Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 633(8029): 459-464, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39169181

RESUMEN

Chaperonins are large barrel-shaped complexes that mediate ATP-dependent protein folding1-3. The bacterial chaperonin GroEL forms juxtaposed rings that bind unfolded protein and the lid-shaped cofactor GroES at their apertures. In vitro analyses of the chaperonin reaction have shown that substrate protein folds, unimpaired by aggregation, while transiently encapsulated in the GroEL central cavity by GroES4-6. To determine the functional stoichiometry of GroEL, GroES and client protein in situ, here we visualized chaperonin complexes in their natural cellular environment using cryo-electron tomography. We find that, under various growth conditions, around 55-70% of GroEL binds GroES asymmetrically on one ring, with the remainder populating symmetrical complexes. Bound substrate protein is detected on the free ring of the asymmetrical complex, defining the substrate acceptor state. In situ analysis of GroEL-GroES chambers, validated by high-resolution structures obtained in vitro, showed the presence of encapsulated substrate protein in a folded state before release into the cytosol. Based on a comprehensive quantification and conformational analysis of chaperonin complexes, we propose a GroEL-GroES reaction cycle that consists of linked asymmetrical and symmetrical subreactions mediating protein folding. Our findings illuminate the native conformational and functional chaperonin cycle directly within cells.


Asunto(s)
Chaperonina 10 , Chaperonina 60 , Microscopía por Crioelectrón , Tomografía con Microscopio Electrónico , Proteínas de Escherichia coli , Escherichia coli , Sitios de Unión , Chaperonina 10/metabolismo , Chaperonina 10/química , Chaperonina 10/ultraestructura , Chaperonina 60/metabolismo , Chaperonina 60/química , Chaperonina 60/ultraestructura , Citosol/química , Citosol/metabolismo , Citosol/ultraestructura , Escherichia coli/química , Escherichia coli/citología , Escherichia coli/crecimiento & desarrollo , Escherichia coli/metabolismo , Escherichia coli/ultraestructura , Modelos Moleculares , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Reproducibilidad de los Resultados , Especificidad por Sustrato , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/ultraestructura
2.
Nature ; 628(8009): 894-900, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38600380

RESUMEN

Fractals are patterns that are self-similar across multiple length-scales1. Macroscopic fractals are common in nature2-4; however, so far, molecular assembly into fractals is restricted to synthetic systems5-12. Here we report the discovery of a natural protein, citrate synthase from the cyanobacterium Synechococcus elongatus, which self-assembles into Sierpinski triangles. Using cryo-electron microscopy, we reveal how the fractal assembles from a hexameric building block. Although different stimuli modulate the formation of fractal complexes and these complexes can regulate the enzymatic activity of citrate synthase in vitro, the fractal may not serve a physiological function in vivo. We use ancestral sequence reconstruction to retrace how the citrate synthase fractal evolved from non-fractal precursors, and the results suggest it may have emerged as a harmless evolutionary accident. Our findings expand the space of possible protein complexes and demonstrate that intricate and regulatable assemblies can evolve in a single substitution.


Asunto(s)
Citrato (si)-Sintasa , Evolución Molecular , Fractales , Multimerización de Proteína , Synechococcus , Microscopía por Crioelectrón , Modelos Moleculares , Synechococcus/enzimología , Citrato (si)-Sintasa/química , Citrato (si)-Sintasa/metabolismo , Citrato (si)-Sintasa/ultraestructura
3.
Proc Natl Acad Sci U S A ; 120(10): e2210891120, 2023 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-36857347

RESUMEN

SMAD-mediated signaling regulates apoptosis, cell cycle arrest, and epithelial-to-mesenchymal transition to safeguard tissue homeostasis. However, it remains elusive how the relatively simple pathway can determine such a broad range of cell fate decisions and how it differentiates between varying ligands. Here, we systematically investigate how SMAD-mediated responses are modulated by various ligands of the transforming growth factor ß (TGFß) family and compare these ligand responses in quiescent and proliferating MCF10A cells. We find that the nature of the phenotypic response is mainly determined by the proliferation status, with migration and cell cycle arrest being dominant in proliferating cells for all tested TGFß family ligands, whereas cell death is the major outcome in quiescent cells. In both quiescent and proliferating cells, the identity of the ligand modulates the strength of the phenotypic response proportional to the dynamics of induced SMAD nuclear-to-cytoplasmic translocation and, as a consequence, the corresponding gene expression changes. Interestingly, the proliferation state of a cell has little impact on the set of genes induced by SMAD signaling; instead, it modulates the relative cellular sensitivity to TGFß superfamily members. Taken together, diversity of SMAD-mediated responses is mediated by differing cellular states, which determine ligand sensitivity and phenotypic effects, while the pathway itself merely serves as a quantitative relay from the cell membrane to the nucleus.


Asunto(s)
Apoptosis , Transducción de Señal , Ligandos , Muerte Celular , Factor de Crecimiento Transformador beta
4.
J Struct Biol ; 213(3): 107750, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34089875

RESUMEN

Cetacean morbillivirus (CeMV) is an emerging and highly infectious paramyxovirus that causes outbreaks in cetaceans and occasionally in pinnipeds, representing a major threat to biodiversity and conservation of endangered marine mammal populations in both hemispheres. As for all non-segmented, negative-sense, single-stranded RNA (ssRNA) viruses, the morbilliviral genome is enwrapped by thousands of nucleoprotein (N) protomers. Each bound to six ribonucleotides, N protomers assemble to form a helical ribonucleoprotein (RNP) complex that serves as scaffold for nucleocapsid formation and as template for viral replication and transcription. While the molecular details on RNP complexes elucidated in human measles virus (MeV) served as paradigm model for these processes in all members of the Morbillivirus genus, no structural information has been obtained from other morbilliviruses, nor has any CeMV structure been solved so far. We report the structure of the CeMV RNP complex, reconstituted in vitro upon binding of recombinant CeMV N to poly-adenine ssRNA hexamers and solved to 4.0 Å resolution by cryo-electron microscopy. In spite of the amino acid sequence similarity and consequently similar folding of the N protomer, the CeMV RNP complex exhibits different helical parameters as compared to previously reported MeV orthologs. The CeMV structure reveals exclusive interactions leading to more extensive protomer-RNA and protomer-protomer interfaces. We identified twelve residues, among those varying between CeMV strains, as putatively important for the stabilization of the RNP complex, which highlights the need to study the potential of CeMV N mutations that modulate nucleocapsid assembly to also affect viral phenotype and host adaptation.


Asunto(s)
Infecciones por Morbillivirus , Morbillivirus , Animales , Microscopía por Crioelectrón , Mamíferos/genética , Morbillivirus/genética , Infecciones por Morbillivirus/epidemiología , Nucleoproteínas/genética , ARN Viral/química , ARN Viral/genética
5.
Biochem Biophys Res Commun ; 538: 54-62, 2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33039147

RESUMEN

Unprecedented by number of casualties and socio-economic burden occurring worldwide, the coronavirus disease 2019 (Covid-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the worst health crisis of this century. In order to develop adequate countermeasures against Covid-19, identification and structural characterization of suitable antiviral targets within the SARS-CoV-2 protein repertoire is urgently needed. The nucleocapsid phosphoprotein (N) is a multifunctional and highly immunogenic determinant of virulence and pathogenicity, whose main functions consist in oligomerizing and packaging the single-stranded RNA (ssRNA) viral genome. Here we report the structural and biophysical characterization of the SARS-CoV-2 N C-terminal domain (CTD), on which both N homo-oligomerization and ssRNA binding depend. Crystal structures solved at 1.44 Å and 1.36 Å resolution describe a rhombus-shape N CTD dimer, which stably exists in solution as validated by size-exclusion chromatography coupled to multi-angle light scattering and analytical ultracentrifugation. Differential scanning fluorimetry revealed moderate thermal stability and a tendency towards conformational change. Microscale thermophoresis demonstrated binding to a 7-bp SARS-CoV-2 genomic ssRNA fragment at micromolar affinity. Furthermore, a low-resolution preliminary model of the full-length SARS-CoV N in complex with ssRNA, obtained by cryo-electron microscopy, provides an initial understanding of self-associating and RNA binding functions exerted by the SARS-CoV-2 N.


Asunto(s)
COVID-19/virología , Proteínas de la Nucleocápside de Coronavirus/química , Proteínas de Unión al ARN/química , SARS-CoV-2/genética , Proteínas de la Nucleocápside de Coronavirus/genética , Microscopía por Crioelectrón , Genoma Viral , Humanos , Fosfoproteínas/química , Fosfoproteínas/genética , Unión Proteica , Dominios Proteicos , Multimerización de Proteína , Proteínas de Unión al ARN/genética
6.
Mol Syst Biol ; 14(1): e7733, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29371237

RESUMEN

The cytokine TGFß provides important information during embryonic development, adult tissue homeostasis, and regeneration. Alterations in the cellular response to TGFß are involved in severe human diseases. To understand how cells encode the extracellular input and transmit its information to elicit appropriate responses, we acquired quantitative time-resolved measurements of pathway activation at the single-cell level. We established dynamic time warping to quantitatively compare signaling dynamics of thousands of individual cells and described heterogeneous single-cell responses by mathematical modeling. Our combined experimental and theoretical study revealed that the response to a given dose of TGFß is determined cell specifically by the levels of defined signaling proteins. This heterogeneity in signaling protein expression leads to decomposition of cells into classes with qualitatively distinct signaling dynamics and phenotypic outcome. Negative feedback regulators promote heterogeneous signaling, as a SMAD7 knock-out specifically affected the signal duration in a subpopulation of cells. Taken together, we propose a quantitative framework that allows predicting and testing sources of cellular signaling heterogeneity.


Asunto(s)
Análisis de la Célula Individual/métodos , Proteína Smad2/metabolismo , Proteína Smad4/metabolismo , Biología de Sistemas/métodos , Factor de Crecimiento Transformador beta/farmacología , Línea Celular , Núcleo Celular/metabolismo , Humanos , Modelos Teóricos , Especificidad de Órganos , Transducción de Señal
7.
Nat Methods ; 9(9): 901-3, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22772729

RESUMEN

The mass spectrometric identification of chemically cross-linked peptides (CXMS) specifies spatial restraints of protein complexes; these values complement data obtained from common structure-determination techniques. Generic methods for determining false discovery rates of cross-linked peptide assignments are currently lacking, thus making data sets from CXMS studies inherently incomparable. Here we describe an automated target-decoy strategy and the software tool xProphet, which solve this problem for large multicomponent protein complexes.


Asunto(s)
Reactivos de Enlaces Cruzados/química , Espectrometría de Masas/métodos , Péptidos/análisis , Péptidos/química , Proteómica/métodos , Algoritmos , Automatización , Interpretación Estadística de Datos , Bases de Datos de Proteínas , Reacciones Falso Positivas , Modelos Moleculares , Conformación Proteica , Programas Informáticos
8.
Proc Natl Acad Sci U S A ; 109(5): 1479-84, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22215586

RESUMEN

Two canonical subunits of the 26S proteasome, Rpn10 and Rpn13, function as ubiquitin (Ub) receptors. The mutual arrangement of these subunits--and all other non-ATPase subunits--in the regulatory particle is unknown. Using electron cryomicroscopy, we calculated difference maps between wild-type 26S proteasome from Saccharomyces cerevisiae and deletion mutants (rpn10Δ, rpn13Δ, and rpn10Δrpn13Δ). These maps allowed us to localize the two Ub receptors unambiguously. Rpn10 and Rpn13 mapped to the apical part of the 26S proteasome, above the N-terminal coiled coils of the AAA-ATPase heterodimers Rpt4/Rpt5 and Rpt1/Rpt2, respectively. On the basis of the mutual positions of Rpn10 and Rpn13, we propose a model for polyubiquitin binding to the 26S proteasome.


Asunto(s)
Microscopía por Crioelectrón/métodos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Animales , Drosophila melanogaster , Espectrometría de Masas , Modelos Moleculares
9.
Proc Natl Acad Sci U S A ; 109(5): 1380-7, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22307589

RESUMEN

The 26S proteasome is at the executive end of the ubiquitin-proteasome pathway for the controlled degradation of intracellular proteins. While the structure of its 20S core particle (CP) has been determined by X-ray crystallography, the structure of the 19S regulatory particle (RP), which recruits substrates, unfolds them, and translocates them to the CP for degradation, has remained elusive. Here, we describe the molecular architecture of the 26S holocomplex determined by an integrative approach based on data from cryoelectron microscopy, X-ray crystallography, residue-specific chemical cross-linking, and several proteomics techniques. The "lid" of the RP (consisting of Rpn3/5/6/7/8/9/11/12) is organized in a modular fashion. Rpn3/5/6/7/9/12 form a horseshoe-shaped heterohexamer, which connects to the CP and roofs the AAA-ATPase module, positioning the Rpn8/Rpn11 heterodimer close to its mouth. Rpn2 is rigid, supporting the lid, while Rpn1 is conformationally variable, positioned at the periphery of the ATPase ring. The ubiquitin receptors Rpn10 and Rpn13 are located in the distal part of the RP, indicating that they were recruited to the complex late in its evolution. The modular structure of the 26S proteasome provides insights into the sequence of events prior to the degradation of ubiquitylated substrates.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Microscopía por Crioelectrón , Cristalografía por Rayos X , Espectrometría de Masas , Modelos Moleculares , Complejo de la Endopetidasa Proteasomal/química , Conformación Proteica , Proteómica , Schizosaccharomyces/enzimología , Especificidad por Sustrato
10.
Proc Natl Acad Sci U S A ; 109(37): 14870-5, 2012 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-22927375

RESUMEN

The 26S proteasome operates at the executive end of the ubiquitin-proteasome pathway. Here, we present a cryo-EM structure of the Saccharomyces cerevisiae 26S proteasome at a resolution of 7.4 Å or 6.7 Å (Fourier-Shell Correlation of 0.5 or 0.3, respectively). We used this map in conjunction with molecular dynamics-based flexible fitting to build a near-atomic resolution model of the holocomplex. The quality of the map allowed us to assign α-helices, the predominant secondary structure element of the regulatory particle subunits, throughout the entire map. We were able to determine the architecture of the Rpn8/Rpn11 heterodimer, which had hitherto remained elusive. The MPN domain of Rpn11 is positioned directly above the AAA-ATPase N-ring suggesting that Rpn11 deubiquitylates substrates immediately following commitment and prior to their unfolding by the AAA-ATPase module. The MPN domain of Rpn11 dimerizes with that of Rpn8 and the C-termini of both subunits form long helices, which are integral parts of a coiled-coil module. Together with the C-terminal helices of the six PCI-domain subunits they form a very large coiled-coil bundle, which appears to serve as a flexible anchoring device for all the lid subunits.


Asunto(s)
Endopeptidasas/química , Modelos Moleculares , Complejo de la Endopetidasa Proteasomal/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/química , Microscopía por Crioelectrón , Simulación de Dinámica Molecular , Estructura Terciaria de Proteína
11.
Proc Natl Acad Sci U S A ; 109(1): 149-54, 2012 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-22187461

RESUMEN

Proteasomes execute the degradation of most cellular proteins. Although the 20S core particle (CP) has been studied in great detail, the structure of the 19S regulatory particle (RP), which prepares ubiquitylated substrates for degradation, has remained elusive. Here, we report the crystal structure of one of the RP subunits, Rpn6, and we describe its integration into the cryo-EM density map of the 26S holocomplex at 9.1 Å resolution. Rpn6 consists of an α-solenoid-like fold and a proteasome COP9/signalosome eIF3 (PCI) module in a right-handed suprahelical configuration. Highly conserved surface areas of Rpn6 interact with the conserved surfaces of the Pre8 (alpha2) and Rpt6 subunits from the alpha and ATPase rings, respectively. The structure suggests that Rpn6 has a pivotal role in stabilizing the otherwise weak interaction between the CP and the RP.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Complejos Multiproteicos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Subunidades de Proteína/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Cristalografía por Rayos X , Proteínas de Drosophila/química , Modelos Moleculares , Datos de Secuencia Molecular , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/ultraestructura , Unión Proteica , Subunidades de Proteína/química , Schizosaccharomyces/enzimología , Soluciones , Propiedades de Superficie
12.
Mol Cell Proteomics ; 11(3): M111.014126, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22286754

RESUMEN

Chemical cross-linking in combination with mass spectrometric analysis offers the potential to obtain low-resolution structural information from proteins and protein complexes. Identification of peptides connected by a cross-link provides direct evidence for the physical interaction of amino acid side chains, information that can be used for computational modeling purposes. Despite impressive advances that were made in recent years, the number of experimentally observed cross-links still falls below the number of possible contacts of cross-linkable side chains within the span of the cross-linker. Here, we propose two complementary experimental strategies to expand cross-linking data sets. First, enrichment of cross-linked peptides by size exclusion chromatography selects cross-linked peptides based on their higher molecular mass, thereby depleting the majority of unmodified peptides present in proteolytic digests of cross-linked samples. Second, we demonstrate that the use of proteases in addition to trypsin, such as Asp-N, can additionally boost the number of observable cross-linking sites. The benefits of both SEC enrichment and multiprotease digests are demonstrated on a set of model proteins and the improved workflow is applied to the characterization of the 20S proteasome from rabbit and Schizosaccharomyces pombe.


Asunto(s)
Cromatografía en Gel , Reactivos de Enlaces Cruzados/farmacología , Péptido Hidrolasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas/metabolismo , Schizosaccharomyces/metabolismo , Animales , Bovinos , Cromatografía Liquida , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Conejos , Espectrometría de Masas en Tándem , Tripsina/farmacología
13.
bioRxiv ; 2024 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-39005358

RESUMEN

Many enzymes assemble into homomeric protein complexes comprising multiple copies of one protein. Because structural form is usually assumed to follow function in biochemistry, these assemblies are thought to evolve because they provide some functional advantage. In many cases, however, no specific advantage is known and, in some cases, quaternary structure varies among orthologs. This has led to the proposition that self-assembly may instead vary neutrally within protein families. The extent of such variation has been difficult to ascertain because quaternary structure has until recently been difficult to measure on large scales. Here, we employ mass photometry, phylogenetics, and structural biology to interrogate the evolution of homo-oligomeric assembly across the entire phylogeny of prokaryotic citrate synthases - an enzyme with a highly conserved function. We discover a menagerie of different assembly types that come and go over the course of evolution, including cases of parallel evolution and reversions from complex to simple assemblies. Functional experiments in vitro and in vivo indicate that evolutionary transitions between different assemblies do not strongly influence enzyme catalysis. Our work suggests that enzymes can wander relatively freely through a large space of possible assemblies and demonstrates the power of characterizing structure-function relationships across entire phylogenies.

14.
Biochem Biophys Res Commun ; 435(2): 250-4, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23643786

RESUMEN

The ubiquitin-proteasome system is responsible for regulated protein degradation in the cell with the 26S proteasome acting as its executive arm. The molecular architecture of this 2.5 MDa complex has been established recently, with the notable exception of the small acidic subunit Sem1. Here, we localize the C-terminal helix of Sem1 binding to the PCI domain of the subunit Rpn7 using cryo-electron microscopy single particle reconstruction of proteasomes purified from yeast cells with sem1 deletion. The approximate position of the N-terminal region of Sem1 bridging the cleft between Rpn7 and Rpn3 was inferred based on site-specific cross-linking data of the 26S proteasome. Our structural studies indicate that Sem1 can assume different conformations in different contexts, which supports the idea that Sem1 functions as a molecular glue stabilizing the Rpn3/Rpn7 heterodimer.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/química , Elementos Reguladores de la Transcripción , Proteínas de Saccharomyces cerevisiae/química , Secuencia de Aminoácidos , Datos de Secuencia Molecular , Subunidades de Proteína/química
15.
Proc Natl Acad Sci U S A ; 107(49): 20992-7, 2010 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-21098295

RESUMEN

The structure of the 26S proteasome from Schizosaccharomyces pombe has been determined to a resolution of 9.1 Å by cryoelectron microscopy and single particle analysis. In addition, chemical cross-linking in conjunction with mass spectrometry has been used to identify numerous residue pairs in close proximity to each other, providing an array of spatial restraints. Taken together these data clarify the topology of the AAA-ATPase module in the 19S regulatory particle and its spatial relationship to the α-ring of the 20S core particle. Image classification and variance analysis reveal a belt of high "activity" surrounding the AAA-ATPase module which is tentatively assigned to the reversible association of proteasome interacting proteins and the conformational heterogeneity among the particles. An integrated model is presented which sheds light on the early steps of protein degradation by the 26S complex.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/química , Schizosaccharomyces/química , Microscopía por Crioelectrón/métodos , Espectrometría de Masas/métodos , Simulación de Dinámica Molecular , Conformación Proteica , Proteínas/metabolismo
16.
Cell Microbiol ; 12(3): 362-71, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19863555

RESUMEN

Cellular organelles are usually linked to the cytoskeleton, which often provides a scaffold for organelle function. In malaria parasites, no link between the cytoskeleton and the major organelles is known. Here we show that during fast, stop-and-go motion of Plasmodium sporozoites, all organelles stay largely fixed in respect to the moving parasite. Cryogenic electron tomography reveals that the nucleus, mitochondrion, apicoplast and the microtubules of Plasmodium sporozoites are linked to the parasite pellicle via long tethering proteins. These tethers originate from the inner membrane complex and are arranged in a periodic fashion following a 32 nm repeat. The tethers pass through a subpellicular structure that encompasses the entire parasite, probably as a network of membrane-associated filaments. While the spatial organization of the large parasite organelles appears dependent on their linkage to the cortex, the specialized secretory vesicles are mostly not linked to microtubules or other cellular structures that could provide support for movement.


Asunto(s)
Citoesqueleto/metabolismo , Membranas Intracelulares/fisiología , Orgánulos/metabolismo , Plasmodium/fisiología , Esporozoítos/fisiología , Animales , Movimiento Celular , Microscopía por Crioelectrón , Orgánulos/ultraestructura , Plasmodium/ultraestructura , Esporozoítos/ultraestructura
17.
Bio Protoc ; 11(18): e4161, 2021 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-34692911

RESUMEN

Genome-wide sequencing of RNA (RNA-seq) has become an inexpensive tool to gain key insights into cellular and disease mechanisms. Sample preparation and sequencing are streamlined and allow the acquisition of hundreds of gene expression profiles in a few days; however, in particular, data processing, curation, and analysis involve numerous steps that can be overwhelming to non-experts. Here, the sample preparation, sequencing, and data processing workflow for RNA-seq expression analysis in yeast is described. While this protocol covers only a small portion of the RNA-seq landscape, the principal workflow common to such experiments is described, allowing the reader to adapt the protocol where necessary. Graphic abstract: Basic workflow of RNA-seq expression analysis.

18.
Nat Plants ; 7(4): 524-538, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33846594

RESUMEN

Biogenesis of photosystem II (PSII), nature's water-splitting catalyst, is assisted by auxiliary proteins that form transient complexes with PSII components to facilitate stepwise assembly events. Using cryo-electron microscopy, we solved the structure of such a PSII assembly intermediate from Thermosynechococcus elongatus at 2.94 Å resolution. It contains three assembly factors (Psb27, Psb28 and Psb34) and provides detailed insights into their molecular function. Binding of Psb28 induces large conformational changes at the PSII acceptor side, which distort the binding pocket of the mobile quinone (QB) and replace the bicarbonate ligand of non-haem iron with glutamate, a structural motif found in reaction centres of non-oxygenic photosynthetic bacteria. These results reveal mechanisms that protect PSII from damage during biogenesis until water splitting is activated. Our structure further demonstrates how the PSII active site is prepared for the incorporation of the Mn4CaO5 cluster, which performs the unique water-splitting reaction.


Asunto(s)
Proteínas Bacterianas/genética , Complejo de Proteína del Fotosistema II/genética , Proteínas Bacterianas/ultraestructura , Fotosíntesis , Complejo de Proteína del Fotosistema II/ultraestructura , Thermosynechococcus/genética , Thermosynechococcus/ultraestructura
19.
Traffic ; 9(8): 1283-98, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18485055

RESUMEN

We previously showed that infection with vaccinia virus (VV) induces cell motility, characterized by contractility and directed migration. Motility is temporally regulated because cells are motile immediately after infection, whereas late in infection motility ceases and cells resettle. Motility and its cessation are accompanied by temporal rearrangements of both the microtubule and the actin networks. Because the F11L gene has previously been implicated in VV-induced migration, we now explore the role of F11L in contractility, migration, the cessation of motility and the cytoskeletal rearrangements. By live cell imaging using a VV that lacks an intact F11L gene, we show that F11L facilitates cell detachment and is required for migration but not for contractility. By light microscopy, F11L expression induces a remodeling of the actin, but not the microtubule, network. The lack of migration correlates with smaller plaques, indicating that this process facilitates cell-to-cell spreading of VV. Late in infection, when motility ceases, cells re-establish cell-to-cell contacts in an F11L-independent manner. We finally show that VV-induced motility and its cessation correlate with a temporal regulation of the guanosine triphosphatase RhoA as well as the expression levels of F11L during the infectious cycle.


Asunto(s)
Regulación Viral de la Expresión Génica , Genes Virales , Virus Vaccinia/metabolismo , Proteínas Virales/química , Actinas/metabolismo , Animales , Adhesión Celular , Movimiento Celular , Citoesqueleto/metabolismo , Ratones , Microtúbulos/metabolismo , Modelos Biológicos , ARN Mensajero/metabolismo , Proteínas Virales/metabolismo , Proteínas Virales/fisiología , Proteína de Unión al GTP rhoA/química
20.
Magn Reson Med ; 64(1): 15-22, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20577978

RESUMEN

A good spatial resolution is essential for high precision segmentations of small structures in magnetic resonance images. However, any increase in the spatial resolution results in a decrease of the signal-to-noise ratio (SNR). In this article, this problem is addressed by a new image restoration technique that is used to partly compensate for the loss in SNR. Specifically, a two-stage hybrid image restoration procedure is proposed where the first stage is a Wiener wavelet filter for an initial denoising. The artifacts that will inevitably be produced by this step are subsequently reduced using a recent variant of anisotropic diffusion. The method is applied to magnetic resonance imaging data acquired on a 7-T magnetic resonance imaging scanner and compared with averaged multiple measurements of the same subject. It was found that the effect of image restoration procedure roughly corresponds to averaging across three repeated measurements.


Asunto(s)
Artefactos , Aumento de la Imagen/métodos , Imagen por Resonancia Magnética/métodos , Adulto , Algoritmos , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA