Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Cell ; 167(1): 145-157.e17, 2016 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-27662087

RESUMO

The type-1 ryanodine receptor (RyR1) is an intracellular calcium (Ca(2+)) release channel required for skeletal muscle contraction. Here, we present cryo-EM reconstructions of RyR1 in multiple functional states revealing the structural basis of channel gating and ligand-dependent activation. Binding sites for the channel activators Ca(2+), ATP, and caffeine were identified at interdomain interfaces of the C-terminal domain. Either ATP or Ca(2+) alone induces conformational changes in the cytoplasmic assembly ("priming"), without pore dilation. In contrast, in the presence of all three activating ligands, high-resolution reconstructions of open and closed states of RyR1 were obtained from the same sample, enabling analyses of conformational changes associated with gating. Gating involves global conformational changes in the cytosolic assembly accompanied by local changes in the transmembrane domain, which include bending of the S6 transmembrane segment and consequent pore dilation, displacement, and deformation of the S4-S5 linker and conformational changes in the pseudo-voltage-sensor domain.


Assuntos
Agonistas dos Canais de Cálcio/química , Ativação do Canal Iônico , Contração Muscular , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Animais , Sítios de Ligação , Cafeína/química , Cálcio/química , Microscopia Crioeletrônica , Ligantes , Domínios Proteicos , Coelhos , Proteínas de Ligação a Tacrolimo/química
2.
Nature ; 629(8011): 481-488, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38632411

RESUMO

The human calcium-sensing receptor (CaSR) detects fluctuations in the extracellular Ca2+ concentration and maintains Ca2+ homeostasis1,2. It also mediates diverse cellular processes not associated with Ca2+ balance3-5. The functional pleiotropy of CaSR arises in part from its ability to signal through several G-protein subtypes6. We determined structures of CaSR in complex with G proteins from three different subfamilies: Gq, Gi and Gs. We found that the homodimeric CaSR of each complex couples to a single G protein through a common mode. This involves the C-terminal helix of each Gα subunit binding to a shallow pocket that is formed in one CaSR subunit by all three intracellular loops (ICL1-ICL3), an extended transmembrane helix 3 and an ordered C-terminal region. G-protein binding expands the transmembrane dimer interface, which is further stabilized by phospholipid. The restraint imposed by the receptor dimer, in combination with ICL2, enables G-protein activation by facilitating conformational transition of Gα. We identified a single Gα residue that determines Gq and Gs versus Gi selectivity. The length and flexibility of ICL2 allows CaSR to bind all three Gα subtypes, thereby conferring capacity for promiscuous G-protein coupling.


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP , Receptores de Detecção de Cálcio , Humanos , Cálcio/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/química , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/química , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/química , Modelos Moleculares , Ligação Proteica , Multimerização Proteica , Receptores de Detecção de Cálcio/metabolismo , Receptores de Detecção de Cálcio/química , Proteínas Heterotriméricas de Ligação ao GTP/química , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Sítios de Ligação , Estrutura Secundária de Proteína , Especificidade por Substrato
3.
Cell ; 153(5): 1108-19, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23706745

RESUMO

Eukaryotic translation initiation begins with assembly of a 43S preinitiation complex. First, methionylated initiator methionine transfer RNA (Met-tRNAi(Met)), eukaryotic initiation factor (eIF) 2, and guanosine triphosphate form a ternary complex (TC). The TC, eIF3, eIF1, and eIF1A cooperatively bind to the 40S subunit, yielding the 43S preinitiation complex, which is ready to attach to messenger RNA (mRNA) and start scanning to the initiation codon. Scanning on structured mRNAs additionally requires DHX29, a DExH-box protein that also binds directly to the 40S subunit. Here, we present a cryo-electron microscopy structure of the mammalian DHX29-bound 43S complex at 11.6 Å resolution. It reveals that eIF2 interacts with the 40S subunit via its α subunit and supports Met-tRNAi(Met) in an unexpected P/I orientation (eP/I). The structural core of eIF3 resides on the back of the 40S subunit, establishing two principal points of contact, whereas DHX29 binds around helix 16. The structure provides insights into eukaryote-specific aspects of translation, including the mechanism of action of DHX29.


Assuntos
Mamíferos/metabolismo , Iniciação Traducional da Cadeia Peptídica , RNA Helicases/química , RNA Ribossômico/química , Ribonucleoproteínas/química , Animais , Sequência de Bases , Sistema Livre de Células , Microscopia Crioeletrônica , Fator de Iniciação 2 em Eucariotos/química , Fator de Iniciação 2 em Eucariotos/metabolismo , Humanos , Mamíferos/genética , Modelos Moleculares , Dados de Sequência Molecular , RNA Helicases/metabolismo , RNA Ribossômico/metabolismo , RNA Ribossômico 18S/química , RNA Ribossômico 18S/metabolismo , Coelhos , Ribonucleoproteínas/metabolismo
5.
Nature ; 584(7820): 304-309, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32581365

RESUMO

The human GABAB receptor-a member of the class C family of G-protein-coupled receptors (GPCRs)-mediates inhibitory neurotransmission and has been implicated in epilepsy, pain and addiction1. A unique GPCR that is known to require heterodimerization for function2-6, the GABAB receptor has two subunits, GABAB1 and GABAB2, that are structurally homologous but perform distinct and complementary functions. GABAB1 recognizes orthosteric ligands7,8, while GABAB2 couples with G proteins9-14. Each subunit is characterized by an extracellular Venus flytrap (VFT) module, a descending peptide linker, a seven-helix transmembrane domain and a cytoplasmic tail15. Although the VFT heterodimer structure has been resolved16, the structure of the full-length receptor and its transmembrane signalling mechanism remain unknown. Here we present a near full-length structure of the GABAB receptor, captured in an inactive state by cryo-electron microscopy. Our structure reveals several ligands that preassociate with the receptor, including two large endogenous phospholipids that are embedded within the transmembrane domains to maintain receptor integrity and modulate receptor function. We also identify a previously unknown heterodimer interface between transmembrane helices 3 and 5 of both subunits, which serves as a signature of the inactive conformation. A unique 'intersubunit latch' within this transmembrane interface maintains the inactive state, and its disruption leads to constitutive receptor activity.


Assuntos
Microscopia Crioeletrônica , Receptores de GABA-B/química , Receptores de GABA-B/ultraestrutura , Cálcio/metabolismo , Etanolaminas/química , Etanolaminas/metabolismo , Humanos , Ligantes , Modelos Moleculares , Fosforilcolina/química , Fosforilcolina/metabolismo , Domínios Proteicos , Multimerização Proteica , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Receptores de GABA-B/metabolismo , Relação Estrutura-Atividade
6.
Nature ; 553(7687): 233-237, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29258289

RESUMO

Calcium-selective transient receptor potential vanilloid subfamily member 6 (TRPV6) channels play a critical role in calcium uptake in epithelial tissues. Altered TRPV6 expression is associated with a variety of human diseases, including cancers. TRPV6 channels are constitutively active and their open probability depends on the lipidic composition of the membrane in which they reside; it increases substantially in the presence of phosphatidylinositol 4,5-bisphosphate. Crystal structures of detergent-solubilized rat TRPV6 in the closed state have previously been solved. Corroborating electrophysiological results, these structures demonstrated that the Ca2+ selectivity of TRPV6 arises from a ring of aspartate side chains in the selectivity filter that binds Ca2+ tightly. However, how TRPV6 channels open and close their pores for ion permeation has remained unclear. Here we present cryo-electron microscopy structures of human TRPV6 in the open and closed states. The channel selectivity filter adopts similar conformations in both states, consistent with its explicit role in ion permeation. The iris-like channel opening is accompanied by an α-to-π-helical transition in the pore-lining transmembrane helix S6 at an alanine hinge just below the selectivity filter. As a result of this transition, the S6 helices bend and rotate, exposing different residues to the ion channel pore in the open and closed states. This gating mechanism, which defines the constitutive activity of TRPV6, is, to our knowledge, unique among tetrameric ion channels and provides structural insights for understanding their diverse roles in physiology and disease.


Assuntos
Canais de Cálcio/metabolismo , Canais de Cálcio/ultraestrutura , Microscopia Crioeletrônica , Células Epiteliais/metabolismo , Ativação do Canal Iônico , Canais de Cátion TRPV/metabolismo , Canais de Cátion TRPV/ultraestrutura , Alanina/metabolismo , Cálcio/metabolismo , Canais de Cálcio/química , Humanos , Transporte de Íons , Conformação Proteica , Rotação , Canais de Cátion TRPV/química
7.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34916296

RESUMO

The human extracellular calcium-sensing (CaS) receptor controls plasma Ca2+ levels and contributes to nutrient-dependent maintenance and metabolism of diverse organs. Allosteric modulation of the CaS receptor corrects disorders of calcium homeostasis. Here, we report the cryogenic-electron microscopy reconstructions of a near-full-length CaS receptor in the absence and presence of allosteric modulators. Activation of the homodimeric CaS receptor requires a break in the transmembrane 6 (TM6) helix of each subunit, which facilitates the formation of a TM6-mediated homodimer interface and expansion of homodimer interactions. This transformation in TM6 occurs without a positive allosteric modulator. Two modulators with opposite functional roles bind to overlapping sites within the transmembrane domain through common interactions, acting to stabilize distinct rotamer conformations of key residues on the TM6 helix. The positive modulator reinforces TM6 distortion and maximizes subunit contact to enhance receptor activity, while the negative modulator strengthens an intact TM6 to dampen receptor function. In both active and inactive states, the receptor displays symmetrical transmembrane conformations that are consistent with its homodimeric assembly.


Assuntos
Cálcio/metabolismo , Regulação da Expressão Gênica/fisiologia , Homeostase/fisiologia , Receptores de Detecção de Cálcio/metabolismo , Microscopia Crioeletrônica , Células HEK293 , Humanos , Modelos Moleculares , Conformação Proteica , Domínios Proteicos , Receptores de Detecção de Cálcio/genética , Transdução de Sinais
8.
Nature ; 549(7670): 60-65, 2017 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-28737760

RESUMO

AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid)-subtype ionotropic glutamate receptors mediate fast excitatory neurotransmission throughout the central nervous system. Gated by the neurotransmitter glutamate, AMPA receptors are critical for synaptic strength, and dysregulation of AMPA receptor-mediated signalling is linked to numerous neurological diseases. Here we use cryo-electron microscopy to solve the structures of AMPA receptor-auxiliary subunit complexes in the apo, antagonist- and agonist-bound states and determine the iris-like mechanism of ion channel opening. The ion channel selectivity filter is formed by the extended portions of the re-entrant M2 loops, while the helical portions of M2 contribute to extensive hydrophobic interfaces between AMPA receptor subunits in the ion channel. We show how the permeation pathway changes upon channel opening and identify conformational changes throughout the entire AMPA receptor that accompany activation and desensitization. Our findings provide a framework for understanding gating across the family of ionotropic glutamate receptors and the role of AMPA receptors in excitatory neurotransmission.


Assuntos
Microscopia Crioeletrônica , Ativação do Canal Iônico , Receptores de AMPA/química , Receptores de AMPA/ultraestrutura , Animais , Canais de Cálcio/metabolismo , Claudinas/metabolismo , Células HEK293 , Humanos , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Modelos Moleculares , Conformação Proteica , Subunidades Proteicas/agonistas , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Ratos , Receptores de AMPA/agonistas , Receptores de AMPA/antagonistas & inibidores , Transmissão Sináptica
9.
Nature ; 517(7532): 44-9, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25470061

RESUMO

Ryanodine receptors (RyRs) mediate the rapid release of calcium (Ca(2+)) from intracellular stores into the cytosol, which is essential for numerous cellular functions including excitation-contraction coupling in muscle. Lack of sufficient structural detail has impeded understanding of RyR gating and regulation. Here we report the closed-state structure of the 2.3-megadalton complex of the rabbit skeletal muscle type 1 RyR (RyR1), solved by single-particle electron cryomicroscopy at an overall resolution of 4.8 Å. We fitted a polyalanine-level model to all 3,757 ordered residues in each protomer, defining the transmembrane pore in unprecedented detail and placing all cytosolic domains as tertiary folds. The cytosolic assembly is built on an extended α-solenoid scaffold connecting key regulatory domains to the pore. The RyR1 pore architecture places it in the six-transmembrane ion channel superfamily. A unique domain inserted between the second and third transmembrane helices interacts intimately with paired EF-hands originating from the α-solenoid scaffold, suggesting a mechanism for channel gating by Ca(2+).


Assuntos
Canal de Liberação de Cálcio do Receptor de Rianodina/química , Canal de Liberação de Cálcio do Receptor de Rianodina/ultraestrutura , Animais , Cálcio/deficiência , Cálcio/metabolismo , Cálcio/farmacologia , Membrana Celular/metabolismo , Microscopia Crioeletrônica , Citosol/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Músculo Esquelético/química , Estrutura Terciária de Proteína , Coelhos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Proteínas de Ligação a Tacrolimo/química , Proteínas de Ligação a Tacrolimo/metabolismo
10.
Proc Natl Acad Sci U S A ; 115(51): 12985-12990, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30509977

RESUMO

Membrane proteins function in native cell membranes, but extraction into isolated particles is needed for many biochemical and structural analyses. Commonly used detergent-extraction methods destroy naturally associated lipid bilayers. Here, we devised a detergent-free method for preparing cell-membrane nanoparticles to study the multidrug exporter AcrB, by cryo-EM at 3.2-Å resolution. We discovered a remarkably well-organized lipid-bilayer structure associated with transmembrane domains of the AcrB trimer. This bilayer patch comprises 24 lipid molecules; inner leaflet chains are packed in a hexagonal array, whereas the outer leaflet has highly irregular but ordered packing. Protein side chains interact with both leaflets and participate in the hexagonal pattern. We suggest that the lipid bilayer supports and harmonizes peristaltic motions through AcrB trimers. In AcrB D407A, a putative proton-relay mutant, lipid bilayer buttresses protein interactions lost in crystal structures after detergent-solubilization. Our detergent-free system preserves lipid-protein interactions for visualization and should be broadly applicable.


Assuntos
Membrana Celular/metabolismo , Detergentes/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/química , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Membrana Celular/química , Cristalografia por Raios X , Detergentes/química , Escherichia coli/crescimento & desenvolvimento , Nanopartículas/química , Nanopartículas/metabolismo , Conformação Proteica
11.
Nature ; 503(7477): 539-43, 2013 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-24185006

RESUMO

Hepatitis C virus (HCV) and classical swine fever virus (CSFV) messenger RNAs contain related (HCV-like) internal ribosome entry sites (IRESs) that promote 5'-end independent initiation of translation, requiring only a subset of the eukaryotic initiation factors (eIFs) needed for canonical initiation on cellular mRNAs. Initiation on HCV-like IRESs relies on their specific interaction with the 40S subunit, which places the initiation codon into the P site, where it directly base-pairs with eIF2-bound initiator methionyl transfer RNA to form a 48S initiation complex. However, all HCV-like IRESs also specifically interact with eIF3 (refs 2, 5-7, 9-12), but the role of this interaction in IRES-mediated initiation has remained unknown. During canonical initiation, eIF3 binds to the 40S subunit as a component of the 43S pre-initiation complex, and comparison of the ribosomal positions of eIF3 and the HCV IRES revealed that they overlap, so that their rearrangement would be required for formation of ribosomal complexes containing both components. Here we present a cryo-electron microscopy reconstruction of a 40S ribosomal complex containing eIF3 and the CSFV IRES. Remarkably, although the position and interactions of the CSFV IRES with the 40S subunit in this complex are similar to those of the HCV IRES in the 40S-IRES binary complex, eIF3 is completely displaced from its ribosomal position in the 43S complex, and instead interacts through its ribosome-binding surface exclusively with the apical region of domain III of the IRES. Our results suggest a role for the specific interaction of HCV-like IRESs with eIF3 in preventing ribosomal association of eIF3, which could serve two purposes: relieving the competition between the IRES and eIF3 for a common binding site on the 40S subunit, and reducing formation of 43S complexes, thereby favouring translation of viral mRNAs.


Assuntos
Vírus da Febre Suína Clássica/genética , Fator de Iniciação 3 em Eucariotos/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Sequências Reguladoras de Ácido Ribonucleico/genética , Subunidades Ribossômicas Menores de Eucariotos/metabolismo , Ribossomos/metabolismo , Animais , Ligação Competitiva , Microscopia Crioeletrônica , Fator de Iniciação 3 em Eucariotos/química , Fator de Iniciação 3 em Eucariotos/ultraestrutura , Humanos , Modelos Moleculares , Biossíntese de Proteínas , Coelhos , Subunidades Ribossômicas Menores de Eucariotos/química , Subunidades Ribossômicas Menores de Eucariotos/ultraestrutura , Ribossomos/química , Ribossomos/ultraestrutura
12.
Nature ; 494(7437): 385-9, 2013 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-23395961

RESUMO

Ribosomes, the protein factories of living cells, translate genetic information carried by messenger RNAs into proteins, and are thus involved in virtually all aspects of cellular development and maintenance. The few available structures of the eukaryotic ribosome reveal that it is more complex than its prokaryotic counterpart, owing mainly to the presence of eukaryote-specific ribosomal proteins and additional ribosomal RNA insertions, called expansion segments. The structures also differ among species, partly in the size and arrangement of these expansion segments. Such differences are extreme in kinetoplastids, unicellular eukaryotic parasites often infectious to humans. Here we present a high-resolution cryo-electron microscopy structure of the ribosome of Trypanosoma brucei, the parasite that is transmitted by the tsetse fly and that causes African sleeping sickness. The atomic model reveals the unique features of this ribosome, characterized mainly by the presence of unusually large expansion segments and ribosomal-protein extensions leading to the formation of four additional inter-subunit bridges. We also find additional rRNA insertions, including one large rRNA domain that is not found in other eukaryotes. Furthermore, the structure reveals the five cleavage sites of the kinetoplastid large ribosomal subunit (LSU) rRNA chain, which is known to be cleaved uniquely into six pieces, and suggests that the cleavage is important for the maintenance of the T. brucei ribosome in the observed structure. We discuss several possible implications of the large rRNA expansion segments for the translation-regulation process. The structure could serve as a basis for future experiments aimed at understanding the functional importance of these kinetoplastid-specific ribosomal features in protein-translation regulation, an essential step towards finding effective and safe kinetoplastid-specific drugs.


Assuntos
Microscopia Crioeletrônica , Ribossomos/ultraestrutura , Trypanosoma brucei brucei/citologia , Trypanosoma brucei brucei/ultraestrutura , Modelos Biológicos , Modelos Moleculares , Conformação Molecular , Biossíntese de Proteínas , RNA de Protozoário/genética , RNA de Protozoário/metabolismo , RNA Ribossômico/genética , RNA Ribossômico/metabolismo , Ribossomos/química , Ribossomos/genética , Trypanosoma brucei brucei/química , Trypanosoma brucei brucei/genética , Leveduras/química
13.
Proc Natl Acad Sci U S A ; 113(43): 12174-12179, 2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27791004

RESUMO

Ribosomes of trypanosomatids, a family of protozoan parasites causing debilitating human diseases, possess multiply fragmented rRNAs that together are analogous to 28S rRNA, unusually large rRNA expansion segments, and r-protein variations compared with other eukaryotic ribosomes. To investigate the architecture of the trypanosomatid ribosomes, we determined the 2.5-Å structure of the Trypanosoma cruzi ribosome large subunit by single-particle cryo-EM. Examination of this structure and comparative analysis of the yeast ribosomal assembly pathway allowed us to develop a stepwise assembly model for the eight pieces of the large subunit rRNAs and a number of ancillary "glue" proteins. This model can be applied to the characterization of Trypanosoma brucei and Leishmania spp. ribosomes as well. Together with other details, our atomic-level structure may provide a foundation for structure-based design of antitrypanosome drugs.


Assuntos
Subunidades Ribossômicas Maiores de Eucariotos/ultraestrutura , Ribossomos/ultraestrutura , Trypanosoma cruzi/química , Doença de Chagas/tratamento farmacológico , Doença de Chagas/parasitologia , Microscopia Crioeletrônica , Cristalografia por Raios X , Humanos , RNA Ribossômico/genética , RNA Ribossômico/ultraestrutura , Proteínas Ribossômicas/química , Proteínas Ribossômicas/genética , Subunidades Ribossômicas Maiores de Eucariotos/química , Subunidades Ribossômicas Maiores de Eucariotos/genética , Ribossomos/genética , Trypanosoma cruzi/genética , Trypanosoma cruzi/ultraestrutura
14.
Nucleic Acids Res ; 42(5): 3409-18, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24335085

RESUMO

Eukaryotic translation termination results from the complex functional interplay between two release factors, eRF1 and eRF3, in which GTP hydrolysis by eRF3 couples codon recognition with peptidyl-tRNA hydrolysis by eRF1. Here, we present a cryo-electron microscopy structure of pre-termination complexes associated with eRF1•eRF3•GDPNP at 9.7 -Šresolution, which corresponds to the initial pre-GTP hydrolysis stage of factor attachment and stop codon recognition. It reveals the ribosomal positions of eRFs and provides insights into the mechanisms of stop codon recognition and triggering of eRF3's GTPase activity.


Assuntos
Terminação Traducional da Cadeia Peptídica , Fatores de Terminação de Peptídeos/química , Ribossomos/química , Códon de Terminação , GTP Fosfo-Hidrolases/metabolismo , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/química , Humanos , Modelos Moleculares , Fatores de Terminação de Peptídeos/metabolismo
15.
J Struct Biol ; 190(3): 348-59, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25913484

RESUMO

Image formation in bright field electron microscopy can be described with the help of the contrast transfer function (CTF). In this work the authors describe the "CTF Estimation Challenge", called by the Madrid Instruct Image Processing Center (I2PC) in collaboration with the National Center for Macromolecular Imaging (NCMI) at Houston. Correcting for the effects of the CTF requires accurate knowledge of the CTF parameters, but these have often been difficult to determine. In this challenge, researchers have had the opportunity to test their ability in estimating some of the key parameters of the electron microscope CTF on a large micrograph data set produced by well-known laboratories on a wide set of experimental conditions. This work presents the first analysis of the results of the CTF Estimation Challenge, including an assessment of the performance of the different software packages under different conditions, so as to identify those areas of research where further developments would be desirable in order to achieve high-resolution structural information.


Assuntos
Substâncias Macromoleculares/química , Microscopia Eletrônica/métodos , Algoritmos , Processamento de Imagem Assistida por Computador/métodos , Software
16.
Proc Natl Acad Sci U S A ; 109(45): 18413-8, 2012 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-23091004

RESUMO

Eukaryotic translation termination results from the complex functional interplay between two eukaryotic release factors, eRF1 and eRF3, and the ribosome, in which GTP hydrolysis by eRF3 couples codon recognition with peptidyl-tRNA hydrolysis by eRF1. Here, using cryo-electron microscopy (cryo-EM) and flexible fitting, we determined the structure of eRF1-eRF3-guanosine 5'-[ß,γ-imido]triphosphate (GMPPNP)-bound ribosomal pretermination complex (pre-TC), which corresponds to the initial, pre-GTP hydrolysis stage of factor attachment. Our results show that eukaryotic translation termination involves a network of interactions between the two release factors and the ribosome. Our structure provides mechanistic insight into the coordination between GTP hydrolysis by eRF3 and subsequent peptide release by eRF1.


Assuntos
Microscopia Crioeletrônica , Mamíferos/metabolismo , Terminação Traducional da Cadeia Peptídica , Fatores de Terminação de Peptídeos/ultraestrutura , Animais , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Humanos , Modelos Moleculares , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica , Aminoacil-RNA de Transferência/química , Aminoacil-RNA de Transferência/metabolismo , Coelhos , Ribossomos/metabolismo , Ribossomos/ultraestrutura , Saccharomyces cerevisiae
17.
J Struct Biol ; 181(2): 190-4, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23228487

RESUMO

Affinity grids (AG) are specialized EM grids that bind macromolecular complexes containing tagged proteins to obtain maximum occupancy for structural analysis through single-particle EM. In this study, utilizing AG, we show that His-tagged activated PKC ßII binds to the small ribosomal subunit (40S). We reconstructed a cryo-EM map which shows that PKC ßII interacts with RACK1, a seven-bladed ß-propeller protein present on the 40S and binds in two different regions close to blades 3 and 4 of RACK1. This study is a first step in understanding the molecular framework of PKC ßII/RACK1 interaction and its role in translation.


Assuntos
Microscopia Crioeletrônica/métodos , Proteínas de Ligação ao GTP/química , Modelos Moleculares , Proteínas de Neoplasias/química , Biossíntese de Proteínas/fisiologia , Conformação Proteica , Proteína Quinase C/química , Receptores de Superfície Celular/química , Subunidades Ribossômicas Menores de Eucariotos/metabolismo , Microscopia Crioeletrônica/instrumentação , Proteínas de Ligação ao GTP/metabolismo , Humanos , Proteínas de Neoplasias/metabolismo , Proteína Quinase C/metabolismo , Receptores de Quinase C Ativada , Receptores de Superfície Celular/metabolismo
18.
Nucleic Acids Res ; 39(7): 2845-54, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21131279

RESUMO

Group II self-splicing introns are phylogenetically diverse retroelements that are widely held to be the ancestors of spliceosomal introns and retrotransposons that insert into DNA. Folding of group II intron RNA is often guided by an intron-encoded protein to form a catalytically active ribonucleoprotein (RNP) complex that plays a key role in the activity of the intron. To date, possible structural differences between the intron RNP in its precursor and spliced forms remain unexplored. In this work, we have trapped the native Lactococcus lactis group II intron RNP complex in its precursor form, by deleting the adenosine nucleophile that initiates splicing. Sedimentation velocity, size-exclusion chromatography and cryo-electron microscopy provide the first glimpse of the intron RNP precursor as a large, loosely packed structure. The dimensions contrast with those of compact spliced introns, implying that the RNP undergoes a dramatic conformational change to achieve the catalytically active state.


Assuntos
Íntrons , Precursores de RNA/química , RNA Catalítico/química , Ribonucleoproteínas/química , Lactococcus lactis/genética , Conformação Molecular , Precursores de RNA/isolamento & purificação , RNA Catalítico/isolamento & purificação , Ribonucleoproteínas/isolamento & purificação , Ribonucleoproteínas/ultraestrutura
19.
Proc Natl Acad Sci U S A ; 106(4): 1063-8, 2009 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-19122150

RESUMO

In translation, elongation factor Tu (EF-Tu) molecules deliver aminoacyl-tRNAs to the mRNA-programmed ribosome. The GTPase activity of EF-Tu is triggered by ribosome-induced conformational changes of the factor that play a pivotal role in the selection of the cognate aminoacyl-tRNAs. We present a 6.7-A cryo-electron microscopy map of the aminoacyl-tRNA x EF-Tu x GDP x kirromycin-bound Escherichia coli ribosome, together with an atomic model of the complex obtained through molecular dynamics flexible fitting. The model reveals the conformational changes in the conserved GTPase switch regions of EF-Tu that trigger hydrolysis of GTP, along with key interactions, including those between the sarcin-ricin loop and the P loop of EF-Tu, and between the effector loop of EF-Tu and a conserved region of the 16S rRNA. Our data suggest that GTP hydrolysis on EF-Tu is controlled through a hydrophobic gate mechanism.


Assuntos
Escherichia coli/metabolismo , Guanosina Trifosfato/metabolismo , Fator Tu de Elongação de Peptídeos/química , Ribossomos/metabolismo , Microscopia Crioeletrônica , Ativação Enzimática , Escherichia coli/enzimologia , Proteínas de Escherichia coli/metabolismo , Histidina/metabolismo , Hidrólise , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Fator Tu de Elongação de Peptídeos/ultraestrutura , Estrutura Secundária de Proteína , RNA de Transferência/metabolismo , Proteínas Ribossômicas/metabolismo , Ribossomos/química , Ribossomos/ultraestrutura , Transdução de Sinais
20.
Nature ; 427(6977): 808-14, 2004 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-14985753

RESUMO

Cotranslational translocation of proteins across or into membranes is a vital process in all kingdoms of life. It requires that the translating ribosome be targeted to the membrane by the signal recognition particle (SRP), an evolutionarily conserved ribonucleoprotein particle. SRP recognizes signal sequences of nascent protein chains emerging from the ribosome. Subsequent binding of SRP leads to a pause in peptide elongation and to the ribosome docking to the membrane-bound SRP receptor. Here we present the structure of a targeting complex consisting of mammalian SRP bound to an active 80S ribosome carrying a signal sequence. This structure, solved to 12 A by cryo-electron microscopy, enables us to generate a molecular model of SRP in its functional conformation. The model shows how the S domain of SRP contacts the large ribosomal subunit at the nascent chain exit site to bind the signal sequence, and that the Alu domain reaches into the elongation-factor-binding site of the ribosome, explaining its elongation arrest activity.


Assuntos
Elongação Traducional da Cadeia Peptídica , Ribossomos/metabolismo , Partícula de Reconhecimento de Sinal/química , Partícula de Reconhecimento de Sinal/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Microscopia Crioeletrônica , Mamíferos , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Sinais Direcionadores de Proteínas/genética , Sinais Direcionadores de Proteínas/fisiologia , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Ribossomos/química , Ribossomos/ultraestrutura , Partícula de Reconhecimento de Sinal/genética , Partícula de Reconhecimento de Sinal/ultraestrutura , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA