Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Genes Dev ; 33(5-6): 333-347, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30796017

RESUMEN

SUMOylation (small ubiquitin-like modifier) in the DNA double-strand break (DSB) response regulates recruitment, activity, and clearance of repair factors. However, our understanding of a role for deSUMOylation in this process is limited. Here we identify different mechanistic roles for deSUMOylation in homologous recombination (HR) and nonhomologous end joining (NHEJ) through the investigation of the deSUMOylase SENP2. We found that regulated deSUMOylation of MDC1 prevents excessive SUMOylation and its RNF4-VCP mediated clearance from DSBs, thereby promoting NHEJ. In contrast, we show that HR is differentially sensitive to SUMO availability and SENP2 activity is needed to provide SUMO. SENP2 is amplified as part of the chromosome 3q amplification in many cancers. Increased SENP2 expression prolongs MDC1 focus retention and increases NHEJ and radioresistance. Collectively, our data reveal that deSUMOylation differentially primes cells for responding to DSBs and demonstrates the ability of SENP2 to tune DSB repair responses.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Reparación del ADN por Unión de Extremidades/genética , Reparación del ADN/genética , Recombinación Homóloga/genética , Sumoilación/genética , Proteínas Adaptadoras Transductoras de Señales , Proteínas de Ciclo Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Cisteína Endopeptidasas/genética , Roturas del ADN de Doble Cadena , Células HEK293 , Células HeLa , Humanos , Rayos Infrarrojos , Proteínas Nucleares/metabolismo , Tolerancia a Radiación/genética , Transducción de Señal/genética , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteína que Contiene Valosina/metabolismo
2.
Nature ; 571(7766): 521-527, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31270457

RESUMEN

The integrity of genomes is constantly threatened by problems encountered by the replication fork. BRCA1, BRCA2 and a subset of Fanconi anaemia proteins protect stalled replication forks from degradation by nucleases, through pathways that involve RAD51. The contribution and regulation of BRCA1 in replication fork protection, and how this role relates to its role in homologous recombination, is unclear. Here we show that BRCA1 in complex with BARD1, and not the canonical BRCA1-PALB2 interaction, is required for fork protection. BRCA1-BARD1 is regulated by a conformational change mediated by the phosphorylation-directed prolyl isomerase PIN1. PIN1 activity enhances BRCA1-BARD1 interaction with RAD51, thereby increasing the presence of RAD51 at stalled replication structures. We identify genetic variants of BRCA1-BARD1 in patients with cancer that exhibit poor protection of nascent strands but retain homologous recombination proficiency, thus defining domains of BRCA1-BARD1 that are required for fork protection and associated with cancer development. Together, these findings reveal a BRCA1-mediated pathway that governs replication fork protection.


Asunto(s)
Proteína BRCA1/química , Proteína BRCA1/metabolismo , Replicación del ADN , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteína BRCA1/genética , Línea Celular Tumoral , Replicación del ADN/genética , Inestabilidad Genómica/genética , Humanos , Isomerismo , Mutación , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , Recombinasa Rad51/metabolismo
3.
J Biol Chem ; 295(21): 7516-7528, 2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32241912

RESUMEN

The ATPase SecA is an essential component of the bacterial Sec machinery, which transports proteins across the cytoplasmic membrane. Most SecA proteins contain a long C-terminal tail (CTT). In Escherichia coli, the CTT contains a structurally flexible linker domain and a small metal-binding domain (MBD). The MBD coordinates zinc via a conserved cysteine-containing motif and binds to SecB and ribosomes. In this study, we screened a high-density transposon library for mutants that affect the susceptibility of E. coli to sodium azide, which inhibits SecA-mediated translocation. Results from sequencing this library suggested that mutations removing the CTT make E. coli less susceptible to sodium azide at subinhibitory concentrations. Copurification experiments suggested that the MBD binds to iron and that azide disrupts iron binding. Azide also disrupted binding of SecA to membranes. Two other E. coli proteins that contain SecA-like MBDs, YecA and YchJ, also copurified with iron, and NMR spectroscopy experiments indicated that YecA binds iron via its MBD. Competition experiments and equilibrium binding measurements indicated that the SecA MBD binds preferentially to iron and that a conserved serine is required for this specificity. Finally, structural modeling suggested a plausible model for the octahedral coordination of iron. Taken together, our results suggest that SecA-like MBDs likely bind to iron in vivo.


Asunto(s)
Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/metabolismo , Hierro/metabolismo , Proteína SecA/metabolismo , Escherichia coli K12/genética , Proteínas de Escherichia coli/genética , Mutación , Unión Proteica , Dominios Proteicos , Proteína SecA/genética , Azida Sódica/farmacología
4.
J Bacteriol ; 199(18)2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28674068

RESUMEN

The EvgS/EvgA two-component system of Escherichia coli is activated in response to low pH and alkali metals and regulates many genes, including those for the glutamate-dependent acid resistance system and a number of efflux pumps. EvgS, the sensor kinase, is one of five unconventional histidine kinases (HKs) in E. coli and has a large periplasmic domain and a cytoplasmic PAS domain in addition to phospho-acceptor, HK and dimerization, internal receiver, and phosphotransfer domains. Mutations that constitutively activate the protein at pH 7 map to the PAS domain. Here, we built a homology model of the periplasmic region of EvgS, based on the structure of the equivalent region of the BvgS homologue, to guide mutagenesis of potential key residues in this region. We show that histidine 226 is required for induction and that it is structurally colocated with a proline residue (P522) at the top of the predicted transmembrane helix that is expected to play a key role in passing information to the cytoplasmic domains. We also show that the constitutive mutations in the PAS domain can be further activated by low external pH. Expression of the cytoplasmic part of the protein alone also gives constitutive activation, which is lost if the constitutive PAS mutations are present. These findings are consistent with a model in which EvgS senses both external and internal pH and is activated by a shift from a tight inactive to a weak active dimer, and we present an analysis of the purified cytoplasmic portion of EvgS that supports this.IMPORTANCE One of the ways bacteria sense their environment is through two-component systems, which have one membrane-bound protein to do the sensing and another inside the cell to turn genes on or off in response to what the membrane-bound protein has detected. The membrane-bound protein must thus be able to detect the stress and signal this detection event to the protein inside the cell. To understand this process, we studied a protein that helps E. coli to survive exposure to low pH, which it must do before taking up residence in the gastrointestinal tract. We describe a predicted structure for the main sensing part of the protein and identify some key residues within it that are involved in the sensing and signaling processes. We propose a mechanism for how the protein may become activated and present some evidence to support our proposal.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Histidina Quinasa/metabolismo , Proteínas Quinasas/metabolismo , Multimerización de Proteína , Análisis Mutacional de ADN , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Histidina Quinasa/química , Histidina Quinasa/genética , Concentración de Iones de Hidrógeno , Conformación Proteica , Proteínas Quinasas/química , Proteínas Quinasas/genética
5.
J Bacteriol ; 199(2)2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27795329

RESUMEN

SecA is an essential component of the Sec machinery in bacteria, which is responsible for transporting proteins across the cytoplasmic membrane. Recent work from our laboratory indicates that SecA binds to ribosomes. Here, we used two different approaches to demonstrate that SecA also interacts with nascent polypeptides in vivo and that these polypeptides are Sec substrates. First, we photo-cross-linked SecA to ribosomes in vivo and identified mRNAs that copurify with SecA. Microarray analysis of the copurifying mRNAs indicated a strong enrichment for proteins containing Sec-targeting sequences. Second, we used a 2-dimensional (2-D) gel approach to analyze radioactively labeled nascent polypeptides that copurify with SecA, including maltose binding protein, a well-characterized SecA substrate. The interaction of SecA with nascent chains was not strongly affected in cells lacking SecB or trigger factor, both of which also interact with nascent Sec substrates. Indeed, the ability of SecB to interact with nascent chains was disrupted in strains in which the interaction between SecA and the ribosome was defective. Analysis of the interaction of SecA with purified ribosomes containing arrested nascent chains in vitro indicates that SecA can begin to interact with a variety of nascent chains when they reach a length of ∼110 amino acids, which is considerably shorter than the length required for interaction with SecB. Our results suggest that SecA cotranslationally recognizes nascent Sec substrates and that this recognition could be required for the efficient delivery of these proteins to the membrane-embedded Sec machinery. IMPORTANCE: SecA is an ATPase that provides the energy for the translocation of proteins across the cytoplasmic membrane by the Sec machinery in bacteria. The translocation of most of these proteins is uncoupled from protein synthesis and is frequently described as "posttranslational." Here, we show that SecA interacts with nascent Sec substrates. This interaction is not dependent on SecB or trigger factor, which also interact with nascent Sec substrates. Moreover, the interaction of SecB with nascent polypeptides is dependent on the interaction of SecA with the ribosome, suggesting that interaction of the nascent chain with SecA precedes interaction with SecB. Our results suggest that SecA could recognize substrate proteins cotranslationally in order to efficiently target them for uncoupled protein translocation.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Bacterianas/metabolismo , Escherichia coli , Regulación Bacteriana de la Expresión Génica/fisiología , Canales de Translocación SEC/metabolismo , Adenosina Trifosfatasas/genética , Proteínas Bacterianas/genética , ADN Bacteriano/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Canales de Translocación SEC/genética , Proteína SecA
6.
Biochem Soc Trans ; 44(2): 619-23, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-27068979

RESUMEN

G-protein-coupled receptors (GPCRs) form the largest class of membrane proteins and are an important target for therapeutic drugs. These receptors are highly dynamic proteins sampling a range of conformational states in order to fulfil their complex signalling roles. In order to fully understand GPCR signalling mechanisms it is necessary to extract the receptor protein out of the plasma membrane. Historically this has universally required detergents which inadvertently strip away the annulus of lipid in close association with the receptor and disrupt lateral pressure exerted by the bilayer. Detergent-solubilized GPCRs are very unstable which presents a serious hurdle to characterization by biophysical methods. A range of strategies have been developed to ameliorate the detrimental effect of removing the receptor from the membrane including amphipols and reconstitution into nanodics stabilized by membrane scaffolding proteins (MSPs) but they all require exposure to detergent. Poly(styrene-co-maleic acid) (SMA) incorporates into membranes and spontaneously forms nanoscale poly(styrene-co-maleic acid) lipid particles (SMALPs), effectively acting like a 'molecular pastry cutter' to 'solubilize' GPCRs in the complete absence of detergent at any stage and with preservation of the native annular lipid throughout the process. GPCR-SMALPs have similar pharmacological properties to membrane-bound receptor, exhibit enhanced stability compared with detergent-solubilized receptors and being non-proteinaceous in nature, are fully compatible with downstream biophysical analysis of the encapsulated GPCR.


Asunto(s)
Lípidos/química , Maleatos/química , Poliestirenos/química , Receptores Acoplados a Proteínas G/química , Conformación Proteica
7.
Biochem J ; 461(2): 269-78, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24758594

RESUMEN

ABC (ATP-binding-cassette) transporters carry out many vital functions and are involved in numerous diseases, but study of the structure and function of these proteins is often hampered by their large size and membrane location. Membrane protein purification usually utilizes detergents to solubilize the protein from the membrane, effectively removing it from its native lipid environment. Subsequently, lipids have to be added back and detergent removed to reconstitute the protein into a lipid bilayer. In the present study, we present the application of a new methodology for the extraction and purification of ABC transporters without the use of detergent, instead, using a copolymer, SMA (polystyrene-co-maleic acid). SMA inserts into a bilayer and assembles into discrete particles, essentially solubilizing the membrane into small discs of bilayer encircled by a polymer, termed SMALPs (SMA lipid particles). We show that this polymer can extract several eukaryotic ABC transporters, P-glycoprotein (ABCB1), MRP1 (multidrug-resistance protein 1; ABCC1), MRP4 (ABCC4), ABCG2 and CFTR (cystic fibrosis transmembrane conductance regulator; ABCC7), from a range of different expression systems. The SMALP-encapsulated ABC transporters can be purified by affinity chromatography, and are able to bind ligands comparably with those in native membranes or detergent micelles. A greater degree of purity and enhanced stability is seen compared with detergent solubilization. The present study demonstrates that eukaryotic ABC transporters can be extracted and purified without ever being removed from their lipid bilayer environment, opening up a wide range of possibilities for the future study of their structure and function.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/aislamiento & purificación , Transportadoras de Casetes de Unión a ATP/aislamiento & purificación , Regulador de Conductancia de Transmembrana de Fibrosis Quística/aislamiento & purificación , Maleatos/química , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/aislamiento & purificación , Proteínas de Neoplasias/aislamiento & purificación , Poliestirenos/química , Proteínas Recombinantes/aislamiento & purificación , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/química , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/química , Animales , Clonación Molecular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/química , Humanos , Cinética , Ligandos , Ratones , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/química , Proteínas de Neoplasias/química , Unión Proteica , Conformación Proteica , Estabilidad Proteica , Proteínas Recombinantes/química
8.
Microb Cell Fact ; 13(1): 127, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25186468

RESUMEN

BACKGROUND: Pichia pastoris is a widely-used host for recombinant protein production; expression is typically driven by methanol-inducible alcohol oxidase (AOX) promoters. Recently this system has become an important source of recombinant G protein-coupled receptors (GPCRs) for structural biology and drug discovery. The influence of diverse culture parameters (such as pH, dissolved oxygen concentration, medium composition, antifoam concentration and culture temperature) on productivity has been investigated for a wide range of recombinant proteins in P. pastoris. In contrast, the impact of the pre-induction phases on yield has not been as closely studied. In this study, we examined the pre-induction phases of P. pastoris bioreactor cultivations producing three different recombinant proteins: the GPCR, human A(2a) adenosine receptor (hA(2a)R), green fluorescent protein (GFP) and human calcitonin gene-related peptide receptor component protein (as a GFP fusion protein; hCGRP-RCP-GFP). RESULTS: Functional hA(2a)R was detected in the pre-induction phases of a 1 L bioreactor cultivation of glycerol-grown P. pastoris. In a separate experiment, a glycerol-grown P. pastoris strain secreted soluble GFP prior to methanol addition. When glucose, which has been shown to repress AOX expression, was the pre-induction carbon source, hA(2a)R and GFP were still produced in the pre-induction phases. Both hA(2a)R and GFP were also produced in methanol-free cultivations; functional protein yields were maintained or increased after depletion of the carbon source. Analysis of the pre-induction phases of 10 L pilot scale cultivations also demonstrated that pre-induction yields were at least maintained after methanol induction, even in the presence of cytotoxic concentrations of methanol. Additional bioreactor data for hCGRP-RCP-GFP and shake-flask data for GFP, horseradish peroxidase (HRP), the human tetraspanins hCD81 and CD82, and the tight-junction protein human claudin-1, demonstrated that bioreactor but not shake-flask cultivations exhibit recombinant protein production in the pre-induction phases of P. pastoris cultures. CONCLUSIONS: The production of recombinant hA(2a)R, GFP and hCGRP-RCP-GFP can be detected in bioreactor cultivations prior to methanol induction, while this is not the case for shake-flask cultivations of GFP, HRP, hCD81, hCD82 and human claudin-1. This confirms earlier suggestions of leaky expression from AOX promoters, which we report here for both glycerol- and glucose-grown cells in bioreactor cultivations. These findings suggest that the productivity of AOX-dependent bioprocesses is not solely dependent on induction by methanol. We conclude that in order to maximize total yields, pre-induction phase cultivation conditions should be optimized, and that increased specific productivity may result in decreased biomass yields.


Asunto(s)
Reactores Biológicos/microbiología , Expresión Génica , Microbiología Industrial/métodos , Pichia/genética , Receptores de Péptido Relacionado con el Gen de Calcitonina/metabolismo , Receptores Purinérgicos P1/metabolismo , Biomasa , Glucosa/metabolismo , Glicerol/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Microbiología Industrial/instrumentación , Pichia/crecimiento & desarrollo , Pichia/metabolismo , Receptores de Péptido Relacionado con el Gen de Calcitonina/genética , Receptores Purinérgicos P1/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
9.
Nanotechnology ; 25(28): 285101, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24972373

RESUMEN

Surfactant-mediated removal of proteins from biomembranes invariably results in partial or complete loss of function and disassembly of multi-protein complexes. We determined the capacity of styrene-co-maleic acid (SMA) co-polymer to remove components of the cell division machinery from the membrane of drug-resistant staphylococcal cells. SMA-lipid nanoparticles solubilized FtsZ-PBP2-PBP2a complexes from intact cells, demonstrating the close physical proximity of these proteins within the lipid bilayer. Exposure of bacteria to (-)-epicatechin gallate, a polyphenolic agent that abolishes ß-lactam resistance in staphylococci, disrupted the association between PBP2 and PBP2a. Thus, SMA purification provides a means to remove native integral membrane protein assemblages with minimal physical disruption and shows promise as a tool for the interrogation of molecular aspects of bacterial membrane protein structure and function.


Asunto(s)
Proteínas Bacterianas/química , Proteínas de la Membrana/química , Proteínas de Unión a las Penicilinas/química , Péptido Sintasas/química , Staphylococcus aureus/química , Tensoactivos/química , Catequina/análogos & derivados , Catequina/química , División Celular/fisiología , Membrana Dobles de Lípidos/química , Maleatos/química , Poliestirenos/química
10.
Biochem Soc Trans ; 39(2): 537-40, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21428935

RESUMEN

Tetraspanins are thought to exert their biological function(s) by co-ordinating the lateral movement and trafficking of associated molecules into tetraspanin-enriched microdomains. A second four-TM (transmembrane) domain protein family, the Claudin superfamily, is the major structural component of cellular TJs (tight junctions). Although the Claudin family displays low sequence homology and appears to be evolutionarily distinct from the tetraspanins, CD81 and Claudin-1 are critical molecules defining HCV (hepatitis C virus) entry; we recently demonstrated that CD81-Claudin-1 complexes have an essential role in this process. To understand the molecular basis of CD81-Claudin-1 complex formation, we produced and purified milligram quantities of full-length CD81 and Claudin-1, alone and in complex, in both detergent and lipid contexts. Structural characterization of these purified proteins will allow us to define the mechanism(s) underlying virus-cell interactions and aid the design of therapeutic agents targeting early steps in the viral life cycle.


Asunto(s)
Antígenos CD/metabolismo , Hepacivirus/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Virales/química , Receptores Virales/metabolismo , Animales , Antígenos CD/química , Claudina-1 , Hepacivirus/fisiología , Humanos , Proteínas de la Membrana/química , Modelos Biológicos , Conformación Molecular , Complejos Multiproteicos/análisis , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Receptores Virales/análisis , Tetraspanina 28 , Internalización del Virus
11.
Biochem Soc Trans ; 39(3): 813-8, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21599653

RESUMEN

In order to study the structure and function of a protein, it is generally required that the protein in question is purified away from all others. For soluble proteins, this process is greatly aided by the lack of any restriction on the free and independent diffusion of individual protein particles in three dimensions. This is not the case for membrane proteins, as the membrane itself forms a continuum that joins the proteins within the membrane with one another. It is therefore essential that the membrane is disrupted in order to allow separation and hence purification of membrane proteins. In the present review, we examine recent advances in the methods employed to separate membrane proteins before purification. These approaches move away from solubilization methods based on the use of small surfactants, which have been shown to suffer from significant practical problems. Instead, the present review focuses on methods that stem from the field of nanotechnology and use a range of reagents that fragment the membrane into nanometre-scale particles containing the protein complete with the local membrane environment. In particular, we examine a method employing the amphipathic polymer poly(styrene-co-maleic acid), which is able to reversibly encapsulate the membrane protein in a 10 nm disc-like structure ideally suited to purification and further biochemical study.


Asunto(s)
Membrana Celular/química , Proteínas de la Membrana/aislamiento & purificación , Tensoactivos/química , Humanos , Maleatos/química , Lípidos de la Membrana/química , Proteínas de la Membrana/química , Modelos Moleculares , Poliestirenos/química , Conformación Proteica , Solubilidad
12.
Biochem Soc Trans ; 39(3): 719-23, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21599640

RESUMEN

Membrane proteins are drug targets for a wide range of diseases. Having access to appropriate samples for further research underpins the pharmaceutical industry's strategy for developing new drugs. This is typically achieved by synthesizing a protein of interest in host cells that can be cultured on a large scale, allowing the isolation of the pure protein in quantities much higher than those found in the protein's native source. Yeast is a popular host as it is a eukaryote with similar synthetic machinery to that of the native human source cells of many proteins of interest, while also being quick, easy and cheap to grow and process. Even in these cells, the production of human membrane proteins can be plagued by low functional yields; we wish to understand why. We have identified molecular mechanisms and culture parameters underpinning high yields and have consolidated our findings to engineer improved yeast host strains. By relieving the bottlenecks to recombinant membrane protein production in yeast, we aim to contribute to the drug discovery pipeline, while providing insight into translational processes.


Asunto(s)
Proteínas de la Membrana/metabolismo , Proteínas Recombinantes/metabolismo , Levaduras/metabolismo , Bioingeniería , Humanos , Proteínas de la Membrana/genética , Proteínas Recombinantes/genética , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Levaduras/citología , Levaduras/genética
13.
Biochim Biophys Acta Biomembr ; 1862(6): 183235, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32126232

RESUMEN

The adenosine 2A receptor (A2AR), a G-protein-coupled receptor (GPCR), was solubilised and purified encapsulated in styrene maleic acid lipid particles (SMALPs). The purified A2AR-SMALP was associated with phospholipids characteristic of the plasma membrane of Pichia pastoris, the host used for its expression, confirming that the A2AR-SMALP encapsulated native lipids. The fluorescence spectrum of the A2AR-SMALP showed a characteristic broad emission peak at 330 nm, produced by endogenous Trp residues. The inverse agonist ZM241385 caused 30% increase in fluorescence emission, unusually accompanied by a red-shift in the emission wavelength. The emission spectrum also showed sub-peaks at 321 nm, 335 nm and 350 nm, indicating that individual Trp inhabited different environments following ZM241385 addition. There was no effect of the agonist NECA on the A2AR-SMALP fluorescence spectrum. Substitution of two Trp residues by Tyr suggested that ZM241385 affected the environment and mobility of Trp2466.48 in TM6 and Trp2687.33 at the extracellular face of TM7, causing transition to a more hydrophobic environment. The fluorescent moiety IAEDANS was site-specifically introduced at the intracellular end of TM6 (residue 2316.33) to report on the dynamic cytoplasmic face of the A2AR. The inverse agonist ZM241385 caused a concentration-dependent increase in fluorescence emission as the IAEDANS moved to a more hydrophobic environment, consistent with closing the G-protein binding crevice. NECA generated only 30% of the effect of ZM241385. This study provides insight into the SMALP environment; encapsulation supported constitutive activity of the A2AR and ZM241385-induced conformational transitions but the agonist NECA generated only small effects.


Asunto(s)
Lípidos/química , Receptor de Adenosina A2A/química , Estireno/química , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Maleatos/química , Pichia/química , Conformación Proteica , Espectrometría de Fluorescencia/métodos , Triazinas/farmacología , Triazoles/farmacología , Triptófano/química
14.
Elife ; 82019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31246174

RESUMEN

In bacteria, the translocation of proteins across the cytoplasmic membrane by the Sec machinery requires the ATPase SecA. SecA binds ribosomes and recognises nascent substrate proteins, but the molecular mechanism of nascent substrate recognition is unknown. We investigated the role of the C-terminal tail (CTT) of SecA in nascent polypeptide recognition. The CTT consists of a flexible linker (FLD) and a small metal-binding domain (MBD). Phylogenetic analysis and ribosome binding experiments indicated that the MBD interacts with 70S ribosomes. Disruption of the MBD only or the entire CTT had opposing effects on ribosome binding, substrate-protein binding, ATPase activity and in vivo function, suggesting that the CTT influences the conformation of SecA. Site-specific crosslinking indicated that F399 in SecA contacts ribosomal protein uL29, and binding to nascent chains disrupts this interaction. Structural studies provided insight into the CTT-mediated conformational changes in SecA. Our results suggest a mechanism for nascent substrate protein recognition.


Asunto(s)
Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Traslocación Bacteriana , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteína SecA/química , Proteína SecA/metabolismo , Secuencia de Aminoácidos , Biocatálisis , Reactivos de Enlaces Cruzados/química , Evolución Molecular , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Péptidos/metabolismo , Filogenia , Unión Proteica , Dominios Proteicos , Pliegue de Proteína , Ribosomas/metabolismo , Especificidad por Sustrato
15.
Sci Rep ; 9(1): 18712, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31822696

RESUMEN

The E. coli membrane protein ZipA, binds to the tubulin homologue FtsZ, in the early stage of cell division. We isolated ZipA in a Styrene Maleic Acid lipid particle (SMALP) preserving its position and integrity with native E. coli membrane lipids. Direct binding of ZipA to FtsZ is demonstrated, including FtsZ fibre bundles decorated with ZipA. Using Cryo-Electron Microscopy, small-angle X-ray and neutron scattering, we determine the encapsulated-ZipA structure in isolation, and in complex with FtsZ to a resolution of 1.6 nm. Three regions can be identified from the structure which correspond to, SMALP encapsulated membrane and ZipA transmembrane helix, a separate short compact tether, and ZipA globular head which binds FtsZ. The complex extends 12 nm from the membrane in a compact structure, supported by mesoscale modelling techniques, measuring the movement and stiffness of the regions within ZipA provides molecular scale analysis and visualisation of the early divisome.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , División Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas Bacterianas/fisiología , Proteínas Portadoras/fisiología , Proteínas Portadoras/ultraestructura , Proteínas de Ciclo Celular/fisiología , Proteínas de Ciclo Celular/ultraestructura , Microscopía por Crioelectrón/métodos , Proteínas del Citoesqueleto/fisiología , Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiología , Proteínas de Escherichia coli/ultraestructura , Proteínas de la Membrana/metabolismo , Unión Proteica
16.
Nat Microbiol ; 4(10): 1692-1705, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31235958

RESUMEN

The Mla pathway is believed to be involved in maintaining the asymmetrical Gram-negative outer membrane via retrograde phospholipid transport. The pathway is composed of three components: the outer membrane MlaA-OmpC/F complex, a soluble periplasmic protein, MlaC, and the inner membrane ATPase, MlaFEDB complex. Here, we solve the crystal structure of MlaC in its phospholipid-free closed apo conformation, revealing a pivoting ß-sheet mechanism that functions to open and close the phospholipid-binding pocket. Using the apo form of MlaC, we provide evidence that the inner-membrane MlaFEDB machinery exports phospholipids to MlaC in the periplasm. Furthermore, we confirm that the phospholipid export process occurs through the MlaD component of the MlaFEDB complex and that this process is independent of ATP. Our data provide evidence of an apparatus for lipid export away from the inner membrane and suggest that the Mla pathway may have a role in anterograde phospholipid transport.


Asunto(s)
Proteínas Bacterianas/metabolismo , Membrana Celular/metabolismo , Proteínas de la Membrana/metabolismo , Fosfolípidos/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Transporte Biológico , Cristalografía por Rayos X , Bacterias Gramnegativas/metabolismo , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Modelos Biológicos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Periplasma/metabolismo , Unión Proteica , Conformación Proteica en Lámina beta
17.
Wellcome Open Res ; 2: 82, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29090272

RESUMEN

Background: Tetraspanins are small transmembrane proteins, found in all higher eukaryotes, that compartmentalize cellular membranes through interactions with partner proteins. CD81 is a prototypical tetraspanin and contributes to numerous physiological and pathological processes, including acting as a critical entry receptor for hepatitis C virus (HCV). Antibody engagement of tetraspanins can induce a variety of effects, including actin cytoskeletal rearrangements, activation of MAPK-ERK signaling and cell migration. However, the epitope specificity of most anti-tetraspanin antibodies is not known, limiting mechanistic interpretation of these studies. Methods: We generated a panel of monoclonal antibodies (mAbs) specific for CD81 second extracellular domain (EC2) and performed detailed epitope mapping with a panel of CD81 mutants. All mAbs were screened for their ability to inhibit HCV infection and E2-CD81 association. Nanoscale distribution of cell surface CD81 was investigated by scanning electron microscopy. Results: The antibodies were classified in two epitope groups targeting opposing sides of EC2. We observed a wide range of anti-HCV potencies that were independent of their epitope grouping, but associated with their relative affinity for cell-surface expressed CD81. Scanning electron microscopy identified at least two populations of CD81; monodisperse and higher-order assemblies, consistent with tetraspanin-enriched microdomains. Conclusions: These novel antibodies provide well-characterised tools to investigate CD81 function, including HCV entry, and have the potential to provide insights into tetraspanin biology in general.

18.
Sci Rep ; 7(1): 8608, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28819315

RESUMEN

Bacterial proteins with MCE domains were first described as being important for Mammalian Cell Entry. More recent evidence suggests they are components of lipid ABC transporters. In Escherichia coli, the single-domain protein MlaD is known to be part of an inner membrane transporter that is important for maintenance of outer membrane lipid asymmetry. Here we describe two multi MCE domain-containing proteins in Escherichia coli, PqiB and YebT, the latter of which is an orthologue of MAM-7 that was previously reported to be an outer membrane protein. We show that all three MCE domain-containing proteins localise to the inner membrane. Bioinformatic analyses revealed that MCE domains are widely distributed across bacterial phyla but multi MCE domain-containing proteins evolved in Proteobacteria from single-domain proteins. Mutants defective in mlaD, pqiAB and yebST were shown to have distinct but partially overlapping phenotypes, but the primary functions of PqiB and YebT differ from MlaD. Complementing our previous findings that all three proteins bind phospholipids, results presented here indicate that multi-domain proteins evolved in Proteobacteria for specific functions in maintaining cell envelope homeostasis.


Asunto(s)
Membrana Celular/metabolismo , Proteínas de Escherichia coli/metabolismo , Homeostasis , Lípidos de la Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Sitios de Unión/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de la Membrana/genética , Mutación , Operón , Fosfolípidos/metabolismo , Unión Proteica , Mapas de Interacción de Proteínas
19.
Nat Protoc ; 11(7): 1149-62, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27254461

RESUMEN

Despite the great importance of membrane proteins, structural and functional studies of these proteins present major challenges. A significant hurdle is the extraction of the functional protein from its natural lipid membrane. Traditionally achieved with detergents, purification procedures can be costly and time consuming. A critical flaw with detergent approaches is the removal of the protein from the native lipid environment required to maintain functionally stable protein. This protocol describes the preparation of styrene maleic acid (SMA) co-polymer to extract membrane proteins from prokaryotic and eukaryotic expression systems. Successful isolation of membrane proteins into SMA lipid particles (SMALPs) allows the proteins to remain with native lipid, surrounded by SMA. We detail procedures for obtaining 25 g of SMA (4 d); explain the preparation of protein-containing SMALPs using membranes isolated from Escherichia coli (2 d) and control protein-free SMALPS using E. coli polar lipid extract (1-2 h); investigate SMALP protein purity by SDS-PAGE analysis and estimate protein concentration (4 h); and detail biophysical methods such as circular dichroism (CD) spectroscopy and sedimentation velocity analytical ultracentrifugation (svAUC) to undertake initial structural studies to characterize SMALPs (∼2 d). Together, these methods provide a practical tool kit for those wanting to use SMALPs to study membrane proteins.


Asunto(s)
Proteínas de Escherichia coli/aislamiento & purificación , Escherichia coli/química , Maleatos/química , Lípidos de la Membrana/aislamiento & purificación , Proteínas de la Membrana/aislamiento & purificación , Poliestirenos/química , Electroforesis en Gel de Poliacrilamida , Modelos Moleculares , Solubilidad
20.
Biosci Rep ; 35(2)2015 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-25720391

RESUMEN

G-protein coupled receptors (GPCRs) constitute the largest class of membrane proteins and are a major drug target. A serious obstacle to studying GPCR structure/function characteristics is the requirement to extract the receptors from their native environment in the plasma membrane, coupled with the inherent instability of GPCRs in the detergents required for their solubilization. In the present study, we report the first solubilization and purification of a functional GPCR [human adenosine A2A receptor (A2AR)], in the total absence of detergent at any stage, by exploiting spontaneous encapsulation by styrene maleic acid (SMA) co-polymer direct from the membrane into a nanoscale SMA lipid particle (SMALP). Furthermore, the A2AR-SMALP, generated from yeast (Pichia pastoris) or mammalian cells, exhibited increased thermostability (~5°C) compared with detergent [DDM (n-dodecyl-ß-D-maltopyranoside)]-solubilized A2AR controls. The A2AR-SMALP was also stable when stored for prolonged periods at 4°C and was resistant to multiple freeze-thaw cycles, in marked contrast with the detergent-solubilized receptor. These properties establish the potential for using GPCR-SMALP in receptor-based drug discovery assays. Moreover, in contrast with nanodiscs stabilized by scaffold proteins, the non-proteinaceous nature of the SMA polymer allowed unobscured biophysical characterization of the embedded receptor. Consequently, CD spectroscopy was used to relate changes in secondary structure to loss of ligand binding ([(3)H]ZM241385) capability. SMALP-solubilization of GPCRs, retaining the annular lipid environment, will enable a wide range of therapeutic targets to be prepared in native-like state to aid drug discovery and understanding of GPCR molecular mechanisms.


Asunto(s)
Receptor de Adenosina A2A/química , Receptor de Adenosina A2A/aislamiento & purificación , Línea Celular , Células HEK293 , Humanos , Maltosa/análogos & derivados , Maltosa/química , Pichia/química , Pichia/genética , Pichia/metabolismo , Estabilidad Proteica , Receptor de Adenosina A2A/biosíntesis , Receptor de Adenosina A2A/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Solubilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA