , Unión Proteica , Conformación Proteica , ARN/genética , División del ARN/genética , Motivos de Unión al ARN/genética , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Elongación Transcripcional/genética
9.
Nat Commun ; 9(1): 4666, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30405110

RESUMEN

Transcription preinitiation complex assembly on the promoters of protein encoding genes is nucleated in vivo by TFIID composed of the TATA-box Binding Protein (TBP) and 13 TBP-associate factors (Tafs) providing regulatory and chromatin binding functions. Here we present the cryo-electron microscopy structure of promoter-bound yeast TFIID at a resolution better than 5 Å, except for a flexible domain. We position the crystal structures of several subunits and, in combination with cross-linking studies, describe the quaternary organization of TFIID. The compact tri lobed architecture is stabilized by a topologically closed Taf5-Taf6 tetramer. We confirm the unique subunit stoichiometry prevailing in TFIID and uncover a hexameric arrangement of Tafs containing a histone fold domain in the Twin lobe.


Asunto(s)
Regiones Promotoras Genéticas/genética , Factor de Transcripción TFIID/química , Factor de Transcripción TFIID/metabolismo , Levaduras/metabolismo , ADN de Hongos/química , ADN de Hongos/metabolismo , Modelos Moleculares , Unión Proteica , Subunidades de Proteína/química , Subunidades de Proteína/aislamiento & purificación , Subunidades de Proteína/metabolismo , Saccharomyces cerevisiae/metabolismo , Factor de Transcripción TFIID/aislamiento & purificación
10.
Elife ; 72018 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-30460895

RESUMEN

The biogenesis of 60S ribosomal subunits is initiated in the nucleus where rRNAs and proteins form pre-60S particles. These pre-60S particles mature by transiently interacting with various assembly factors. The ~5000 amino-acid AAA+ ATPase Rea1 (or Midasin) generates force to mechanically remove assembly factors from pre-60S particles, which promotes their export to the cytosol. Here we present three Rea1 cryoEM structures. We visualise the Rea1 engine, a hexameric ring of AAA+ domains, and identify an α-helical bundle of AAA2 as a major ATPase activity regulator. The α-helical bundle interferes with nucleotide-induced conformational changes that create a docking site for the substrate binding MIDAS domain on the AAA +ring. Furthermore, we reveal the architecture of the Rea1 linker, which is involved in force generation and extends from the AAA+ ring. The data presented here provide insights into the mechanism of one of the most complex ribosome maturation factors.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/química , Adenosina Trifosfato/química , ARN Ribosómico/química , Proteínas Ribosómicas/química , Subunidades Ribosómicas Grandes de Eucariotas/genética , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , ATPasas Asociadas con Actividades Celulares Diversas/genética , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Adenosina Difosfato/química , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Sitios de Unión , Fenómenos Biomecánicos , Clonación Molecular , Microscopía por Crioelectrón , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Cinética , Modelos Moleculares , Biogénesis de Organelos , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , ARN de Hongos/química , ARN de Hongos/metabolismo , ARN Ribosómico/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/enzimología , Subunidades Ribosómicas Grandes de Eucariotas/ultraestructura , Saccharomyces cerevisiae/enzimología , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidad por Sustrato
11.
Mol Cell ; 69(5): 816-827.e4, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29499136

RESUMEN

Transcriptional pausing by RNA polymerases (RNAPs) is a key mechanism to regulate gene expression in all kingdoms of life and is a prerequisite for transcription termination. The essential bacterial transcription factor NusA stimulates both pausing and termination of transcription, thus playing a central role. Here, we report single-particle electron cryo-microscopy reconstructions of NusA bound to paused E. coli RNAP elongation complexes with and without a pause-enhancing hairpin in the RNA exit channel. The structures reveal four interactions between NusA and RNAP that suggest how NusA stimulates RNA folding, pausing, and termination. An asymmetric translocation intermediate of RNA and DNA converts the active site of the enzyme into an inactive state, providing a structural explanation for the inhibition of catalysis. Comparing RNAP at different stages of pausing provides insights on the dynamic nature of the process and the role of NusA as a regulatory factor.


Asunto(s)
ARN Polimerasas Dirigidas por ADN , Proteínas de Escherichia coli , Escherichia coli , Pliegue del ARN , ARN Bacteriano , Terminación de la Transcripción Genética , Factores de Elongación Transcripcional , Dominio Catalítico , ADN Bacteriano/química , ADN Bacteriano/metabolismo , ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/metabolismo , Escherichia coli/química , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , ARN Bacteriano/biosíntesis , ARN Bacteriano/química , Factores de Elongación Transcripcional/química , Factores de Elongación Transcripcional/metabolismo
12.
J Struct Biol ; 202(3): 191-199, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29337113

RESUMEN

A current bottleneck in structure determination of macromolecular complexes by cryo electron microscopy (cryo-EM) is the large amount of data needed to obtain high-resolution 3D reconstructions, including through sorting into different conformations and compositions with advanced image processing. Additionally, it may be difficult to visualize small ligands that bind in sub-stoichiometric levels. Volta phase plates (VPP) introduce a phase shift in the contrast transfer and drastically increase the contrast of the recorded low-dose cryo-EM images while preserving high frequency information. Here we present a comparative study to address the behavior of different data sets during image processing and quantify important parameters during structure refinement. The automated data collection was done from the same human ribosome sample either as a conventional defocus range dataset or with a Volta phase plate close to focus (cfVPP) or with a small defocus (dfVPP). The analysis of image processing parameters shows that dfVPP data behave more robustly during cryo-EM structure refinement because particle alignments, Euler angle assignments and 2D & 3D classifications behave more stably and converge faster. In particular, less particle images are required to reach the same resolution in the 3D reconstructions. Finally, we find that defocus range data collection is also applicable to VPP. This study shows that data processing and cryo-EM map interpretation, including atomic model refinement, are facilitated significantly by performing VPP cryo-EM, which will have an important impact on structural biology.


Asunto(s)
Microscopía por Crioelectrón/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Sustancias Macromoleculares/química , Recolección de Datos , Humanos , Ligandos , Sustancias Macromoleculares/ultraestructura , Ribosomas/química , Ribosomas/ultraestructura
13.
Nat Commun ; 8(1): 1556, 2017 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-29146944

RESUMEN

The transcription co-activator complex SAGA is recruited to gene promoters by sequence-specific transcriptional activators and by chromatin modifications to promote pre-initiation complex formation. The yeast Tra1 subunit is the major target of acidic activators such as Gal4, VP16, or Gcn4 but little is known about its structural organization. The 430 kDa Tra1 subunit and its human homolog the transformation/transcription domain-associated protein TRRAP are members of the phosphatidyl 3-kinase-related kinase (PIKK) family. Here, we present the cryo-EM structure of the entire SAGA complex where the major target of activator binding, the 430 kDa Tra1 protein, is resolved with an average resolution of 5.7 Å. The high content of alpha-helices in Tra1 enabled tracing of the majority of its main chain. Our results highlight the integration of Tra1 within the major epigenetic regulator SAGA.


Asunto(s)
Cromatina/metabolismo , Proteínas Fúngicas/metabolismo , Histona Acetiltransferasas/metabolismo , Transactivadores/metabolismo , Secuencia de Aminoácidos , Cromatina/química , Cromatina/ultraestructura , Microscopía por Crioelectrón , Proteínas Fúngicas/química , Proteínas Fúngicas/ultraestructura , Histona Acetiltransferasas/química , Histona Acetiltransferasas/ultraestructura , Humanos , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Saccharomycetales/química , Saccharomycetales/metabolismo , Homología de Secuencia de Aminoácido , Transactivadores/química , Transactivadores/ultraestructura
15.
Mol Cell ; 66(3): 384-397.e8, 2017 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-28475873

RESUMEN

Linker histones associate with nucleosomes to promote the formation of higher-order chromatin structure, but the underlying molecular details are unclear. We investigated the structure of a 197 bp nucleosome bearing symmetric 25 bp linker DNA arms in complex with vertebrate linker histone H1. We determined electron cryo-microscopy (cryo-EM) and crystal structures of unbound and H1-bound nucleosomes and validated these structures by site-directed protein cross-linking and hydroxyl radical footprinting experiments. Histone H1 shifts the conformational landscape of the nucleosome by drawing the two linkers together and reducing their flexibility. The H1 C-terminal domain (CTD) localizes primarily to a single linker, while the H1 globular domain contacts the nucleosome dyad and both linkers, associating more closely with the CTD-distal linker. These findings reveal that H1 imparts a strong degree of asymmetry to the nucleosome, which is likely to influence the assembly and architecture of higher-order structures.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , ADN/metabolismo , Histonas/metabolismo , Nucleosomas/metabolismo , Animales , Emparejamiento Base , Sitios de Unión , Cromatina/química , Cromatina/genética , Cromatina/ultraestructura , Microscopía por Crioelectrón , ADN/química , ADN/genética , Histonas/química , Humanos , Modelos Moleculares , Nucleosomas/química , Nucleosomas/genética , Nucleosomas/ultraestructura , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad , Factores de Tiempo , Xenopus laevis/genética , Xenopus laevis/metabolismo
16.
Sci Rep ; 6: 38399, 2016 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-27924919

RESUMEN

The conserved SecYEG protein-conducting channel and the accessory proteins SecDF-YajC and YidC constitute the bacterial holo-translocon (HTL), capable of protein-secretion and membrane-protein insertion. By employing an integrative approach combining small-angle neutron scattering (SANS), low-resolution electron microscopy and biophysical analyses we determined the arrangement of the proteins and lipids within the super-complex. The results guided the placement of X-ray structures of individual HTL components and allowed the proposal of a model of the functional translocon. Their arrangement around a central lipid-containing pool conveys an unexpected, but compelling mechanism for membrane-protein insertion. The periplasmic domains of YidC and SecD are poised at the protein-channel exit-site of SecY, presumably to aid the emergence of translocating polypeptides. The SecY lateral gate for membrane-insertion is adjacent to the membrane 'insertase' YidC. Absolute-scale SANS employing a novel contrast-match-point analysis revealed a dynamic complex adopting open and compact configurations around an adaptable central lipid-filled chamber, wherein polytopic membrane-proteins could fold, sheltered from aggregation and proteolysis.


Asunto(s)
Proteínas de Escherichia coli/química , Escherichia coli/química , Proteínas de Transporte de Membrana/química , Canales de Translocación SEC/química , Sitios de Unión , Clonación Molecular , Microscopía por Crioelectrón , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Methanocaldococcus/química , Methanocaldococcus/genética , Methanocaldococcus/metabolismo , Modelos Moleculares , Difracción de Neutrones , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Canales de Translocación SEC/genética , Canales de Translocación SEC/metabolismo , Dispersión del Ángulo Pequeño , Homología Estructural de Proteína , Especificidad por Sustrato , Thermus thermophilus/química , Thermus thermophilus/genética , Thermus thermophilus/metabolismo
17.
Mol Cell ; 63(4): 674-685, 2016 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-27499292

RESUMEN

CENP-A is a histone variant, which replaces histone H3 at centromeres and confers unique properties to centromeric chromatin. The crystal structure of CENP-A nucleosome suggests flexible nucleosomal DNA ends, but their dynamics in solution remains elusive and their implication in centromere function is unknown. Using electron cryo-microscopy, we determined the dynamic solution properties of the CENP-A nucleosome. Our biochemical, proteomic, and genetic data reveal that higher flexibility of DNA ends impairs histone H1 binding to the CENP-A nucleosome. Substituting the 2-turn αN-helix of CENP-A with the 3-turn αN-helix of H3 results in compact particles with rigidified DNA ends, able to bind histone H1. In vivo replacement of CENP-A with H3-CENP-A hybrid nucleosomes leads to H1 recruitment, delocalization of kinetochore proteins, and significant mitotic and cytokinesis defects. Our data reveal that the evolutionarily conserved flexible ends of the CENP-A nucleosomes are essential to ensure the fidelity of the mitotic pathway.


Asunto(s)
Autoantígenos/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , ADN/metabolismo , Histonas/metabolismo , Cinetocoros/metabolismo , Mitosis/fisiología , Nucleosomas/metabolismo , Animales , Autoantígenos/genética , Autoantígenos/ultraestructura , Sitios de Unión , Proteína A Centromérica , Proteínas Cromosómicas no Histona/deficiencia , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/ultraestructura , Microscopía por Crioelectrón , Citocinesis , ADN/química , Genotipo , Células HeLa , Humanos , Cinetocoros/ultraestructura , Ratones , Ratones Noqueados , Modelos Moleculares , Mutación , Conformación de Ácido Nucleico , Nucleosomas/ultraestructura , Fenotipo , Unión Proteica , Conformación Proteica en Hélice alfa , Relación Estructura-Actividad , Transfección
18.
Nat Commun ; 7: 12126, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27418187

RESUMEN

Eukaryotic RNA polymerase I (Pol I) is specialized in rRNA gene transcription synthesizing up to 60% of cellular RNA. High level rRNA production relies on efficient binding of initiation factors to the rRNA gene promoter and recruitment of Pol I complexes containing initiation factor Rrn3. Here, we determine the cryo-EM structure of the Pol I-Rrn3 complex at 7.5 Å resolution, and compare it with Rrn3-free monomeric and dimeric Pol I. We observe that Rrn3 contacts the Pol I A43/A14 stalk and subunits A190 and AC40, that association re-organizes the Rrn3 interaction interface, thereby preventing Pol I dimerization; and Rrn3-bound and monomeric Pol I differ from the dimeric enzyme in cleft opening, and localization of the A12.2 C-terminus in the active centre. Our findings thus support a dual role for Rrn3 in transcription initiation to stabilize a monomeric initiation competent Pol I and to drive pre-initiation complex formation.


Asunto(s)
Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , ARN Polimerasa I/química , ARN Polimerasa I/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Microscopía por Crioelectrón/métodos , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , Regiones Promotoras Genéticas , Dominios Proteicos , Multimerización de Proteína , ARN Polimerasa I/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Transcripción Genética
19.
Nat Commun ; 6: 6011, 2015 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-25586196

RESUMEN

General transcription factor TFIID is a cornerstone of RNA polymerase II transcription initiation in eukaryotic cells. How human TFIID-a megadalton-sized multiprotein complex composed of the TATA-binding protein (TBP) and 13 TBP-associated factors (TAFs)-assembles into a functional transcription factor is poorly understood. Here we describe a heterotrimeric TFIID subcomplex consisting of the TAF2, TAF8 and TAF10 proteins, which assembles in the cytoplasm. Using native mass spectrometry, we define the interactions between the TAFs and uncover a central role for TAF8 in nucleating the complex. X-ray crystallography reveals a non-canonical arrangement of the TAF8-TAF10 histone fold domains. TAF2 binds to multiple motifs within the TAF8 C-terminal region, and these interactions dictate TAF2 incorporation into a core-TFIID complex that exists in the nucleus. Our results provide evidence for a stepwise assembly pathway of nuclear holo-TFIID, regulated by nuclear import of preformed cytoplasmic submodules.


Asunto(s)
Citoplasma/metabolismo , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factor de Transcripción TFIID/química , Secuencias de Aminoácidos , Calorimetría , Núcleo Celular/metabolismo , Cristalografía por Rayos X , Células HeLa , Histonas/química , Humanos , Espectrometría de Masas/métodos , Modelos Moleculares , Unión Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo , Resonancia por Plasmón de Superficie , Factor de Transcripción TFIID/metabolismo , Factores de Transcripción/metabolismo
20.
Nature ; 493(7434): 699-702, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23292512

RESUMEN

The initiation of gene transcription by RNA polymerase II is regulated by a plethora of proteins in human cells. The first general transcription factor to bind gene promoters is transcription factor IID (TFIID). TFIID triggers pre-initiation complex formation, functions as a coactivator by interacting with transcriptional activators and reads epigenetic marks. TFIID is a megadalton-sized multiprotein complex composed of TATA-box-binding protein (TBP) and 13 TBP-associated factors (TAFs). Despite its crucial role, the detailed architecture and assembly mechanism of TFIID remain elusive. Histone fold domains are prevalent in TAFs, and histone-like tetramer and octamer structures have been proposed in TFIID. A functional core-TFIID subcomplex was revealed in Drosophila nuclei, consisting of a subset of TAFs (TAF4, TAF5, TAF6, TAF9 and TAF12). These core subunits are thought to be present in two copies in holo-TFIID, in contrast to TBP and other TAFs that are present in a single copy, conveying a transition from symmetry to asymmetry in the TFIID assembly pathway. Here we present the structure of human core-TFIID determined by cryo-electron microscopy at 11.6 Å resolution. Our structure reveals a two-fold symmetric, interlaced architecture, with pronounced protrusions, that accommodates all conserved structural features of the TAFs including the histone folds. We further demonstrate that binding of one TAF8-TAF10 complex breaks the original symmetry of core-TFIID. We propose that the resulting asymmetric structure serves as a functional scaffold to nucleate holo-TFIID assembly, by accreting one copy each of the remaining TAFs and TBP.


Asunto(s)
Modelos Moleculares , Factor de Transcripción TFIID/química , Células Cultivadas , Microscopía por Crioelectrón , Células HeLa , Humanos , Unión Proteica , Estructura Terciaria de Proteína , Factor de Transcripción TFIID/genética , Factor de Transcripción TFIID/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...