Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 170(1): 48-60.e11, 2017 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-28666122

RESUMEN

Type I CRISPR systems feature a sequential dsDNA target searching and degradation process, by crRNA-displaying Cascade and nuclease-helicase fusion enzyme Cas3, respectively. Here we present two cryo-EM snapshots of the Thermobifida fusca type I-E Cascade: (1) unwinding 11 bp of dsDNA at the seed-sequence region to scout for sequence complementarity, and (2) further unwinding of the entire protospacer to form a full R-loop. These structures provide the much-needed temporal and spatial resolution to resolve key mechanistic steps leading to Cas3 recruitment. In the early steps, PAM recognition causes severe DNA bending, leading to spontaneous DNA unwinding to form a seed-bubble. The full R-loop formation triggers conformational changes in Cascade, licensing Cas3 to bind. The same process also generates a bulge in the non-target DNA strand, enabling its handover to Cas3 for cleavage. The combination of both negative and positive checkpoints ensures stringent yet efficient target degradation in type I CRISPR-Cas systems.


Asunto(s)
Actinobacteria/genética , Actinobacteria/ultraestructura , Sistemas CRISPR-Cas , Hibridación de Ácido Nucleico , Actinobacteria/química , Actinobacteria/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Secuencia de Bases , Proteínas Asociadas a CRISPR/química , Proteínas Asociadas a CRISPR/metabolismo , Proteínas Asociadas a CRISPR/ultraestructura , Microscopía por Crioelectrón , Modelos Moleculares , ARN Bacteriano/química , ARN Bacteriano/metabolismo , ARN Pequeño no Traducido/química , ARN Pequeño no Traducido/metabolismo
2.
Cell ; 163(5): 1138-1152, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26548953

RESUMEN

Diverse repertoires of antigen-receptor genes that result from combinatorial splicing of coding segments by V(D)J recombination are hallmarks of vertebrate immunity. The (RAG1-RAG2)2 recombinase (RAG) recognizes recombination signal sequences (RSSs) containing a heptamer, a spacer of 12 or 23 base pairs, and a nonamer (12-RSS or 23-RSS) and introduces precise breaks at RSS-coding segment junctions. RAG forms synaptic complexes only with one 12-RSS and one 23-RSS, a dogma known as the 12/23 rule that governs the recombination fidelity. We report cryo-electron microscopy structures of synaptic RAG complexes at up to 3.4 Å resolution, which reveal a closed conformation with base flipping and base-specific recognition of RSSs. Distortion at RSS-coding segment junctions and base flipping in coding segments uncover the two-metal-ion catalytic mechanism. Induced asymmetry involving tilting of the nonamer-binding domain dimer of RAG1 upon binding of HMGB1-bent 12-RSS or 23-RSS underlies the molecular mechanism for the 12/23 rule.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Homeodominio/química , Recombinación V(D)J , Secuencia de Aminoácidos , Animales , Microscopía por Crioelectrón , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/ultraestructura , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/ultraestructura , Humanos , Ratones , Datos de Secuencia Molecular , Complejos Multiproteicos/química , Complejos Multiproteicos/ultraestructura , Mutación , Alineación de Secuencia , Pez Cebra
3.
Cell ; 159(4): 857-68, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25417161

RESUMEN

Dyneins power microtubule motility using ring-shaped, AAA-containing motor domains. Here, we report X-ray and electron microscopy (EM) structures of yeast dynein bound to different ATP analogs, which collectively provide insight into the roles of dynein's two major ATPase sites, AAA1 and AAA3, in the conformational change mechanism. ATP binding to AAA1 triggers a cascade of conformational changes that propagate to all six AAA domains and cause a large movement of the "linker," dynein's mechanical element. In contrast to the role of AAA1 in driving motility, nucleotide transitions in AAA3 gate the transmission of conformational changes between AAA1 and the linker, suggesting that AAA3 acts as a regulatory switch. Further structural and mutational studies also uncover a role for the linker in regulating the catalytic cycle of AAA1. Together, these results reveal how dynein's two major ATP-binding sites initiate and modulate conformational changes in the motor domain during motility.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Dineínas/química , Dineínas/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/química , Adenosina Trifosfato/metabolismo , Regulación Alostérica , Catálisis , Cristalografía por Rayos X , Dictyostelium/química , Dineínas/ultraestructura , Microscopía Electrónica , Modelos Moleculares , Estructura Terciaria de Proteína , Proteínas de Saccharomyces cerevisiae/ultraestructura
4.
Brief Bioinform ; 25(3)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38701416

RESUMEN

Predicting protein function is crucial for understanding biological life processes, preventing diseases and developing new drug targets. In recent years, methods based on sequence, structure and biological networks for protein function annotation have been extensively researched. Although obtaining a protein in three-dimensional structure through experimental or computational methods enhances the accuracy of function prediction, the sheer volume of proteins sequenced by high-throughput technologies presents a significant challenge. To address this issue, we introduce a deep neural network model DeepSS2GO (Secondary Structure to Gene Ontology). It is a predictor incorporating secondary structure features along with primary sequence and homology information. The algorithm expertly combines the speed of sequence-based information with the accuracy of structure-based features while streamlining the redundant data in primary sequences and bypassing the time-consuming challenges of tertiary structure analysis. The results show that the prediction performance surpasses state-of-the-art algorithms. It has the ability to predict key functions by effectively utilizing secondary structure information, rather than broadly predicting general Gene Ontology terms. Additionally, DeepSS2GO predicts five times faster than advanced algorithms, making it highly applicable to massive sequencing data. The source code and trained models are available at https://github.com/orca233/DeepSS2GO.


Asunto(s)
Algoritmos , Biología Computacional , Redes Neurales de la Computación , Estructura Secundaria de Proteína , Proteínas , Proteínas/química , Proteínas/metabolismo , Proteínas/genética , Biología Computacional/métodos , Bases de Datos de Proteínas , Ontología de Genes , Análisis de Secuencia de Proteína/métodos , Programas Informáticos
5.
Nature ; 581(7808): 323-328, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32433611

RESUMEN

Triacylglycerols store metabolic energy in organisms and have industrial uses as foods and fuels. Excessive accumulation of triacylglycerols in humans causes obesity and is associated with metabolic diseases1. Triacylglycerol synthesis is catalysed by acyl-CoA diacylglycerol acyltransferase (DGAT) enzymes2-4, the structures and catalytic mechanisms of which remain unknown. Here we determined the structure of dimeric human DGAT1, a member of the membrane-bound O-acyltransferase (MBOAT) family, by cryo-electron microscopy at approximately 3.0 Å resolution. DGAT1 forms a homodimer through N-terminal segments and a hydrophobic interface, with putative active sites within the membrane region. A structure obtained with oleoyl-CoA substrate resolved at approximately 3.2 Å shows that the CoA moiety binds DGAT1 on the cytosolic side and the acyl group lies deep within a hydrophobic channel, positioning the acyl-CoA thioester bond near an invariant catalytic histidine residue. The reaction centre is located inside a large cavity, which opens laterally to the membrane bilayer, providing lipid access to the active site. A lipid-like density-possibly representing an acyl-acceptor molecule-is located within the reaction centre, orthogonal to acyl-CoA. Insights provided by the DGAT1 structures, together with mutagenesis and functional studies, provide the basis for a model of the catalysis of triacylglycerol synthesis by DGAT.


Asunto(s)
Biocatálisis , Microscopía por Crioelectrón , Diacilglicerol O-Acetiltransferasa/metabolismo , Diacilglicerol O-Acetiltransferasa/ultraestructura , Triglicéridos/biosíntesis , Acilcoenzima A/química , Acilcoenzima A/metabolismo , Acilcoenzima A/ultraestructura , Aciltransferasas/química , Aciltransferasas/metabolismo , Dominio Catalítico , Membrana Celular/química , Membrana Celular/metabolismo , Diacilglicerol O-Acetiltransferasa/química , Histidina/química , Histidina/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Multimerización de Proteína , Especificidad por Sustrato
6.
Nature ; 582(7810): 129-133, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32494073

RESUMEN

Mitochondria take up Ca2+ through the mitochondrial calcium uniporter complex to regulate energy production, cytosolic Ca2+ signalling and cell death1,2. In mammals, the uniporter complex (uniplex) contains four core components: the pore-forming MCU protein, the gatekeepers MICU1 and MICU2, and an auxiliary subunit, EMRE, essential for Ca2+ transport3-8. To prevent detrimental Ca2+ overload, the activity of MCU must be tightly regulated by MICUs, which sense changes in cytosolic Ca2+ concentrations to switch MCU on and off9,10. Here we report cryo-electron microscopic structures of the human mitochondrial calcium uniporter holocomplex in inhibited and Ca2+-activated states. These structures define the architecture of this multicomponent Ca2+-uptake machinery and reveal the gating mechanism by which MICUs control uniporter activity. Our work provides a framework for understanding regulated Ca2+ uptake in mitochondria, and could suggest ways of modulating uniporter activity to treat diseases related to mitochondrial Ca2+ overload.


Asunto(s)
Canales de Calcio/química , Canales de Calcio/metabolismo , Microscopía por Crioelectrón , Sitios de Unión/efectos de los fármacos , Calcio/metabolismo , Calcio/farmacología , Canales de Calcio/ultraestructura , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura
7.
Nat Methods ; 19(5): 576-585, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35501384

RESUMEN

High-resolution structural studies are essential for understanding the folding and function of diverse RNAs. Herein, we present a nanoarchitectural engineering strategy for efficient structural determination of RNA-only structures using single-particle cryogenic electron microscopy (cryo-EM). This strategy-ROCK (RNA oligomerization-enabled cryo-EM via installing kissing loops)-involves installing kissing-loop sequences onto the functionally nonessential stems of RNAs for homomeric self-assembly into closed rings with multiplied molecular weights and mitigated structural flexibility. ROCK enables cryo-EM reconstruction of the Tetrahymena group I intron at 2.98-Å resolution overall (2.85 Å for the core), allowing de novo model building of the complete RNA, including the previously unknown peripheral domains. ROCK is further applied to two smaller RNAs-the Azoarcus group I intron and the FMN riboswitch, revealing the conformational change of the former and the bound ligand in the latter. ROCK holds promise to greatly facilitate the use of cryo-EM in RNA structural studies.


Asunto(s)
ARN , Riboswitch , Microscopía por Crioelectrón , Ligandos , ARN/genética , Imagen Individual de Molécula
8.
Nature ; 567(7749): 486-490, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30894744

RESUMEN

In Gram-negative bacteria, lipopolysaccharide is essential for outer membrane formation and antibiotic resistance. The seven lipopolysaccharide transport (Lpt) proteins A-G move lipopolysaccharide from the inner to the outer membrane. The ATP-binding cassette transporter LptB2FG, which tightly associates with LptC, extracts lipopolysaccharide out of the inner membrane. The mechanism of the LptB2FG-LptC complex (LptB2FGC) and the role of LptC in lipopolysaccharide transport are poorly understood. Here we characterize the structures of LptB2FG and LptB2FGC in nucleotide-free and vanadate-trapped states, using single-particle cryo-electron microscopy. These structures resolve the bound lipopolysaccharide, reveal transporter-lipopolysaccharide interactions with side-chain details and uncover how the capture and extrusion of lipopolysaccharide are coupled to conformational rearrangements of LptB2FGC. LptC inserts its transmembrane helix between the two transmembrane domains of LptB2FG, which represents a previously unknown regulatory mechanism for ATP-binding cassette transporters. Our results suggest a role for LptC in achieving efficient lipopolysaccharide transport, by coordinating the action of LptB2FG in the inner membrane and Lpt protein interactions in the periplasm.


Asunto(s)
Microscopía por Crioelectrón , Proteínas de Escherichia coli/metabolismo , Escherichia coli/química , Escherichia coli/metabolismo , Lipopolisacáridos/química , Lipopolisacáridos/metabolismo , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/química , Complejos Multiproteicos/ultraestructura , Escherichia coli/ultraestructura , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/ultraestructura , Proteínas de la Membrana/química , Proteínas de la Membrana/ultraestructura , Modelos Moleculares , Complejos Multiproteicos/metabolismo , Unión Proteica/efectos de los fármacos , Dominios Proteicos/efectos de los fármacos , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Relación Estructura-Actividad , Vanadatos/química , Vanadatos/metabolismo , Vanadatos/farmacología
9.
Nature ; 565(7739): 318-323, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30542158

RESUMEN

HIV-1 envelope glycoprotein (Env), which consists of trimeric (gp160)3 cleaved to (gp120 and gp41)3, interacts with the primary receptor CD4 and a coreceptor (such as chemokine receptor CCR5) to fuse viral and target-cell membranes. The gp120-coreceptor interaction has previously been proposed as the most crucial trigger for unleashing the fusogenic potential of gp41. Here we report a cryo-electron microscopy structure of a full-length gp120 in complex with soluble CD4 and unmodified human CCR5, at 3.9 Å resolution. The V3 loop of gp120 inserts into the chemokine-binding pocket formed by seven transmembrane helices of CCR5, and the N terminus of CCR5 contacts the CD4-induced bridging sheet of gp120. CCR5 induces no obvious allosteric changes in gp120 that can propagate to gp41; it does bring the Env trimer close to the target membrane. The N terminus of gp120, which is gripped by gp41 in the pre-fusion or CD4-bound Env, flips back in the CCR5-bound conformation and may irreversibly destabilize gp41 to initiate fusion. The coreceptor probably functions by stabilizing and anchoring the CD4-induced conformation of Env near the cell membrane. These results advance our understanding of HIV-1 entry into host cells and may guide the development of vaccines and therapeutic agents.


Asunto(s)
Antígenos CD4/química , Antígenos CD4/ultraestructura , Proteína gp120 de Envoltorio del VIH/química , Proteína gp120 de Envoltorio del VIH/ultraestructura , Receptores CCR5/química , Receptores CCR5/ultraestructura , Receptores del VIH/química , Receptores del VIH/ultraestructura , Fármacos Anti-VIH/química , Fármacos Anti-VIH/metabolismo , Sitios de Unión , Antígenos CD4/aislamiento & purificación , Antígenos CD4/metabolismo , Línea Celular , Quimiocina CCL5/química , Quimiocina CCL5/metabolismo , Proteína gp120 de Envoltorio del VIH/aislamiento & purificación , Proteína gp120 de Envoltorio del VIH/metabolismo , Proteína gp41 de Envoltorio del VIH/química , Proteína gp41 de Envoltorio del VIH/metabolismo , Proteína gp41 de Envoltorio del VIH/ultraestructura , Humanos , Ligandos , Maraviroc/química , Maraviroc/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica , Receptores CCR5/aislamiento & purificación , Receptores CCR5/metabolismo , Receptores del VIH/antagonistas & inhibidores , Receptores del VIH/metabolismo
10.
Nature ; 572(7770): 488-492, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31367042

RESUMEN

Cation-chloride cotransporters (CCCs) mediate the electroneutral transport of chloride, potassium and/or sodium across the membrane. They have critical roles in regulating cell volume, controlling ion absorption and secretion across epithelia, and maintaining intracellular chloride homeostasis. These transporters are primary targets for some of the most commonly prescribed drugs. Here we determined the cryo-electron microscopy structure of the Na-K-Cl cotransporter NKCC1, an extensively studied member of the CCC family, from Danio rerio. The structure defines the architecture of this protein family and reveals how cytosolic and transmembrane domains are strategically positioned for communication. Structural analyses, functional characterizations and computational studies reveal the ion-translocation pathway, ion-binding sites and key residues for transport activity. These results provide insights into ion selectivity, coupling and translocation, and establish a framework for understanding the physiological functions of CCCs and interpreting disease-related mutations.


Asunto(s)
Microscopía por Crioelectrón , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Miembro 2 de la Familia de Transportadores de Soluto 12/ultraestructura , Pez Cebra , Secuencia de Aminoácidos , Animales , Sitios de Unión , Cationes Monovalentes/metabolismo , Cloruros/metabolismo , Citosol/metabolismo , Síndrome de Gitelman/genética , Humanos , Transporte Iónico , Modelos Moleculares , Simulación de Dinámica Molecular , Potasio/metabolismo , Dominios Proteicos , Sodio/metabolismo , Miembro 2 de la Familia de Transportadores de Soluto 12/química , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Pez Cebra/genética
11.
Annu Rev Physiol ; 83: 153-181, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33141631

RESUMEN

Cholesterol homeostasis and trafficking are critical to the maintenance of the asymmetric plasma membrane of eukaryotic cells. Disruption or dysfunction of cholesterol trafficking leads to numerous human diseases. ATP-binding cassette (ABC) transporters play several critical roles in this process, and mutations in these sterol transporters lead to disorders such as Tangier disease and sitosterolemia. Biochemical and structural information on ABC sterol transporters is beginning to emerge, with published structures of ABCA1 and ABCG5/G8; these two proteins function in the reverse cholesterol transport pathway and mediate the efflux of cholesterol and xenosterols to high-density lipoprotein and bile salt micelles, respectively. Although both of these transporters belong to the ABC family and mediate the efflux of a sterol substrate, they have many distinct differences. Here, we summarize the current understanding of sterol transport driven by ABC transporters, with an emphasis on these two extensively characterized transporters.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Transporte Biológico/fisiología , Esteroles/metabolismo , Animales , Colesterol/metabolismo , Humanos
12.
Nature ; 559(7715): 575-579, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29995856

RESUMEN

Mitochondrial calcium uptake is critical for regulating ATP production, intracellular calcium signalling, and cell death. This uptake is mediated by a highly selective calcium channel called the mitochondrial calcium uniporter (MCU). Here, we determined the structures of the pore-forming MCU proteins from two fungi by X-ray crystallography and single-particle cryo-electron microscopy. The stoichiometry, overall architecture, and individual subunit structure differed markedly from those described in the recent nuclear magnetic resonance structure of Caenorhabditis elegans MCU. We observed a dimer-of-dimer architecture across species and chemical environments, which was corroborated by biochemical experiments. Structural analyses and functional characterization uncovered the roles of key residues in the pore. These results reveal a new ion channel architecture, provide insights into calcium coordination, selectivity and conduction, and establish a structural framework for understanding the mechanism of mitochondrial calcium uniporter function.


Asunto(s)
Canales de Calcio/química , Canales de Calcio/ultraestructura , Microscopía por Crioelectrón , Fusarium/química , Metarhizium/química , Animales , Caenorhabditis elegans/química , Calcio/metabolismo , Canales de Calcio/metabolismo , Cristalografía por Rayos X , Activación del Canal Iónico , Modelos Moleculares , Dominios Proteicos , Multimerización de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Reproducibilidad de los Resultados , Solubilidad
13.
Brain ; 145(5): 1839-1853, 2022 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34919654

RESUMEN

CACNA1I is implicated in the susceptibility to schizophrenia by large-scale genetic association studies of single nucleotide polymorphisms. However, the channelopathy of CACNA1I in schizophrenia is unknown. CACNA1I encodes CaV3.3, a neuronal voltage-gated calcium channel that underlies a subtype of T-type current that is important for neuronal excitability in the thalamic reticular nucleus and other regions of the brain. Here, we present an extensive functional characterization of 57 naturally occurring rare and common missense variants of CACNA1I derived from a Swedish schizophrenia cohort of more than 10 000 individuals. Our analysis of this allelic series of coding CACNA1I variants revealed that reduced CaV3.3 channel current density was the dominant phenotype associated with rare CACNA1I coding alleles derived from control subjects, whereas rare CACNA1I alleles from schizophrenia patients encoded CaV3.3 channels with altered responses to voltages. CACNA1I variants associated with altered current density primarily impact the ionic channel pore and those associated with altered responses to voltage impact the voltage-sensing domain. CaV3.3 variants associated with altered voltage dependence of the CaV3.3 channel and those associated with peak current density deficits were significantly segregated across affected and unaffected groups (Fisher's exact test, P = 0.034). Our results, together with recent data from the SCHEMA (Schizophrenia Exome Sequencing Meta-Analysis) cohort, suggest that reduced CaV3.3 function may protect against schizophrenia risk in rare cases. We subsequently modelled the effect of the biophysical properties of CaV3.3 channel variants on thalamic reticular nucleus excitability and found that compared with common variants, ultrarare CaV3.3-coding variants derived from control subjects significantly decreased thalamic reticular nucleus excitability (P = 0.011). When all rare variants were analysed, there was a non-significant trend between variants that reduced thalamic reticular nucleus excitability and variants that either had no effect or increased thalamic reticular nucleus excitability across disease status. Taken together, the results of our functional analysis of an allelic series of >50 CACNA1I variants in a schizophrenia cohort reveal that loss of function of CaV3.3 is a molecular phenotype associated with reduced disease risk burden, and our approach may serve as a template strategy for channelopathies in polygenic disorders.


Asunto(s)
Canales de Calcio Tipo T , Canalopatías , Esquizofrenia , Alelos , Canales de Calcio Tipo T/genética , Canalopatías/genética , Humanos , Mutación Missense , Esquizofrenia/genética , Suecia
14.
Nature ; 549(7671): 233-237, 2017 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-28869968

RESUMEN

Lipopolysaccharide (LPS) in the outer membrane of Gram-negative bacteria is critical for the assembly of their cell envelopes. LPS synthesized in the cytoplasmic leaflet of the inner membrane is flipped to the periplasmic leaflet by MsbA, an ATP-binding cassette transporter. Despite substantial efforts, the structural mechanisms underlying MsbA-driven LPS flipping remain elusive. Here we use single-particle cryo-electron microscopy to elucidate the structures of lipid-nanodisc-embedded MsbA in three functional states. The 4.2 Å-resolution structure of the transmembrane domains of nucleotide-free MsbA reveals that LPS binds deep inside MsbA at the height of the periplasmic leaflet, establishing extensive hydrophilic and hydrophobic interactions with MsbA. Two sub-nanometre-resolution structures of MsbA with ADP-vanadate and ADP reveal an unprecedented closed and an inward-facing conformation, respectively. Our study uncovers the structural basis for LPS recognition, delineates the conformational transitions of MsbA to flip LPS, and paves the way for structural characterization of other lipid flippases.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/ultraestructura , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Microscopía por Crioelectrón , Escherichia coli , Lipopolisacáridos/metabolismo , Transportadoras de Casetes de Unión a ATP/química , Adenosina Difosfato/química , Adenosina Difosfato/metabolismo , Proteínas Bacterianas/química , Transporte Biológico , Membrana Celular/química , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Escherichia coli/citología , Escherichia coli/enzimología , Escherichia coli/ultraestructura , Interacciones Hidrofóbicas e Hidrofílicas , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Modelos Moleculares , Nanoestructuras/química , Nanoestructuras/ultraestructura , Periplasma/química , Periplasma/metabolismo , Periplasma/ultraestructura , Unión Proteica , Dominios Proteicos
15.
Nature ; 548(7667): 352-355, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28682307

RESUMEN

Misfolded endoplasmic reticulum proteins are retro-translocated through the membrane into the cytosol, where they are poly-ubiquitinated, extracted from the membrane, and degraded by the proteasome-a pathway termed endoplasmic reticulum-associated protein degradation (ERAD). Proteins with misfolded domains in the endoplasmic reticulum lumen or membrane are discarded through the ERAD-L and ERAD-M pathways, respectively. In Saccharomyces cerevisiae, both pathways require the ubiquitin ligase Hrd1, a multi-spanning membrane protein with a cytosolic RING finger domain. Hrd1 is the crucial membrane component for retro-translocation, but it is unclear whether it forms a protein-conducting channel. Here we present a cryo-electron microscopy structure of S. cerevisiae Hrd1 in complex with its endoplasmic reticulum luminal binding partner, Hrd3. Hrd1 forms a dimer within the membrane with one or two Hrd3 molecules associated at its luminal side. Each Hrd1 molecule has eight transmembrane segments, five of which form an aqueous cavity extending from the cytosol almost to the endoplasmic reticulum lumen, while a segment of the neighbouring Hrd1 molecule forms a lateral seal. The aqueous cavity and lateral gate are reminiscent of features of protein-conducting conduits that facilitate polypeptide movement in the opposite direction-from the cytosol into or across membranes. Our results suggest that Hrd1 forms a retro-translocation channel for the movement of misfolded polypeptides through the endoplasmic reticulum membrane.


Asunto(s)
Microscopía por Crioelectrón , Degradación Asociada con el Retículo Endoplásmico , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/ultraestructura , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/ultraestructura , Saccharomyces cerevisiae/química , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/ultraestructura , Interacciones Hidrofóbicas e Hidrofílicas , Glicoproteínas de Membrana/química , Modelos Moleculares , Conformación Proteica , Saccharomyces cerevisiae/ultraestructura , Proteínas de Saccharomyces cerevisiae/química , Ubiquitina-Proteína Ligasas/química
16.
J Biol Chem ; 293(14): 5360-5373, 2018 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-29462788

RESUMEN

Many pathogenic bacteria, including Streptococcus gordonii, possess a pathway for the cellular export of a single serine-rich-repeat protein that mediates the adhesion of bacteria to host cells and the extracellular matrix. This adhesin protein is O-glycosylated by several cytosolic glycosyltransferases and requires three accessory Sec proteins (Asp1-3) for export, but how the adhesin protein is processed for export is not well understood. Here, we report that the S. gordonii adhesin GspB is sequentially O-glycosylated by three enzymes (GtfA/B, Nss, and Gly) that attach N-acetylglucosamine and glucose to Ser/Thr residues. We also found that modified GspB is transferred from the last glycosyltransferase to the Asp1/2/3 complex. Crystal structures revealed that both Asp1 and Asp3 are related to carbohydrate-binding proteins, suggesting that they interact with carbohydrates and bind glycosylated adhesin, a notion that was supported by further analyses. We further observed that Asp1 also has an affinity for phospholipids, which is attenuated by Asp2. In summary, our findings support a model in which the GspB adhesin is sequentially glycosylated by GtfA/B, Nss, and Gly and then transferred to the Asp1/2/3 complex in which Asp1 mediates the interaction of the Asp1/2/3 complex with the lipid bilayer for targeting of matured GspB to the export machinery.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Streptococcus gordonii/metabolismo , Acetilglucosamina/metabolismo , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Citosol/metabolismo , Glicosilación , Glicosiltransferasas/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Unión Proteica , Transporte de Proteínas/fisiología , Streptococcus gordonii/fisiología
17.
Nature ; 504(7478): 107-12, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24305160

RESUMEN

Transient receptor potential (TRP) channels are sensors for a wide range of cellular and environmental signals, but elucidating how these channels respond to physical and chemical stimuli has been hampered by a lack of detailed structural information. Here we exploit advances in electron cryo-microscopy to determine the structure of a mammalian TRP channel, TRPV1, at 3.4 Å resolution, breaking the side-chain resolution barrier for membrane proteins without crystallization. Like voltage-gated channels, TRPV1 exhibits four-fold symmetry around a central ion pathway formed by transmembrane segments 5-6 (S5-S6) and the intervening pore loop, which is flanked by S1-S4 voltage-sensor-like domains. TRPV1 has a wide extracellular 'mouth' with a short selectivity filter. The conserved 'TRP domain' interacts with the S4-S5 linker, consistent with its contribution to allosteric modulation. Subunit organization is facilitated by interactions among cytoplasmic domains, including amino-terminal ankyrin repeats. These observations provide a structural blueprint for understanding unique aspects of TRP channel function.


Asunto(s)
Microscopía por Crioelectrón , Modelos Moleculares , Canales Catiónicos TRPV/química , Animales , Repetición de Anquirina , Células HEK293 , Humanos , Estructura Terciaria de Proteína , Ratas
18.
Nature ; 504(7478): 113-8, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24305161

RESUMEN

Transient receptor potential (TRP) channels are polymodal signal detectors that respond to a wide range of physical and chemical stimuli. Elucidating how these channels integrate and convert physiological signals into channel opening is essential to understanding how they regulate cell excitability under normal and pathophysiological conditions. Here we exploit pharmacological probes (a peptide toxin and small vanilloid agonists) to determine structures of two activated states of the capsaicin receptor, TRPV1. A domain (consisting of transmembrane segments 1-4) that moves during activation of voltage-gated channels remains stationary in TRPV1, highlighting differences in gating mechanisms for these structurally related channel superfamilies. TRPV1 opening is associated with major structural rearrangements in the outer pore, including the pore helix and selectivity filter, as well as pronounced dilation of a hydrophobic constriction at the lower gate, suggesting a dual gating mechanism. Allosteric coupling between upper and lower gates may account for rich physiological modulation exhibited by TRPV1 and other TRP channels.


Asunto(s)
Modelos Moleculares , Canales Catiónicos TRPV/química , Canales Catiónicos TRPV/fisiología , Animales , Mutación , Estructura Terciaria de Proteína , Ratas , Canales Catiónicos TRPV/genética
19.
Nature ; 496(7445): 377-81, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23485968

RESUMEN

A hallmark of histone H3 lysine 9 (H3K9)-methylated heterochromatin, conserved from the fission yeast Schizosaccharomyces pombe to humans, is its ability to spread to adjacent genomic regions. Central to heterochromatin spread is heterochromatin protein 1 (HP1), which recognizes H3K9-methylated chromatin, oligomerizes and forms a versatile platform that participates in diverse nuclear functions, ranging from gene silencing to chromosome segregation. How HP1 proteins assemble on methylated nucleosomal templates and how the HP1-nucleosome complex achieves functional versatility remain poorly understood. Here we show that binding of the key S. pombe HP1 protein, Swi6, to methylated nucleosomes drives a switch from an auto-inhibited state to a spreading-competent state. In the auto-inhibited state, a histone-mimic sequence in one Swi6 monomer blocks methyl-mark recognition by the chromodomain of another monomer. Auto-inhibition is relieved by recognition of two template features, the H3K9 methyl mark and nucleosomal DNA. Cryo-electron-microscopy-based reconstruction of the Swi6-nucleosome complex provides the overall architecture of the spreading-competent state in which two unbound chromodomain sticky ends appear exposed. Disruption of the switch between the auto-inhibited and spreading-competent states disrupts heterochromatin assembly and gene silencing in vivo. These findings are reminiscent of other conditionally activated polymerization processes, such as actin nucleation, and open up a new class of regulatory mechanisms that operate on chromatin in vivo.


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Heterocromatina/metabolismo , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Secuencia de Aminoácidos , Animales , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/ultraestructura , Microscopía por Crioelectrón , Silenciador del Gen , Heterocromatina/química , Heterocromatina/ultraestructura , Histonas/química , Histonas/metabolismo , Metilación , Modelos Moleculares , Datos de Secuencia Molecular , Nucleosomas/química , Nucleosomas/genética , Nucleosomas/metabolismo , Nucleosomas/ultraestructura , Estructura Terciaria de Proteína , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/antagonistas & inhibidores , Proteínas de Schizosaccharomyces pombe/ultraestructura , Xenopus laevis
20.
Nat Struct Mol Biol ; 31(4): 657-666, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38316880

RESUMEN

Mitochondrial adenosine triphosphate (ATP) synthase uses the proton gradient across the inner mitochondrial membrane to synthesize ATP. Structural and single molecule studies conducted mostly at neutral or basic pH have provided details of the reaction mechanism of ATP synthesis. However, pH of the mitochondrial matrix is slightly acidic during hypoxia and pH-dependent conformational changes in the ATP synthase have been reported. Here we use single-particle cryo-EM to analyze the conformational ensemble of the yeast (Saccharomyces cerevisiae) ATP synthase at pH 6. Of the four conformations resolved in this study, three are reaction intermediates. In addition to canonical catalytic dwell and binding dwell structures, we identify two unique conformations with nearly identical positions of the central rotor but different catalytic site conformations. These structures provide new insights into the catalytic mechanism of the ATP synthase and highlight elastic coupling between the catalytic and proton translocating domains.


Asunto(s)
Protones , Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolismo , Adenosina Trifosfato/metabolismo , ATPasas de Translocación de Protón Mitocondriales/química , Conformación Proteica , Concentración de Iones de Hidrógeno
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA