Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Nature ; 585(7823): 129-134, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32848250

RESUMO

Transmembrane channels and pores have key roles in fundamental biological processes1 and in biotechnological applications such as DNA nanopore sequencing2-4, resulting in considerable interest in the design of pore-containing proteins. Synthetic amphiphilic peptides have been found to form ion channels5,6, and there have been recent advances in de novo membrane protein design7,8 and in redesigning naturally occurring channel-containing proteins9,10. However, the de novo design of stable, well-defined transmembrane protein pores that are capable of conducting ions selectively or are large enough to enable the passage of small-molecule fluorophores remains an outstanding challenge11,12. Here we report the computational design of protein pores formed by two concentric rings of α-helices that are stable and monodisperse in both their water-soluble and their transmembrane forms. Crystal structures of the water-soluble forms of a 12-helical pore and a 16-helical pore closely match the computational design models. Patch-clamp electrophysiology experiments show that, when expressed in insect cells, the transmembrane form of the 12-helix pore enables the passage of ions across the membrane with high selectivity for potassium over sodium; ion passage is blocked by specific chemical modification at the pore entrance. When incorporated into liposomes using in vitro protein synthesis, the transmembrane form of the 16-helix pore-but not the 12-helix pore-enables the passage of biotinylated Alexa Fluor 488. A cryo-electron microscopy structure of the 16-helix transmembrane pore closely matches the design model. The ability to produce structurally and functionally well-defined transmembrane pores opens the door to the creation of designer channels and pores for a wide variety of applications.


Assuntos
Simulação por Computador , Genes Sintéticos/genética , Canais Iônicos/química , Canais Iônicos/genética , Modelos Moleculares , Biologia Sintética , Linhagem Celular , Microscopia Crioeletrônica , Cristalografia por Raios X , Condutividade Elétrica , Escherichia coli/genética , Escherichia coli/metabolismo , Hidrazinas , Canais Iônicos/metabolismo , Transporte de Íons , Lipossomos/metabolismo , Técnicas de Patch-Clamp , Porinas/química , Porinas/genética , Porinas/metabolismo , Engenharia de Proteínas , Estrutura Secundária de Proteína , Solubilidade , Água/química
2.
Nature ; 565(7737): 106-111, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30568301

RESUMO

Specificity of interactions between two DNA strands, or between protein and DNA, is often achieved by varying bases or side chains coming off the DNA or protein backbone-for example, the bases participating in Watson-Crick pairing in the double helix, or the side chains contacting DNA in TALEN-DNA complexes. By contrast, specificity of protein-protein interactions usually involves backbone shape complementarity1, which is less modular and hence harder to generalize. Coiled-coil heterodimers are an exception, but the restricted geometry of interactions across the heterodimer interface (primarily at the heptad a and d positions2) limits the number of orthogonal pairs that can be created simply by varying side-chain interactions3,4. Here we show that protein-protein interaction specificity can be achieved using extensive and modular side-chain hydrogen-bond networks. We used the Crick generating equations5 to produce millions of four-helix backbones with varying degrees of supercoiling around a central axis, identified those accommodating extensive hydrogen-bond networks, and used Rosetta to connect pairs of helices with short loops and to optimize the remainder of the sequence. Of 97 such designs expressed in Escherichia coli, 65 formed constitutive heterodimers, and the crystal structures of four designs were in close agreement with the computational models and confirmed the designed hydrogen-bond networks. In cells, six heterodimers were fully orthogonal, and in vitro-following mixing of 32 chains from 16 heterodimer designs, denaturation in 5 M guanidine hydrochloride and reannealing-almost all of the interactions observed by native mass spectrometry were between the designed cognate pairs. The ability to design orthogonal protein heterodimers should enable sophisticated protein-based control logic for synthetic biology, and illustrates that nature has not fully explored the possibilities for programmable biomolecular interaction modalities.


Assuntos
Simulação por Computador , Engenharia de Proteínas , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas/química , Proteínas/metabolismo , DNA/química , DNA/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Guanidina/farmacologia , Ligação de Hidrogênio , Modelos Moleculares , Ligação Proteica , Desnaturação Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína , Proteínas/genética
3.
Nature ; 557(7706): 590-594, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29769724

RESUMO

Potassium-sensitive hypokalaemic and normokalaemic periodic paralysis are inherited skeletal muscle diseases characterized by episodes of flaccid muscle weakness1,2. They are caused by single mutations in positively charged residues ('gating charges') in the S4 transmembrane segment of the voltage sensor of the voltage-gated sodium channel Nav1.4 or the calcium channel Cav1.11,2. Mutations of the outermost gating charges (R1 and R2) cause hypokalaemic periodic paralysis1,2 by creating a pathogenic gating pore in the voltage sensor through which cations leak in the resting state3,4. Mutations of the third gating charge (R3) cause normokalaemic periodic paralysis 5 owing to cation leak in both activated and inactivated states 6 . Here we present high-resolution structures of the model bacterial sodium channel NavAb with the analogous gating-charge mutations7,8, which have similar functional effects as in the human channels. The R2G and R3G mutations have no effect on the backbone structures of the voltage sensor, but they create an aqueous cavity near the hydrophobic constriction site that controls gating charge movement through the voltage sensor. The R3G mutation extends the extracellular aqueous cleft through the entire length of the activated voltage sensor, creating an aqueous path through the membrane. Conversely, molecular modelling shows that the R2G mutation creates a continuous aqueous path through the membrane only in the resting state. Crystal structures of NavAb(R2G) in complex with guanidinium define a potential drug target site. Molecular dynamics simulations illustrate the mechanism of Na+ permeation through the mutant gating pore in concert with conformational fluctuations of the gating charge R4. Our results reveal pathogenic mechanisms of periodic paralysis at the atomic level and suggest designs of drugs that may prevent ionic leak and provide symptomatic relief from hypokalaemic and normokalaemic periodic paralysis.


Assuntos
Ativação do Canal Iônico , Canal de Sódio Disparado por Voltagem NAV1.4/química , Canal de Sódio Disparado por Voltagem NAV1.4/metabolismo , Paralisias Periódicas Familiares/metabolismo , Sítios de Ligação , Condutividade Elétrica , Guanidina/metabolismo , Humanos , Paralisia Periódica Hipopotassêmica/genética , Paralisia Periódica Hipopotassêmica/metabolismo , Ativação do Canal Iônico/genética , Simulação de Dinâmica Molecular , Mutação , Canal de Sódio Disparado por Voltagem NAV1.4/genética , Paralisias Periódicas Familiares/genética , Sódio/metabolismo , Termodinâmica
4.
Nature ; 562(7726): 286-290, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30283133

RESUMO

Membrane-bound O-acyltransferases (MBOATs) are a superfamily of integral transmembrane enzymes that are found in all kingdoms of life1. In bacteria, MBOATs modify protective cell-surface polymers. In vertebrates, some MBOAT enzymes-such as acyl-coenzyme A:cholesterol acyltransferase and diacylglycerol acyltransferase 1-are responsible for lipid biosynthesis or phospholipid remodelling2,3. Other MBOATs, including porcupine, hedgehog acyltransferase and ghrelin acyltransferase, catalyse essential lipid modifications of secreted proteins such as Wnt, hedgehog and ghrelin, respectively4-10. Although many MBOAT proteins are important drug targets, little is known about their molecular architecture and functional mechanisms. Here we present crystal structures of DltB, an MBOAT responsible for the D-alanylation of cell-wall teichoic acid in Gram-positive bacteria11-16, both alone and in complex with the D-alanyl donor protein DltC. DltB contains a ring of 11 peripheral transmembrane helices, which shield a highly conserved extracellular structural funnel extending into the middle of the lipid bilayer. The conserved catalytic histidine residue is located at the bottom of this funnel and is connected to the intracellular DltC through a narrow tunnel. Mutation of either the catalytic histidine or the DltC-binding site of DltB abolishes the D-alanylation of lipoteichoic acid and sensitizes the Gram-positive bacterium Bacillus subtilis to cell-wall stress, which suggests cross-membrane catalysis involving the tunnel. Structure-guided sequence comparison among DltB and vertebrate MBOATs reveals a conserved structural core and suggests that MBOATs from different organisms have similar catalytic mechanisms. Our structures provide a template for understanding structure-function relationships in MBOATs and for developing therapeutic MBOAT inhibitors.


Assuntos
Aciltransferases/química , Aciltransferases/metabolismo , Bicamadas Lipídicas/metabolismo , Aciltransferases/genética , Sequência de Aminoácidos , Animais , Bacillus subtilis/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Biocatálise , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Parede Celular/metabolismo , Sequência Conservada , Cristalografia por Raios X , Histidina/genética , Histidina/metabolismo , Bicamadas Lipídicas/química , Lipopolissacarídeos/metabolismo , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Modelos Moleculares , Mutação , Ligação Proteica , Relação Estrutura-Atividade , Ácidos Teicoicos/metabolismo
5.
Proteins ; 90(10): 1800-1806, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35305033

RESUMO

Membrane transport proteins, which include transporters and channels, are delicate protein machineries that mediate the exchange of a variety of substances across biomembranes. Accumulated structural and functional knowledge allows for the de novo design of transport proteins with new structures that do not exist in nature. Analysis based on these novel proteins provides new insights into the principles that govern protein assembly, conformational change, and substrate recognition. Here, we review the advances in the de novo design of transporters and channels over recent years and highlight the challenges and opportunities in this field.


Assuntos
Proteínas de Transporte , Proteínas de Membrana Transportadoras , Transporte Biológico , Proteínas de Transporte/química , Proteínas de Membrana Transportadoras/química
6.
Nature ; 525(7568): 212-217, 2015 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-26280335

RESUMO

Dysfunction of the intramembrane protease γ-secretase is thought to cause Alzheimer's disease, with most mutations derived from Alzheimer's disease mapping to the catalytic subunit presenilin 1 (PS1). Here we report an atomic structure of human γ-secretase at 3.4 Šresolution, determined by single-particle cryo-electron microscopy. Mutations derived from Alzheimer's disease affect residues at two hotspots in PS1, each located at the centre of a distinct four transmembrane segment (TM) bundle. TM2 and, to a lesser extent, TM6 exhibit considerable flexibility, yielding a plastic active site and adaptable surrounding elements. The active site of PS1 is accessible from the convex side of the TM horseshoe, suggesting considerable conformational changes in nicastrin extracellular domain after substrate recruitment. Component protein APH-1 serves as a scaffold, anchoring the lone transmembrane helix from nicastrin and supporting the flexible conformation of PS1. Ordered phospholipids stabilize the complex inside the membrane. Our structure serves as a molecular basis for mechanistic understanding of γ-secretase function.


Assuntos
Secretases da Proteína Precursora do Amiloide/química , Secretases da Proteína Precursora do Amiloide/ultraestrutura , Microscopia Crioeletrônica , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/ultraestrutura , Presenilina-1/química , Presenilina-1/ultraestrutura , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Sítios de Ligação , Humanos , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Mutação , Presenilina-1/genética , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo
7.
Nature ; 512(7513): 166-170, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-25043039

RESUMO

The γ-secretase complex, comprising presenilin 1 (PS1), PEN-2, APH-1 and nicastrin, is a membrane-embedded protease that controls a number of important cellular functions through substrate cleavage. Aberrant cleavage of the amyloid precursor protein (APP) results in aggregation of amyloid-ß, which accumulates in the brain and consequently causes Alzheimer's disease. Here we report the three-dimensional structure of an intact human γ-secretase complex at 4.5 Å resolution, determined by cryo-electron-microscopy single-particle analysis. The γ-secretase complex comprises a horseshoe-shaped transmembrane domain, which contains 19 transmembrane segments (TMs), and a large extracellular domain (ECD) from nicastrin, which sits immediately above the hollow space formed by the TM horseshoe. Intriguingly, nicastrin ECD is structurally similar to a large family of peptidases exemplified by the glutamate carboxypeptidase PSMA. This structure serves as an important basis for understanding the functional mechanisms of the γ-secretase complex.


Assuntos
Secretases da Proteína Precursora do Amiloide/química , Modelos Moleculares , Microscopia Crioeletrônica , Cristalografia por Raios X , Humanos , Estrutura Terciária de Proteína
8.
Nature ; 483(7391): 632-6, 2012 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-22407317

RESUMO

Food-borne hemorrhagic Escherichia coli, exemplified by the strains O157:H7 and O104:H4 (refs 1, 2), require elaborate acid-resistance systems (ARs) to survive the extremely acidic environment such as the stomach (pH ≈ 2). AR2 expels intracellular protons through the decarboxylation of L-glutamate (Glu) in the cytoplasm and exchange of the reaction product γ-aminobutyric acid (GABA) with extracellular Glu. The latter process is mediated by the Glu-GABA antiporter GadC, a representative member of the amino-acid-polyamine-organocation superfamily of membrane transporters. The functional mechanism of GadC remains largely unknown. Here we show, with the use of an in vitro proteoliposome-based assay, that GadC transports GABA/Glu only under acidic conditions, with no detectable activity at pH values higher than 6.5. We determined the crystal structure of E. coli GadC at 3.1 Å resolution under basic conditions. GadC, comprising 12 transmembrane segments (TMs), exists in a closed state, with its carboxy-terminal domain serving as a plug to block an otherwise inward-open conformation. Structural and biochemical analyses reveal the essential transport residues, identify the transport path and suggest a conserved transport mechanism involving the rigid-body rotation of a helical bundle for GadC and other amino acid antiporters.


Assuntos
Antiporters/química , Antiporters/metabolismo , Escherichia coli O157/química , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Ácido Glutâmico/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Ácido gama-Aminobutírico/metabolismo , Transporte Biológico , Cristalografia por Raios X , Modelos Moleculares , Estrutura Terciária de Proteína , Proteolipídeos/metabolismo , Rotação , Relação Estrutura-Atividade
9.
Proc Natl Acad Sci U S A ; 111(5): 1813-8, 2014 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-24449903

RESUMO

Vitamin C, also known as ascorbate, is required in numerous essential metabolic reactions in eukaryotes. The eukaryotic ascorbate-dependent oxidoreductase cytochrome b561 (Cyt b561), a family of highly conserved transmembrane enzymes, plays an important role in ascorbate recycling and iron absorption. Although Cyt b561 was identified four decades ago, its atomic structure and functional mechanism remain largely unknown. Here, we report the high-resolution crystal structures of cytochrome b561 from Arabidopsis thaliana in both substrate-free and substrate-bound states. Cyt b561 forms a homodimer, with each protomer consisting of six transmembrane helices and two heme groups. The negatively charged substrate ascorbate, or monodehydroascorbate, is enclosed in a positively charged pocket on either side of the membrane. Two highly conserved amino acids, Lys(81) and His(106), play an essential role in substrate recognition and catalysis. Our structural and biochemical analyses allow the proposition of a general electron transfer mechanism for members of the Cyt b561 family.


Assuntos
Arabidopsis/enzimologia , Ácido Ascórbico/metabolismo , Membrana Celular/enzimologia , Grupo dos Citocromos b/química , Grupo dos Citocromos b/metabolismo , Oxirredutases/química , Oxirredutases/metabolismo , Aminoácidos/metabolismo , Sítios de Ligação , Sequência Conservada , Elétrons , Células Eucarióticas/citologia , Células Eucarióticas/enzimologia , Heme/metabolismo , Modelos Biológicos , Modelos Moleculares
10.
Proc Natl Acad Sci U S A ; 111(37): 13349-54, 2014 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-25197054

RESUMO

γ-Secretase is an intramembrane protease responsible for the generation of amyloid-ß (Aß) peptides. Aberrant accumulation of Aß leads to the formation of amyloid plaques in the brain of patients with Alzheimer's disease. Nicastrin is the putative substrate-recruiting component of the γ-secretase complex. No atomic-resolution structure had been identified on γ-secretase or any of its four components, hindering mechanistic understanding of γ-secretase function. Here we report the crystal structure of nicastrin from Dictyostelium purpureum at 1.95-Å resolution. The extracellular domain of nicastrin contains a large lobe and a small lobe. The large lobe of nicastrin, thought to be responsible for substrate recognition, associates with the small lobe through a hydrophobic pivot at the center. The putative substrate-binding pocket is shielded from the small lobe by a lid, which blocks substrate entry. These structural features suggest a working model of nicastrin function. Analysis of nicastrin structure provides insights into the assembly and architecture of the γ-secretase complex.


Assuntos
Secretases da Proteína Precursora do Amiloide/química , Dictyostelium/enzimologia , Glicoproteínas de Membrana/química , Sítios de Ligação , Cristalografia por Raios X , Humanos , Modelos Moleculares , Estrutura Terciária de Proteína , Especificidade por Substrato
11.
Nature ; 462(7272): 467-72, 2009 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-19940917

RESUMO

FocA is a representative member of the formate-nitrite transporter family, which transports short-chain acids in bacteria, archaea, fungi, algae and parasites. The structure and transport mechanism of the formate-nitrite transporter family remain unknown. Here we report the crystal structure of Escherichia coli FocA at 2.25 A resolution. FocA forms a symmetric pentamer, with each protomer consisting of six transmembrane segments. Despite a lack of sequence homology, the overall structure of the FocA protomer closely resembles that of aquaporin and strongly argues that FocA is a channel, rather than a transporter. Structural analysis identifies potentially important channel residues, defines the channel path and reveals two constriction sites. Unlike aquaporin, FocA is impermeable to water but allows the passage of formate. A structural and biochemical investigation provides mechanistic insights into the channel activity of FocA.


Assuntos
Aquaporinas/química , Proteínas de Escherichia coli/química , Proteínas de Membrana Transportadoras/química , Aquaporinas/metabolismo , Cristalografia por Raios X , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Formiatos/metabolismo , Lipossomos/química , Lipossomos/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Modelos Moleculares , Mimetismo Molecular , Mutação , Permeabilidade , Estrutura Quaternária de Proteína , Relação Estrutura-Atividade , Água/análise , Água/metabolismo
12.
J Biol Chem ; 288(21): 15148-53, 2013 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-23589309

RESUMO

GadC, a central component of the Escherichia coli acid resistance system, is a Glu/GABA antiporter. A previous structural study and biochemical characterization showed that GadC exhibits a stringent pH dependence for substrate transport, with no detectable activity at pH values above 6.5. However, the substrate selectivity and the mechanism of pH-dependent transport activity of GadC remain enigmatic. In this study, we demonstrate that GadC selectively transports Glu with no net charge and GABA with a positive charge. A C-plug-truncated variant of GadC (residues 1-470) transported Gln (a mimic of Glu with no net charge), but not Glu, even at pH 8.0. The pH-dependent transport of Gln by this GadC variant was shifted ~1 unit toward a higher pH compared with Glu transport. Taken together, the results identify the substrate selectivity for GadC and show that the protonation states of substrates are crucial for transport.


Assuntos
Transporte Biológico Ativo/fisiologia , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Glutamina/metabolismo , Proteínas de Membrana/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Glutamina/genética , Concentração de Íons de Hidrogênio , Proteínas de Membrana/genética , Especificidade por Substrato/fisiologia
13.
Nat Commun ; 15(1): 5618, 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38965227

RESUMO

Naturally generated lipid nanoparticles termed extracellular vesicles (EVs) hold significant promise as engineerable therapeutic delivery vehicles. However, active loading of protein cargo into EVs in a manner that is useful for delivery remains a challenge. Here, we demonstrate that by rationally designing proteins to traffic to the plasma membrane and associate with lipid rafts, we can enhance loading of protein cargo into EVs for a set of structurally diverse transmembrane and peripheral membrane proteins. We then demonstrate the capacity of select lipid tags to mediate increased EV loading and functional delivery of an engineered transcription factor to modulate gene expression in target cells. We envision that this technology could be leveraged to develop new EV-based therapeutics that deliver a wide array of macromolecular cargo.


Assuntos
Vesículas Extracelulares , Nanopartículas , Vesículas Extracelulares/metabolismo , Humanos , Nanopartículas/química , Engenharia de Proteínas/métodos , Microdomínios da Membrana/metabolismo , Lipídeos/química , Membrana Celular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Animais , Sistemas de Liberação de Medicamentos , Transporte Proteico , Células HEK293 , Lipossomos
14.
Nat Commun ; 15(1): 3162, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38605024

RESUMO

The organization of membrane proteins between and within membrane-bound compartments is critical to cellular function. Yet we lack approaches to regulate this organization in a range of membrane-based materials, such as engineered cells, exosomes, and liposomes. Uncovering and leveraging biophysical drivers of membrane protein organization to design membrane systems could greatly enhance the functionality of these materials. Towards this goal, we use de novo protein design, molecular dynamic simulations, and cell-free systems to explore how membrane-protein hydrophobic mismatch could be used to tune protein cotranslational integration and organization in synthetic lipid membranes. We find that membranes must deform to accommodate membrane-protein hydrophobic mismatch, which reduces the expression and co-translational insertion of membrane proteins into synthetic membranes. We use this principle to sort proteins both between and within membranes, thereby achieving one-pot assembly of vesicles with distinct functions and controlled split-protein assembly, respectively. Our results shed light on protein organization in biological membranes and provide a framework to design self-organizing membrane-based materials with applications such as artificial cells, biosensors, and therapeutic nanoparticles.


Assuntos
Células Artificiais , Proteínas de Membrana , Membrana Celular/metabolismo , Membranas/metabolismo , Proteínas de Membrana/metabolismo , Lipossomos , Bicamadas Lipídicas/química
15.
Nat Commun ; 15(1): 5221, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38890329

RESUMO

Latent bioreactive unnatural amino acids (Uaas) have been widely used in the development of covalent drugs and identification of protein interactors, such as proteins, DNA, RNA and carbohydrates. However, it is challenging to perform high-throughput identification of Uaa cross-linking products due to the complexities of protein samples and the data analysis processes. Enrichable Uaas can effectively reduce the complexities of protein samples and simplify data analysis, but few cross-linked peptides were identified from mammalian cell samples with these Uaas. Here we develop an enrichable and multiple amino acids reactive Uaa, eFSY, and demonstrate that eFSY is MS cleavable when eFSY-Lys and eFSY-His are the cross-linking products. An identification software, AixUaa is developed to decipher eFSY mass cleavable data. We systematically identify direct interactomes of Thioredoxin 1 (Trx1) and Selenoprotein M (SELM) with eFSY and AixUaa.


Assuntos
Aminoácidos , Tiorredoxinas , Aminoácidos/metabolismo , Aminoácidos/química , Humanos , Tiorredoxinas/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/química , Reagentes de Ligações Cruzadas/química , Ligação Proteica , Peptídeos/metabolismo , Peptídeos/química , Selenoproteínas/metabolismo , Selenoproteínas/genética , Selenoproteínas/química , Software , Proteínas/metabolismo , Proteínas/química , Células HEK293
16.
Biochem J ; 441(2): 591-7, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21957909

RESUMO

The Prp19-associated complex [NTC (nineteen complex)] plays a crucial role in intron removal during premature mRNA splicing in eukaryotes. Only one component of the NTC, Cwc2, is capable of binding RNA. In the present study we report the 1.9 Å (1 Å=0.1 nm) X-ray structure of the Cwc2 core domain, which is both necessary and sufficient for RNA binding. The Cwc2 core domain contains two sub-domains, a CCCH-type ZnF (zinc finger) and a RRM (RNA recognition motif). Unexpectedly, the ZnF domain and the RRM form a single folding unit, glued together by extensive hydrophobic interactions and hydrogen bonds. Structure-guided mutational analysis revealed that the intervening loop [known as the RB loop (RNA-binding loop)] between ZnF and RRM plays an essential role in RNA binding. In addition, a number of highly conserved positively charged residues on the ß-strands of RRM make an important contribution to RNA binding. Intriguingly, these residues and a portion of the RB loop constitute an extended basic surface strip that encircles Cwc2 halfway. The present study serves as a framework for understanding the regulatory function of the NTC in RNA splicing.


Assuntos
Splicing de RNA , Proteínas de Ligação a RNA/química , Proteínas de Saccharomyces cerevisiae/química , Spliceossomos/química , Motivos de Aminoácidos , Cristalografia por Raios X , Modelos Moleculares , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Fatores de Processamento de RNA , Dedos de Zinco
17.
Curr Opin Struct Biol ; 74: 102381, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35537282

RESUMO

In recent decades, major progress has been made in the design of water-soluble proteins, yet the design of transmembrane proteins has lagged considerably. Despite their biological and pharmaceutical importance, only a limited number of transmembrane proteins have been successfully designed owing to the complexity of the membrane environment and difficulties in experimental characterization. Here, we introduce principles for transmembrane protein design in general and discuss design examples, including scaffold proteins and functional proteins. We also discuss how developments in design methods have advanced the field and what we may achieve with recent breakthroughs in structural biology.


Assuntos
Biologia Computacional , Proteínas de Membrana , Proteínas de Membrana/química
18.
Nat Commun ; 12(1): 4541, 2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34315898

RESUMO

Wntless (WLS), an evolutionarily conserved multi-pass transmembrane protein, is essential for secretion of Wnt proteins. Wnt-triggered signaling pathways control many crucial life events, whereas aberrant Wnt signaling is tightly associated with many human diseases including cancers. Here, we report the cryo-EM structure of human WLS in complex with Wnt3a, the most widely studied Wnt, at 2.2 Å resolution. The transmembrane domain of WLS bears a GPCR fold, with a conserved core cavity and a lateral opening. Wnt3a interacts with WLS at multiple interfaces, with the lipid moiety on Wnt3a traversing a hydrophobic tunnel of WLS transmembrane domain and inserting into membrane. A ß-hairpin of Wnt3a containing the conserved palmitoleoylation site interacts with WLS extensively, which is crucial for WLS-mediated Wnt secretion. The flexibility of the Wnt3a loop/hairpin regions involved in the multiple binding sites indicates induced fit might happen when Wnts are bound to different binding partners. Our findings provide important insights into the molecular mechanism of Wnt palmitoleoylation, secretion and signaling.


Assuntos
Microscopia Crioeletrônica , Receptores Acoplados a Proteínas G/ultraestrutura , Proteína Wnt3A/ultraestrutura , Receptores Frizzled/metabolismo , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Modelos Moleculares , Conformação Proteica , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Proteína Wnt3A/química , Proteína Wnt3A/metabolismo
19.
ACS Nano ; 15(3): 5671-5678, 2021 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-33586956

RESUMO

An atomically dispersed structure is attractive for electrochemically converting carbon dioxide (CO2) to fuels and feedstock due to its unique properties and activity. Most single-atom electrocatalysts are reported to reduce CO2 to carbon monoxide (CO). Herein, we develop atomically dispersed indium (In) on a nitrogen-doped carbon skeleton (In-N-C) as an efficient catalyst to produce formic acid/formate in aqueous media, reaching a turnover frequency as high as 26771 h-1 at -0.99 V relative to a reversible hydrogen electrode (RHE). Electrochemical measurements show that trace amounts of In loaded on the carbon matrix significantly improve the electrocatalytic behavior for the CO2 reduction reaction, outperforming conventional metallic In catalysts. Further experiments and density functional theory (DFT) calculations reveal that the formation of intermediate *OCHO on isolated In sites plays a pivotal role in the efficiency of the CO2-to-formate process, which has a lower energy barrier than that on metallic In.

20.
Nat Commun ; 12(1): 3384, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34099674

RESUMO

Despite recent success in computational design of structured cyclic peptides, de novo design of cyclic peptides that bind to any protein functional site remains difficult. To address this challenge, we develop a computational "anchor extension" methodology for targeting protein interfaces by extending a peptide chain around a non-canonical amino acid residue anchor. To test our approach using a well characterized model system, we design cyclic peptides that inhibit histone deacetylases 2 and 6 (HDAC2 and HDAC6) with enhanced potency compared to the original anchor (IC50 values of 9.1 and 4.4 nM for the best binders compared to 5.4 and 0.6 µM for the anchor, respectively). The HDAC6 inhibitor is among the most potent reported so far. These results highlight the potential for de novo design of high-affinity protein-peptide interfaces, as well as the challenges that remain.


Assuntos
Desenho de Fármacos , Inibidores de Histona Desacetilases/farmacologia , Peptídeos Cíclicos/farmacologia , Relação Estrutura-Atividade , Domínio Catalítico/efeitos dos fármacos , Cristalografia por Raios X , Ensaios Enzimáticos , Histona Desacetilase 2/antagonistas & inibidores , Histona Desacetilase 2/isolamento & purificação , Histona Desacetilase 2/metabolismo , Histona Desacetilase 2/ultraestrutura , Desacetilase 6 de Histona/antagonistas & inibidores , Desacetilase 6 de Histona/genética , Desacetilase 6 de Histona/isolamento & purificação , Desacetilase 6 de Histona/ultraestrutura , Inibidores de Histona Desacetilases/química , Concentração Inibidora 50 , Simulação de Acoplamento Molecular , Ressonância Magnética Nuclear Biomolecular , Biblioteca de Peptídeos , Peptídeos Cíclicos/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA