Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
J Biol Chem ; 297(6): 101419, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34801550

RESUMO

A profound understanding of the molecular interactions between receptors and ligands is important throughout diverse research, such as protein design, drug discovery, or neuroscience. What determines specificity and how do proteins discriminate against similar ligands? In this study, we analyzed factors that determine binding in two homologs belonging to the well-known superfamily of periplasmic binding proteins, PotF and PotD. Building on a previously designed construct, modes of polyamine binding were swapped. This change of specificity was approached by analyzing local differences in the binding pocket as well as overall conformational changes in the protein. Throughout the study, protein variants were generated and characterized structurally and thermodynamically, leading to a specificity swap and improvement in affinity. This dataset not only enriches our knowledge applicable to rational protein design but also our results can further lay groundwork for engineering of specific biosensors as well as help to explain the adaptability of pathogenic bacteria.


Assuntos
Escherichia coli K12/química , Proteínas de Escherichia coli/química , Proteínas Periplásmicas de Ligação/química , Receptores de Amina Biogênica/química , Espermidina/química , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Proteínas Periplásmicas de Ligação/genética , Proteínas Periplásmicas de Ligação/metabolismo , Ligação Proteica , Receptores de Amina Biogênica/genética , Receptores de Amina Biogênica/metabolismo , Espermidina/metabolismo
2.
mBio ; 12(5): e0213021, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34607455

RESUMO

Gram-negative bacteria have a multicomponent and constitutively active periplasmic chaperone system to ensure the quality control of their outer membrane proteins (OMPs). Recently, OMPs have been identified as a new class of vulnerable targets for antibiotic development, and therefore a comprehensive understanding of OMP quality control network components will be critical for discovering antimicrobials. Here, we demonstrate that the periplasmic chaperone Spy protects certain OMPs against protein-unfolding stress and can functionally compensate for other periplasmic chaperones, namely Skp and FkpA, in the Escherichia coli K-12 MG1655 strain. After extensive in vivo genetic experiments for functional characterization of Spy, we use nuclear magnetic resonance and circular dichroism spectroscopy to elucidate the mechanism by which Spy binds and folds two different OMPs. Along with holding OMP substrates in a dynamic conformational ensemble, Spy binding enables OmpX to form a partially folded ß-strand secondary structure. The bound OMP experiences temperature-dependent conformational exchange within the chaperone, pointing to a multitude of local dynamics. Our findings thus deepen the understanding of functional compensation among periplasmic chaperones during OMP biogenesis and will promote the development of innovative antimicrobials against pathogenic Gram-negative bacteria. IMPORTANCE Outer membrane proteins (OMPs) play critical roles in bacterial pathogenicity and provide a new niche for antibiotic development. A comprehensive understanding of the OMP quality control network will strongly impact antimicrobial discovery. Here, we systematically demonstrate that the periplasmic chaperone Spy has a role in maintaining the homeostasis of certain OMPs. Remarkably, Spy utilizes a unique chaperone mechanism to bind OmpX and allows it to form a partially folded ß-strand secondary structure in a dynamic exchange of conformations. This mechanism differs from that of other E. coli periplasmic chaperones such as Skp and SurA, both of which maintain OMPs in disordered conformations. Our study thus deepens the understanding of the complex OMP quality control system and highlights the differences in the mechanisms of ATP-independent chaperones.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/metabolismo , Membrana Celular/metabolismo , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Hidrolases/química , Hidrolases/metabolismo , Proteínas Periplásmicas/metabolismo , Trifosfato de Adenosina/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Membrana Celular/química , Membrana Celular/genética , Escherichia coli K12/química , Escherichia coli K12/genética , Proteínas de Escherichia coli/genética , Hidrolases/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Peptidilprolil Isomerase/genética , Peptidilprolil Isomerase/metabolismo , Proteínas Periplásmicas/genética , Ligação Proteica , Conformação Proteica em alfa-Hélice , Dobramento de Proteína
3.
J Biol Chem ; 297(4): 101168, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34487760

RESUMO

LysO, a prototypical member of the LysO family, mediates export of L-lysine (Lys) and resistance to the toxic Lys antimetabolite, L-thialysine (Thl) in Escherichia coli. Here, we have addressed unknown aspects of LysO function pertaining to its membrane topology and the mechanism by which it mediates Lys/Thl export. Using substituted cysteine (Cys) accessibility, here we delineated the membrane topology of LysO. Our studies support a model in which both the N- and C-termini of LysO are present at the periplasmic face of the membrane with a transmembrane (TM) domain comprising eight TM segments (TMSs) between them. In addition, a feature of intramembrane solvent exposure in LysO is inferred with the identification of membrane-located solvent-exposed Cys residues. Isosteric substitutions of a pair of conserved acidic residues, one E233, located in the solvent-exposed TMS7 and the other D261, in a solvent-exposed intramembrane segment located between TMS7 and TMS8, abolished LysO function in vivo. Thl, but not Lys, elicited proton release in inside-out membrane vesicles, a process requiring the presence of both E233 and D261. We postulate that Thl may be exported in antiport with H+ and that Lys may be a low-affinity export substrate. Our findings are compatible with a physiological scenario wherein in vivo LysO exports the naturally occurring antimetabolite Thl with higher affinity over the essential cellular metabolite Lys, thus affording protection from Thl toxicity and limiting wasteful export of Lys.


Assuntos
Sistemas de Transporte de Aminoácidos Básicos/química , Membrana Celular/química , Escherichia coli K12/química , Proteínas de Escherichia coli/química , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Transporte Biológico Ativo , Membrana Celular/genética , Membrana Celular/metabolismo , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Domínios Proteicos , Relação Estrutura-Atividade
4.
Rapid Commun Mass Spectrom ; 35(1): e8941, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-32885498

RESUMO

RATIONALE: Tracing isotopically labeled water into proteins allows for the detection of species-specific metabolic activity in complex communities. However, a stress response may alter the newly synthesized proteins. METHODS: We traced 18-oxygen from heavy water into proteins of Escherichia coli K12 grown from permissive to retardant temperatures. All samples were analyzed using UPLC/Orbitrap Q-Exactive-MS/MS operating in positive electrospray ionization mode. RESULTS: We found that warmer temperatures resulted in significantly (P-value < 0.05) higher incorporation of 18-oxygen as seen by both substrate utilization as relative isotope abundance (RIA) and growth as labeling ratio (LR). However, the absolute number of peptides with incorporation of 18-oxygen showed no significant correlation to temperature, potentially caused by the synthesis of different proteins at low temperatures, namely, proteins related to cold stress response. CONCLUSIONS: Our results unveil the species-specific cold stress response of E. coli K12 that could be misinterpreted as general growth; this is why the quantity as RIA and LR but also the quality as absolute number of peptides with incorporation (relative abundance, RA) and their function must be considered to fully understand the activity of microbial communities.


Assuntos
Resposta ao Choque Frio/fisiologia , Escherichia coli K12 , Proteínas de Escherichia coli , Marcação por Isótopo/métodos , Isótopos de Oxigênio , Cromatografia Líquida de Alta Pressão/métodos , Temperatura Baixa , Escherichia coli K12/química , Escherichia coli K12/metabolismo , Escherichia coli K12/fisiologia , Proteínas de Escherichia coli/análise , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Isótopos de Oxigênio/análise , Isótopos de Oxigênio/metabolismo , Espectrometria de Massas em Tandem/métodos
5.
Biochemistry ; 59(22): 2069-2077, 2020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32437133

RESUMO

YcjR from Escherichia coli K-12 MG1655 catalyzes the manganese-dependent reversible epimerization of 3-keto-α-d-gulosides to the corresponding 3-keto-α-d-glucosides as a part of a proposed catabolic pathway for the transformation of d-gulosides to d-glucosides. The three-dimensional structure of the manganese-bound enzyme was determined by X-ray crystallography. The divalent manganese ion is coordinated to the enzyme by ligation to Glu-146, Asp-179, His-205, and Glu-240. When either of the two active site glutamate residues is mutated to glutamine, the enzyme loses all catalytic activity for the epimerization of α-methyl-3-keto-d-glucoside at C4. However, the E240Q mutant can catalyze hydrogen-deuterium exchange of the proton at C4 of α-methyl-3-keto-d-glucoside in solvent D2O. The E146Q mutant does not catalyze this exchange reaction. These results indicate that YcjR catalyzes the isomerization of 3-keto-d-glucosides via proton abstraction at C4 by Glu-146 to form a cis-enediolate intermediate that is subsequently protonated on the opposite face by Glu-240 to generate the corresponding 3-keto-d-guloside. This conclusion is supported by docking of the cis-enediolate intermediate into the active site of YcjR based on the known binding orientation of d-fructose and d-psicose in the active site of d-psicose-3-epimerase.


Assuntos
Escherichia coli K12/química , Proteínas de Escherichia coli/metabolismo , Glucosídeos/metabolismo , Cristalografia por Raios X , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/química , Glucosídeos/química , Modelos Moleculares , Conformação Molecular , Estereoisomerismo
6.
Nat Struct Mol Biol ; 26(10): 910-918, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31570873

RESUMO

Protein cleavage inside the cell membrane triggers various pathophysiological signaling pathways, but the mechanism of catalysis is poorly understood. We solved ten structures of the Escherichia coli rhomboid protease in a bicelle membrane undergoing time-resolved steps that encompass the entire proteolytic reaction on a transmembrane substrate and an aldehyde inhibitor. Extensive gate opening accompanied substrate, but not inhibitor, binding, revealing that substrates and inhibitors take different paths to the active site. Catalysis unexpectedly commenced with, and was guided through subsequent catalytic steps by, motions of an extracellular loop, with local contributions from active site residues. We even captured the elusive tetrahedral intermediate that is uncleaved but covalently attached to the catalytic serine, about which the substrate was forced to bend dramatically. This unexpectedly stable intermediate indicates rhomboid catalysis uses an unprecedented reaction coordinate that may involve mechanically stressing the peptide bond, and could be selectively targeted by inhibitors.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Endopeptidases/metabolismo , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Membrana/metabolismo , Cristalografia por Raios X , Proteínas de Ligação a DNA/química , Endopeptidases/química , Escherichia coli K12/química , Proteínas de Escherichia coli/química , Proteínas de Membrana/química , Modelos Moleculares , Peptídeos/metabolismo , Conformação Proteica , Proteólise , Especificidade por Substrato
7.
Nat Struct Mol Biol ; 26(10): 919-929, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31570874

RESUMO

Cotranslational protein targeting is a conserved process for membrane protein biogenesis. In Escherichia coli, the essential ATPase SecA was found to cotranslationally target a subset of nascent membrane proteins to the SecYEG translocase at the plasma membrane. The molecular mechanism of this pathway remains unclear. Here we use biochemical and cryoelectron microscopy analyses to show that the amino-terminal amphipathic helix of SecA and the ribosomal protein uL23 form a composite binding site for the transmembrane domain (TMD) on the nascent protein. This binding mode further enables recognition of charged residues flanking the nascent TMD and thus explains the specificity of SecA recognition. Finally, we show that membrane-embedded SecYEG promotes handover of the translating ribosome from SecA to the translocase via a concerted mechanism. Our work provides a molecular description of the SecA-mediated cotranslational targeting pathway and demonstrates an unprecedented role of the ribosome in shielding nascent TMDs.


Assuntos
Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas Ribossômicas/metabolismo , Proteínas SecA/metabolismo , Sítios de Ligação , Microscopia Crioeletrônica , Escherichia coli K12/química , Proteínas de Escherichia coli/química , Modelos Moleculares , Biossíntese de Proteínas , Domínios Proteicos , Estrutura Secundária de Proteína , Proteínas Ribossômicas/química , Canais de Translocação SEC/química , Canais de Translocação SEC/metabolismo , Proteínas SecA/química
8.
Chem Commun (Camb) ; 55(68): 10158-10161, 2019 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-31389420

RESUMO

We developed a chemical method to covalently functionalize cellulose nanofibers and cellulose paper with mannoside ligands displaying a strong affinity for the FimH adhesin from pathogenic E. coli strains. Mannose-grafted cellulose proved efficient to selectively bind FimH lectin and discriminate pathogenic E. coli strains from non-pathogenic ones. These modified papers are valuable tools for diagnosing infections promoted by E. coli, such as cystitis or inflammatory bowel diseases, and the concept may be applicable to other life-threatening pathogens.


Assuntos
Celulose/química , Escherichia coli K12/isolamento & purificação , Mananas/química , Nanofibras/química , Adesinas de Escherichia coli/metabolismo , Aderência Bacteriana/efeitos dos fármacos , Técnicas de Tipagem Bacteriana/instrumentação , Técnicas de Tipagem Bacteriana/métodos , Linhagem Celular Tumoral , Celulose/metabolismo , Escherichia coli K12/química , Fezes/microbiologia , Proteínas de Fímbrias/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Mananas/metabolismo , Papel , Ligação Proteica
9.
J Phys Chem B ; 123(33): 7092-7102, 2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31340643

RESUMO

Lipoproteins are essential for bacterial survival. Bacterial lipoprotein biosynthesis is accomplished by sequential modification by three enzymes in the inner membrane, all of which are emerging antimicrobial targets. The X-ray crystal structure of prolipoprotein diacylglyceryl transferase (Lgt) and apolipoprotein N-acyl transferase (Lnt) has been reported. However, the mechanisms of the post-translational modification catalyzed by these enzymes have not been understood. Here, we studied the mechanism of the transacylation reaction catalyzed by Lgt, the first enzyme for lipoprotein modification using molecular docking, molecular dynamics, and quantum mechanics/molecular mechanics (QM/MM) calculations. Our results suggest that Arg143, Arg239, and Glu202 play a critical role in stabilizing the glycerol-1-phosphate head group and activating the glycerol C3-O ester bond of the phosphatidylglycerol (PG) substrate. With PG binding, the opening of the L6-7 loop mediated by the highly conserved Arg236 residue as a gatekeeper is observed, which facilitates the release of the modified lipoprotein product, as well as the entry of another PG substrate. Further QM/MM studies revealed that His103 acts as a catalytic base to abstract a proton from the cysteine residue of the preproliprotein, initiating the diacylglyceryl transfer from PG to preprolipoprotein. This is the first study on the mechanism of lipoprotein modification catalyzed by a post-translocational processing enzyme. The transacylation mechanism of Lgt would shed light on the development of novel antimicrobial therapies targeting the challenging enzymes involved in the post-translocational modification pathway of lipoproteins.


Assuntos
Escherichia coli K12/enzimologia , Fosfatidilgliceróis/metabolismo , Transferases/metabolismo , Acilação , Cristalografia por Raios X , Escherichia coli K12/química , Escherichia coli K12/metabolismo , Simulação de Acoplamento Molecular , Fosfatidilgliceróis/química , Conformação Proteica , Teoria Quântica , Especificidade por Substrato , Transferases/química
10.
Colloids Surf B Biointerfaces ; 182: 110361, 2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31351270

RESUMO

Curli are amyloid fibrils that polymerize extracellularly from curlin, a protein that is secreted by many enteric bacteria and is important for biofilm formation. Presented here is a systematic study of the effects of curli on bacteria-clay interactions. The aggregation trends of curli-producing and curli-deficient bacteria with clay minerals were followed using gradient-sedimentation experiments, Lumisizer measurements, bright-field and electron microscopy. The results revealed that curli-producing bacteria auto-aggregated into high-density flocs (1.23 g/cm3), ranging in size from 10 to 50 µm, that settle spontaneously. In contrast, curli-deficient bacteria remained relatively stable in solution as individual cells (1-2 µm, 1.18 g/cm3), even at high ionic strength (350 mM). The stability of clay suspensions mixed with curli-deficient bacteria depended on clay type and ionic strength, the general trends being consistent with the classic DLVO theory. However, suspensions of curli-producing bacteria mixed with clays were highly unstable regardless of clay type and solution chemistry, suggesting extensive interactions between the clays and the bacteria-curli aggregates. SEM measurements revealed interesting differences in morphologies of the aggregates; montmorillonite particles coated the bacterial auto-aggregates whereas the kaolinite platelets were embedded within the larger curli-bacteria aggregates. These new observations regarding the densities, aggregation trends, and morphologies of bacteria-curli and bacteria-curli-clay complexes make it clear that production of surface appendages, such as curli, need to be considered when addressing the fate, activity and transport of bacteria - particularly in aquatic environments.


Assuntos
Proteínas de Bactérias/química , Bentonita/química , Argila/química , Escherichia coli K12/química , Caulim/química , Amiloide/química , Aderência Bacteriana , Proteínas de Bactérias/biossíntese , Escherichia coli K12/metabolismo , Floculação , Cinética , Concentração Osmolar
11.
Langmuir ; 35(26): 8840-8849, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31177781

RESUMO

Tuning surface composition and stiffness is now an established strategy to improve the integration of medical implants. Recent evidence suggests that matrix stiffness affects bacterial adhesion, but contradictory findings have been reported in the literature. Distinguishing between the effects of bacterial adhesion and attachment strength on these surfaces may help interpret these findings. Here, we develop a precision microfluidic shear assay to quantify bacterial adhesion strength on stiffness-tunable and biomolecule-coated silicone materials. We demonstrate that bacteria are more strongly attached to soft silicones, compared to stiff silicones; as determined by retention against increasing shear flows. Interestingly, this effect is reduced when the surface is coated with matrix biomolecules. These results demonstrate that bacteria do sense and respond to stiffness of the surrounding environment and that precisely defined assays are needed to understand the interplay among surface mechanics, composition, and bacterial binding.


Assuntos
Materiais Revestidos Biocompatíveis/química , Escherichia coli K12/química , Técnicas Analíticas Microfluídicas , Silicones/química , Aderência Bacteriana , Materiais Revestidos Biocompatíveis/síntese química , Tamanho da Partícula , Resistência ao Cisalhamento , Propriedades de Superfície
12.
Biochemistry ; 58(26): 2875-2882, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31199144

RESUMO

In the absence of arabinose, the dimeric Escherichia coli regulatory protein of the l-arabinose operon, AraC, represses expression by looping the DNA between distant half-sites. Binding of arabinose to the dimerization domains forces AraC to preferentially bind two adjacent DNA half-sites, which stimulates RNA polymerase transcription of the araBAD catabolism genes. Prior genetic and biochemical studies hypothesized that arabinose allosterically induces a helix-coil transition of a linker between the dimerization and DNA binding domains that switches the AraC conformation to an inducing state [Brown, M. J., and Schleif, R. F. (2019) Biochemistry, preceding paper in this issue (DOI: 10.1021/acs.biochem.9b00234)]. To test this hypothesis, hydrogen-deuterium exchange mass spectrometry was utilized to identify structural regions involved in the conformational activation of AraC by arabinose. Comparison of the hydrogen-deuterium exchange kinetics of individual dimeric dimerization domains and the full-length dimeric AraC protein in the presence and absence of arabinose reveals a prominent arabinose-induced destabilization of the amide hydrogen-bonded structure of linker residues (I167 and N168). This destabilization is demonstrated to result from an increased probability to form a helix capping motif at the C-terminal end of the dimerizing α-helix of the dimerization domain that preceeds the interdomain linker. These conformational changes could allow for quaternary repositioning of the DNA binding domains required for induction of the araBAD promoter through rotation of peptide backbone dihedral angles of just a couple of residues. Subtle changes in exchange rates are also visible around the arabinose binding pocket and in the DNA binding domain.


Assuntos
Fator de Transcrição AraC/metabolismo , Arabinose/metabolismo , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/metabolismo , Fator de Transcrição AraC/química , Sítios de Ligação , DNA Bacteriano/metabolismo , Infecções por Escherichia coli/microbiologia , Escherichia coli K12/química , Proteínas de Escherichia coli/química , Humanos , Modelos Moleculares , Domínios Proteicos , Multimerização Proteica
13.
Anal Chem ; 91(14): 9221-9228, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31187982

RESUMO

Interactions of glycans with proteins, cells, and microorganisms play important roles in cell-cell adhesion and host-pathogen interaction. Glycan microarray technology, in which multiple glycan structures are immobilized on a single glass slide and interrogated with glycan-binding proteins (GBPs), has become an indispensable tool in the study of protein-glycan interactions. Despite its great success, the current format of the glycan microarray requires expensive, specialized instrumentation and labor-intensive assay and image processing procedures, which limit automation and possibilities for high-throughput analyses. Furthermore, the current microarray is not suitable for assaying interaction with intact cells due to their large size compared to the two-dimensional microarray surface. To address these limitations, we developed the next-generation glycan microarray (NGGM) based on artificial DNA coding of glycan structures. In this novel approach, a glycan library is presented as a mixture of glycans and glycoconjugates, each of which is coded with a unique oligonucleotide sequence (code). The glycan mixture is interrogated by GBPs followed by the separation of unbound coded glycans. The DNA sequences that identify individual bound glycans are quantitatively sequenced (decoded) by powerful next-generation sequencing (NGS) technology, and copied numbers of the DNA codes represent relative binding specificities of corresponding glycan structures to GBPs. We demonstrate that NGGM generates glycan-GBP binding data that are consistent with that generated in a slide-based glycan microarray. More importantly, the solution phase binding assay is directly applicable to identifying glycan binding to intact cells, which is often challenging using glass slide-based glycan microarrays.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , DNA/química , Glicoconjugados/metabolismo , Análise em Microsséries/métodos , Polissacarídeos/metabolismo , Acinetobacter baumannii/química , Animais , Química Click , Escherichia coli K12/química , Glicoconjugados/química , Sequenciamento de Nucleotídeos em Larga Escala , Polissacarídeos/química , Ligação Proteica , Staphylococcus aureus/química , Suínos
14.
Biochem Biophys Res Commun ; 514(1): 37-43, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31014676

RESUMO

HigA functions as the antitoxin in HigB-HigA toxin-antitoxin system. It neutralizes HigB-mediated toxicity by forming a stable toxin-antitoxin complex. Here the crystal structure of isolated HigA from Escherichia coli str. K-12 has been determined to 2.0 Šresolution. The structural differences between HigA and HigA in HigBA complex imply that HigA undergoes drastic conformational changes upon the binding of HigB. The conformational changes are achieved by rigid motions of N-terminal and C-terminal domains of HigA around its central linker domain, which is different from other known forms of regulation patterns in other organisms. As a transcriptional regulator, HigA bind to its operator DNA through the C-terminal HTH motif, in which key residues were identified in this study.


Assuntos
Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/metabolismo , Cristalografia por Raios X , Infecções por Escherichia coli/microbiologia , Escherichia coli K12/química , Proteínas de Escherichia coli/química , Humanos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Multimerização Proteica
15.
Biochem Biophys Res Commun ; 512(3): 448-452, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-30902387

RESUMO

Purified membrane proteins are most frequently studied solubilized in detergent, but the properties of detergent micelles are very different from those of lipid bilayers. Therefore, there is an increasing interest in studying membrane proteins under conditions that resemble the membrane protein native environment more closely. Although there are indications of differences between membrane proteins in detergent and in lipid bilayers, direct functional and structural comparisons are very hard to find. Nanodiscs have been established as a new platform that consists of two molecules of a membrane scaffold protein that surround a small lipid-bilayer patch. Here, we undertook the task of comparing the function and conformational states of the transport protein MsbA in detergent and nanodiscs using ATPase activity and luminescence resonance energy transfer (LRET) measurements to assess differences in activity and conformational states, respectively. MsbA is a prototypical member of the ATP binding cassette protein superfamily. MsbA activity was higher in nanodiscs vs detergent, which had clear structural correlates: an increase in the fraction of molecules displaying closed nucleotide-binding domain dimers in the apo state, and a decrease in the distance of the "dissociated" nucleotide-binding domains. Our LRET studies support the notion that the widely separated nucleotide binding domains observed in the MsbA x-ray structures in detergent do not correspond to physiological conformations. Although our studies focus on a particular ABC exporter, the possibility of similar environment effects on other membrane proteins should be carefully considered.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Proteínas de Bactérias/química , Detergentes/química , Escherichia coli K12/química , Bicamadas Lipídicas/química , Transportadores de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X , Escherichia coli K12/metabolismo , Modelos Moleculares , Nanoestruturas/química , Conformação Proteica , Multimerização Proteica
16.
J Mol Biol ; 431(10): 2006-2019, 2019 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-30914293

RESUMO

Type II single-span membrane proteins, such as CadC or RodZ, lacking a signal sequence and having a far-downstream hydrophobic segment, require the SecA secretion motor for insertion into the inner membrane of Escherichia coli. Using two chimeric single-span proteins containing a designed hydrophobic segment H, we have determined the requirements for SecA-mediated secretion, the molecular distinction between TM domains and signal peptides, and the propensity for hydrophobic H-segments to remain embedded within the bilayer after targeting. By means of engineered H-segments and a strategically placed SPase I cleavage site, we determined how targeting and stability of the chimeric proteins are affected by the length and hydrophobicity of the H-segment. Very hydrophobic segments (e.g., 16 Leu) are stably incorporated into the inner membrane, resulting in a C-terminal anchored membrane protein, while a 24L construct was not targeted to the membrane by SecA and remained in the cytoplasm. However, a construct carrying preMalE at the N-terminus led to SecA targeting to SecYEG via the native signal sequence and stable insertion of the downstream 24L H-segment. We show that the RseP intramembrane protease degrades weakly stable H-segments and is a useful tool for investigating the borderline between stable and unstable TM segments. Using RseP- cells, we find that moderately hydrophobic sequences (e.g., 5Leu + 11Ala) are targeted to SecYEG by SecA and inserted, but subsequently drop out of the membrane into the cytoplasm. Therefore, the free energy of transfer from translocon to bilayer is different from the transfer free energy from membrane to water.


Assuntos
Escherichia coli K12/química , Proteínas de Escherichia coli/química , Proteínas SecA/química , Sequência de Aminoácidos , Infecções por Escherichia coli/microbiologia , Humanos , Hidrogênio/química , Interações Hidrofóbicas e Hidrofílicas , Bicamadas Lipídicas/química , Domínios Proteicos
17.
Mol Immunol ; 108: 1-7, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30739075

RESUMO

Our previous data demonstrated that Friend leukemia virus integration 1 (Fli-1), an ETS transcription factor, governs pericyte loss and vascular dysfunction in cecal ligation and puncture-induced murine sepsis by regulating essential pyroptosis markers including caspase-1. However, whether Fli-1 regulates caspase-1 expression levels in vitro and how Fli-1 regulates caspase-1 remain unknown. Our present work further demonstrated that overexpressed Fli-1 significantly increased caspase-1 and IL-18 expression levels in cultured mouse lung pericytes. Bacterial outer membrane vesicles (OMVs) have been found to induce cell pyroptosis through transferring LPS intracellularly. Using OMVs to induce an in vitro model of pyroptosis, we observed that OMVs significantly increased protein levels of Fli-1 in mouse lung pericytes. Furthermore, knockdown of Fli-1 by siRNA blocked OMVs-induced caspase-1, caspase-11 and IL-18 expression levels. As caspase-1 was predicted as a potential target of Fli-1, we cloned murine caspase-1 promoter into a luciferase construct. Our data demonstrate for the first time that Fli-1 regulates caspase-1 expression by directly binding to its promoter regions measured by chromatin immunoprecipitation (ChIP) assay and luciferase reporter system. In summary, our findings demonstrated a novel role and mechanism of Fli-1 in regulating caspase-1 expression in lung pericytes.


Assuntos
Caspase 1/imunologia , Regulação Enzimológica da Expressão Gênica/imunologia , Proteína Proto-Oncogênica c-fli-1/imunologia , Animais , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/farmacologia , Caspase 1/genética , Escherichia coli K12/química , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Interleucina-18/genética , Interleucina-18/imunologia , Pulmão , Camundongos , Pericitos , Proteína Proto-Oncogênica c-fli-1/genética
18.
Biosens Bioelectron ; 130: 95-102, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30731351

RESUMO

The paper describes a new biological sensor which represents a resonator based on a segment of a rectangular waveguide of 8 GHz band with shear dimensions of 28.5 × 12.6 mm2. On one side, the resonator is bounded by a metallic short-circuited wall; on the other side, it is bounded by a lithium niobate plate with a porous polystyrene film. This film, applied by centrifugation and modified in high-frequency discharge plasma in argon, was used to immobilize cells of Escherichia coli K-12. This resonator was connected through a coaxial-waveguide adapter to the S parameter meter, by means of which the reflection coefficient S11 in the plane of the lithium niobate plate was measured. The addition of an aqueous solution of ampicillin at 4-50 µg/ml to immobilized cells led to a significant change in the reflection coefficient of S11 from - 10.15 dB to - 15.09 dB. At the same time, the resonance frequency changed insignificantly within the range 8.06-8.068 GHz. The optimal time for modifying the polystyrene film for obtaining the required porosity and the optimal time for the immobilization of the bacterial cells were determined. The immobilized cells retained their activity for 4 months at a temperature of 4 °C. The study showed the promise of such a biosensor to determine ß-lactam antibiotics in aqueous solutions by using ampicillin as an example. The limit of detection of the developed biosensor with respect to ampicillin was established (4 µg/ml).


Assuntos
Ampicilina/isolamento & purificação , Técnicas Biossensoriais , Escherichia coli K12/química , Ampicilina/química , Micro-Ondas , Nióbio/química , Óxidos/química , Temperatura , Água/química
19.
Org Biomol Chem ; 17(8): 2223-2231, 2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-30720838

RESUMO

N-Methylation of DNA/RNA bases can be regulatory or damaging and is linked to diseases including cancer and genetic disorders. Bacterial AlkB and human FTO are DNA/RNA demethylases belonging to the Fe(ii) and 2-oxoglutarate oxygenase superfamily. Modelling studies reveal conformational dynamics influence structure-function relationships of AlkB and FTO, e.g. why 1-methyladenine is a better substrate for AlkB than 6-methyladenine. Simulations show that the flexibility of the double stranded DNA substrate in AlkB influences correlated motions, including between the core jelly-roll fold and an active site loop involved in substrate binding. The FTO N- and C-terminal domains move in respect to one another in a manner likely important for substrate binding. Substitutions, including clinically observed ones, influencing catalysis contribute to the network of correlated motions in AlkB and FTO. Overall, the calculations highlight the importance of the overall protein environment and its flexibility to the geometry of the reactant complexes.


Assuntos
Enzimas AlkB/química , Dioxigenase FTO Dependente de alfa-Cetoglutarato/química , Escherichia coli K12/enzimologia , Proteínas de Escherichia coli/química , Adenina/análogos & derivados , Adenina/metabolismo , Enzimas AlkB/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Metilação de DNA , DNA de Cadeia Simples/metabolismo , Escherichia coli K12/química , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/metabolismo , Humanos , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Especificidade por Substrato
20.
Int J Mol Sci ; 20(3)2019 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-30764475

RESUMO

The outer membrane of Gram-negative bacteria contains ß-barrel proteins that form high-conducting ion channels providing a path for hydrophilic molecules, including antibiotics. Traditionally, these proteins have been considered to exist only in an open state so that regulation of outer membrane permeability was accomplished via protein expression. However, electrophysiological recordings show that ß-barrel channels respond to transmembrane voltages by characteristically switching from a high-conducting, open state, to a so-called 'closed' state, with reduced permeability and possibly exclusion of large metabolites. Here, we use the bacterial porin OmpF from E. coli as a model system to gain insight on the control of outer membrane permeability by bacterial porins through the modulation of their open state. Using planar bilayer electrophysiology, we perform an extensive study of the role of membrane lipids in the OmpF channel closure by voltage. We pay attention not only to the effects of charges in the hydrophilic lipid heads but also to the contribution of the hydrophobic tails in the lipid-protein interactions. Our results show that gating kinetics is governed by lipid characteristics so that each stage of a sequential closure is different from the previous one, probably because of intra- or intermonomeric rearrangements.


Assuntos
Escherichia coli K12/metabolismo , Bicamadas Lipídicas/metabolismo , Fosfolipídeos/metabolismo , Porinas/metabolismo , Acilação , Escherichia coli K12/química , Interações Hidrofóbicas e Hidrofílicas , Ativação do Canal Iônico , Bicamadas Lipídicas/química , Modelos Moleculares , Fosfolipídeos/química , Porinas/química , Conformação Proteica em Folha beta , Multimerização Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...