Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Microbiol ; 111(1): 269-286, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30353924

RESUMO

Agrobacterium tumefaciens transfers oncogenic T-DNA via the type IV secretion system (T4SS) into plants causing tumor formation. The acvB gene encodes a virulence factor of unknown function required for plant transformation. Here we specify AcvB as a periplasmic lysyl-phosphatidylglycerol (L-PG) hydrolase, which modulates L-PG homeostasis. Through functional characterization of recombinant AcvB variants, we showed that the C-terminal domain of AcvB (residues 232-456) is sufficient for full enzymatic activity and defined key residues for catalysis. Absence of the hydrolase resulted in ~10-fold increase in L-PG in Agrobacterium membranes and abolished T-DNA transfer and tumor formation. Overproduction of the L-PG synthase gene (lpiA) in wild-type A. tumefaciens resulted in a similar increase in the L-PG content (~7-fold) and a virulence defect even in the presence of intact AcvB. These results suggest that elevated L-PG amounts (either by overproduction of the synthase or absence of the hydrolase) are responsible for the virulence phenotype. Gradually increasing the L-PG content by complementation with different acvB variants revealed that cellular L-PG levels above 3% of total phospholipids interfere with T-DNA transfer. Cumulatively, this study identified AcvB as a novel virulence factor required for membrane lipid homeostasis and T-DNA transfer.


Assuntos
Agrobacterium tumefaciens/patogenicidade , Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Homeostase , Lisina/metabolismo , Fosfatidilgliceróis/metabolismo , Fatores de Virulência/metabolismo , Agrobacterium tumefaciens/crescimento & desenvolvimento , Proteínas de Bactérias/genética , Domínio Catalítico , Análise Mutacional de DNA , DNA Bacteriano/metabolismo , Proteínas de Ligação a DNA/genética , Deleção de Genes , Teste de Complementação Genética , Proteínas Periplásmicas/genética , Proteínas Periplásmicas/metabolismo , Doenças das Plantas/microbiologia , Solanum tuberosum/microbiologia , Transformação Genética , Virulência , Fatores de Virulência/genética
2.
Biochem J ; 475(11): 1885-1907, 2018 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-29717024

RESUMO

A quantitative Pseudomonas aeruginosa proteomics approach revealed increased abundance of the so-far uncharacterized protein PA3911 in anaerobic biofilms grown under conditions of the cystic fibrosis lung. Physiological relevance of ORF PA3911 was demonstrated, inter alia, using phenotype microarray experiments. The mutant strain showed increased susceptibility in the presence of antimicrobials (minocycline, nafcillin, oxacillin, chloramphenicol and thiamphenicol), enhanced twitching motility and significantly impaired biofilm formation. PA3911 is a soluble, cytoplasmic protein in P. aeruginosa In protein-lipid overlay experiments, purified PA3911 bound specifically to phosphatidic acid (PA), the central hub of phospholipid metabolism. Structure-guided site-directed mutagenesis was used to explore the proposed ligand-binding cavity of PA3911. Protein variants of Leu56, Leu58, Val69 and Leu114 were shown to impair PA interaction. A comparative shotgun lipidomics approach demonstrated a multifaceted response of P. aeruginosa to anaerobic conditions at the lipid head group and fatty acid level. Lipid homeostasis in the PA3911 mutant strain was imbalanced with respect to lysophosphatidylcholine, phosphatidylcholine and diacylglycerol under anaerobic and/or aerobic conditions. The impact of the newly identified PA-binding protein on lipid homeostasis and the related macroscopic phenotypes of P. aeruginosa are discussed.


Assuntos
Proteínas de Bactérias/metabolismo , Biofilmes , Ácidos Fosfatídicos/metabolismo , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/fisiologia , Adaptação Biológica , Anaerobiose , Proteínas de Bactérias/genética , Homeostase , Humanos , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/genética
3.
Biochem J ; 474(1): 163-178, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27803248

RESUMO

The molybdenum cofactor (Moco) is a redox active prosthetic group, essentially required for numerous enzyme-catalyzed two electron transfer reactions. Moco is synthesized by an evolutionarily old and highly conserved multistep pathway. In the last step of Moco biosynthesis, the molybdenum center is inserted into the final Moco precursor adenylated molybdopterin (MPT-AMP). This unique and yet poorly characterized maturation reaction finally yields physiologically active Moco. In the model plant Arabidopsis, the two domain enzyme, Cnx1, is required for Moco formation. Recently, a genetic screen identified novel Arabidopsis cnx1 mutant plant lines each harboring a single amino acid exchange in the N-terminal Cnx1E domain. Biochemical characterization of the respective recombinant Cnx1E variants revealed two different amino acid exchanges (S197F and G175D) that impair Cnx1E dimerization, thus linking Cnx1E oligomerization to Cnx1 functionality. Analysis of the Cnx1E structure identified Cnx1E active site-bound molybdate and magnesium ions, which allowed to fine-map the Cnx1E MPT-AMP-binding site.


Assuntos
Proteínas de Arabidopsis , Arabidopsis , Calnexina , Multimerização Proteica/fisiologia , Substituição de Aminoácidos , Arabidopsis/química , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Calnexina/química , Calnexina/genética , Calnexina/metabolismo , Domínio Catalítico , Coenzimas/química , Coenzimas/genética , Coenzimas/metabolismo , Metaloproteínas/química , Metaloproteínas/genética , Metaloproteínas/metabolismo , Cofatores de Molibdênio , Mutação de Sentido Incorreto , Estrutura Secundária de Proteína , Pteridinas/química , Pteridinas/metabolismo
4.
Proc Natl Acad Sci U S A ; 112(34): 10691-6, 2015 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-26261323

RESUMO

The cytoplasmic membrane is probably the most important physical barrier between microbes and the surrounding habitat. Aminoacylation of the polar head group of the phospholipid phosphatidylglycerol (PG) catalyzed by Ala-tRNA(Ala)-dependent alanyl-phosphatidylglycerol synthase (A-PGS) or by Lys-tRNA(Lys)-dependent lysyl-phosphatidylglycerol synthase (L-PGS) enables bacteria to cope with cationic peptides that are harmful to the integrity of the cell membrane. Accordingly, these synthases also have been designated as multiple peptide resistance factors (MprF). They consist of a separable C-terminal catalytic domain and an N-terminal transmembrane flippase domain. Here we present the X-ray crystallographic structure of the catalytic domain of A-PGS from the opportunistic human pathogen Pseudomonas aeruginosa. In parallel, the structure of the related lysyl-phosphatidylglycerol-specific L-PGS domain from Bacillus licheniformis in complex with the substrate analog L-lysine amide is presented. Both proteins reveal a continuous tunnel that allows the hydrophobic lipid substrate PG and the polar aminoacyl-tRNA substrate to access the catalytic site from opposite directions. Substrate recognition of A-PGS versus L-PGS was investigated using misacylated tRNA variants. The structural work presented here in combination with biochemical experiments using artificial tRNA or artificial lipid substrates reveals the tRNA acceptor stem, the aminoacyl moiety, and the polar head group of PG as the main determinants for substrate recognition. A mutagenesis approach yielded the complementary amino acid determinants of tRNA interaction. These results have broad implications for the design of L-PGS and A-PGS inhibitors that could render microbial pathogens more susceptible to antimicrobial compounds.


Assuntos
Aminoaciltransferases/química , Bacillus/enzimologia , Proteínas de Bactérias/química , Fosfatidilgliceróis/metabolismo , Pseudomonas aeruginosa/enzimologia , Fatores R , RNA de Transferência de Alanina/metabolismo , RNA de Transferência de Lisina/metabolismo , Aminoacilação , Aminoaciltransferases/metabolismo , Bacillus/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Domínio Catalítico , Cristalografia por Raios X , Interações Hidrofóbicas e Hidrofílicas , Lisina/biossíntese , Modelos Moleculares , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação de Ácido Nucleico , Fosfatidilgliceróis/biossíntese , Conformação Proteica , Pseudomonas aeruginosa/genética , Proteínas Recombinantes de Fusão/química , Relação Estrutura-Atividade , Especificidade por Substrato
5.
Science ; 348(6239): 1106-12, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-26045430

RESUMO

The discovery of Streptomyces-produced streptomycin founded the age of tuberculosis therapy. Despite the subsequent development of a curative regimen for this disease, tuberculosis remains a worldwide problem, and the emergence of multidrug-resistant Mycobacterium tuberculosis has prioritized the need for new drugs. Here we show that new optimized derivatives from Streptomyces-derived griselimycin are highly active against M. tuberculosis, both in vitro and in vivo, by inhibiting the DNA polymerase sliding clamp DnaN. We discovered that resistance to griselimycins, occurring at very low frequency, is associated with amplification of a chromosomal segment containing dnaN, as well as the ori site. Our results demonstrate that griselimycins have high translational potential for tuberculosis treatment, validate DnaN as an antimicrobial target, and capture the process of antibiotic pressure-induced gene amplification.


Assuntos
Antituberculosos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Terapia de Alvo Molecular , Mycobacterium tuberculosis/efeitos dos fármacos , Peptídeos Cíclicos/farmacologia , Tuberculose Resistente a Múltiplos Medicamentos/tratamento farmacológico , Animais , Antituberculosos/química , Antituberculosos/uso terapêutico , Linhagem Celular Tumoral , Cristalografia por Raios X , DNA Polimerase Dirigida por DNA , Modelos Animais de Doenças , Desenho de Fármacos , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Mycobacterium smegmatis/efeitos dos fármacos , Mycobacterium smegmatis/enzimologia , Mycobacterium tuberculosis/enzimologia , Peptídeos Cíclicos/química , Peptídeos Cíclicos/uso terapêutico , Estrutura Secundária de Proteína , Streptomyces/química , Streptomyces/efeitos dos fármacos , Streptomyces/metabolismo , Tuberculose Resistente a Múltiplos Medicamentos/microbiologia
6.
J Mol Biol ; 426(19): 3272-3286, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25083922

RESUMO

The isobacteriochlorin heme d1 serves as an essential cofactor in the cytochrome cd1 nitrite reductase NirS that plays an important role for denitrification. During the biosynthesis of heme d1, the enzyme siroheme decarboxylase catalyzes the conversion of siroheme to 12,18-didecarboxysiroheme. This enzyme was discovered recently (Bali S, Lawrence AD, Lobo SA, Saraiva LM, Golding BT, Palmer DJ et al. Molecular hijacking of siroheme for the synthesis of heme and d1 heme. Proc Natl Acad Sci USA 2011;108:18260-5) and is only scarcely characterized. Here, we present the crystal structure of the siroheme decarboxylase from Hydrogenobacter thermophilus representing the first three-dimensional structure for this type of enzyme. The overall structure strikingly resembles those of transcriptional regulators of the Lrp/AsnC family. Moreover, the structure of the enzyme in complex with a substrate analog reveals first insights into its active-site architecture. Through site-directed mutagenesis and subsequent biochemical characterization of the enzyme variants, two conserved histidine residues within the active site are identified to be involved in substrate binding and catalysis. Based on our results, we propose a potential catalytic mechanism for the enzymatic reaction catalyzed by the siroheme decarboxylase.


Assuntos
Bactérias/enzimologia , Proteínas de Bactérias/química , Carboxiliases/química , Histidina/química , Ferro/química , Uroporfirinas/química , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/ultraestrutura , Sítios de Ligação , Carboxiliases/ultraestrutura , Domínio Catalítico , Descarboxilação , Heme/análogos & derivados , Heme/biossíntese , Heme/química , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Alinhamento de Sequência
7.
Structure ; 21(12): 2098-9, 2013 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-24315456

RESUMO

Secretins are major constituents of bacterial type III secretion systems (T3SS). In this issue of Structure, Kowal and colleagues report on the cryo-EM structure of the native YscC secretin from Yersinia, revealing its internal symmetry and mode of length adaptation.


Assuntos
Proteínas de Bactérias/química , Secretina/química , Tripsina/metabolismo , Yersinia enterocolitica/metabolismo
8.
Elife ; 2: e00792, 2013 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-23908767

RESUMO

Injectisomes are multi-protein transmembrane machines allowing pathogenic bacteria to inject effector proteins into eukaryotic host cells, a process called type III secretion. Here we present the first three-dimensional structure of Yersinia enterocolitica and Shigella flexneri injectisomes in situ and the first structural analysis of the Yersinia injectisome. Unexpectedly, basal bodies of injectisomes inside the bacterial cells showed length variations of 20%. The in situ structures of the Y. enterocolitica and S. flexneri injectisomes had similar dimensions and were significantly longer than the isolated structures of related injectisomes. The crystal structure of the inner membrane injectisome component YscD appeared elongated compared to a homologous protein, and molecular dynamics simulations documented its elongation elasticity. The ring-shaped secretin YscC at the outer membrane was stretched by 30-40% in situ, compared to its isolated liposome-embedded conformation. We suggest that elasticity is critical for some two-membrane spanning protein complexes to cope with variations in the intermembrane distance. DOI:http://dx.doi.org/10.7554/eLife.00792.001.


Assuntos
Proteínas de Membrana/metabolismo , Yersinia enterocolitica/metabolismo , Microscopia Crioeletrônica , Proteínas de Membrana/química , Pressão Osmótica , Conformação Proteica
9.
Proc Natl Acad Sci U S A ; 110(6): 2094-8, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23341615

RESUMO

Photosynthesis uses chlorophylls for the conversion of light into chemical energy, the driving force of life on Earth. During chlorophyll biosynthesis in photosynthetic bacteria, cyanobacteria, green algae and gymnosperms, dark-operative protochlorophyllide oxidoreductase (DPOR), a nitrogenase-like metalloenzyme, catalyzes the chemically challenging two-electron reduction of the fully conjugated ring system of protochlorophyllide a. The reduction of the C-17=C-18 double bond results in the characteristic ring architecture of all chlorophylls, thereby altering the absorption properties of the molecule and providing the basis for light-capturing and energy-transduction processes of photosynthesis. We report the X-ray crystallographic structure of the substrate-bound, ADP-aluminium fluoride-stabilized (ADP·AlF(3)-stabilized) transition state complex between the DPOR components L(2) and (NB)(2) from the marine cyanobacterium Prochlorococcus marinus. Our analysis permits a thorough investigation of the dynamic interplay between L(2) and (NB)(2). Upon complex formation, substantial ATP-dependent conformational rearrangements of L(2) trigger the protein-protein interactions with (NB)(2) as well as the electron transduction via redox-active [4Fe-4S] clusters. We also present the identification of artificial "small-molecule substrates" of DPOR in correlation with those of nitrogenase. The catalytic differences and similarities between DPOR and nitrogenase have broad implications for the energy transduction mechanism of related multiprotein complexes that are involved in the reduction of chemically stable double and/or triple bonds.


Assuntos
Difosfato de Adenosina/química , Difosfato de Adenosina/metabolismo , Compostos de Alumínio/química , Compostos de Alumínio/metabolismo , Fluoretos/química , Fluoretos/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/química , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Protoclorifilida/química , Protoclorifilida/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X , Estabilidade Enzimática , Substâncias Macromoleculares/química , Substâncias Macromoleculares/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Mutagênese Sítio-Dirigida , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Prochlorococcus/enzimologia , Prochlorococcus/genética , Conformação Proteica , Subunidades Proteicas , Homologia de Sequência de Aminoácidos
10.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 1): 114-20, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23275169

RESUMO

In recent decades, several canonical serine protease inhibitor families have been classified and characterized. In contrast to most trypsin inhibitors, those from garden four o'clock (Mirabilis jalapa) and spinach (Spinacia oleracea) do not share sequence similarity and have been proposed to form the new Mirabilis serine protease inhibitor family. These 30-40-amino-acid inhibitors possess a defined disulfide-bridge topology and belong to the cystine-knot miniproteins (knottins). To date, no atomic structure of this inhibitor family has been solved. Here, the first structure of S. oleracea trypsin inhibitor III (SOTI-III), in complex with bovine pancreatic trypsin, is reported. The inhibitor was synthesized by solid-phase peptide synthesis on a multi-milligram scale and was assayed to test its inhibitory activity and binding properties. The structure confirmed the proposed cystine-bridge topology. The structural features of SOTI-III suggest that it belongs to a new canonical serine protease inhibitor family with promising properties for use in protein-engineering and medical applications.


Assuntos
Aprotinina/química , Proteínas de Plantas/química , Spinacia oleracea/química , Animais , Bovinos , Cristalização , Cristalografia por Raios X , Mirabilis/química , Proteínas de Plantas/farmacologia , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/farmacologia
11.
J Mol Biol ; 425(8): 1302-17, 2013 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-23274142

RESUMO

The complement system as a major part of innate immunity is the first line of defense against invading microorganisms. Orchestrated by more than 60 proteins, its major task is to discriminate between host cells and pathogens and to initiate immune response. Additional recognition of necrotic or apoptotic cells demands a fine-tune regulation of this powerful system. C4b-binding protein (C4BP) is the major inhibitor of the classical complement and lectin pathway. The crystal structure of the human C4BP oligomerization domain in its 7α isoform and molecular simulations provide first structural insights of C4BP oligomerization. The heptameric core structure is stabilized by intermolecular disulfide bonds. In addition, thermal shift assays indicate that layers of electrostatic interactions mainly contribute to the extraordinary thermodynamic stability of the complex. These findings make C4BP a promising scaffold for multivalent ligand display with applications in immunology and biological chemistry.


Assuntos
Proteína de Ligação ao Complemento C4b/química , Cristalografia por Raios X , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Conformação Proteica , Multimerização Proteica , Eletricidade Estática , Termodinâmica
12.
J Biol Chem ; 287(43): 35796-803, 2012 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-22936808

RESUMO

Pathogens often rely on thermosensing to adjust virulence gene expression. In yersiniae, important virulence-associated traits are under the control of the master regulator RovA, which uses a built-in thermosensor to control its activity. Thermal upshifts encountered upon host entry induce conformational changes in the RovA dimer that attenuate DNA binding and render the protein more susceptible to proteolysis. Here, we report the crystal structure of RovA in the free and DNA-bound forms and provide evidence that thermo-induced loss of RovA activity is promoted mainly by a thermosensing loop in the dimerization domain and residues in the adjacent C-terminal helix. These determinants allow partial unfolding of the regulator upon an upshift to 37 °C. This structural distortion is transmitted to the flexible DNA-binding domain of RovA. RovA contacts mainly the DNA backbone in a low-affinity binding mode, which allows the immediate release of RovA from its operator sites. We also show that SlyA, a close homolog of RovA from Salmonella with a very similar structure, is not a thermosensor and remains active and stable at 37 °C. Strikingly, changes in only three amino acids, reflecting evolutionary replacements in SlyA, result in a complete loss of the thermosensing properties of RovA and prevent degradation. In conclusion, only minor alterations can transform a thermotolerant regulator into a thermosensor that allows adjustment of virulence and fitness determinants to their thermal environment.


Assuntos
Proteínas de Bactérias/química , Dobramento de Proteína , Fatores de Transcrição/química , Fatores de Virulência/química , Yersinia pseudotuberculosis/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , DNA Bacteriano/química , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Temperatura Alta , Ligação Proteica , Estrutura Terciária de Proteína , Proteólise , Salmonella/química , Salmonella/genética , Salmonella/metabolismo , Salmonella/patogenicidade , Homologia Estrutural de Proteína , Relação Estrutura-Atividade , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/patogenicidade
13.
J Biol Chem ; 287(46): 39224-32, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-22965230

RESUMO

Hepatitis C virus (HCV) NS3-4A protease is essential for viral replication. All current small molecular weight drugs against NS3-4A are substrate peptidomimetics that have a similar binding and resistance profile. We developed inhibitory peptides (IPs) capping the active site and binding via a novel "tyrosine" finger at an alternative NS3-4A site that is of particular interest for further HCV drug development. The peptides are not cleaved due to a combination of geometrical constraints and impairment of the oxyanion hole function. Selection and optimization through combinatorial phagemid display, protein crystallography, and further modifications resulted in a 32-amino acid peptide with a K(i) of 0.53 nm. Inhibition of viral replication in cell culture was demonstrated by fusion to a cell-penetrating peptide. Negligible susceptibility to known (A156V and R155K) resistance mutations of the NS3-4A protease was observed. This work shows for the first time that antiviral peptides can target an intracellular site and reveals a novel druggable site on the HCV protease.


Assuntos
Proteínas de Transporte/química , Mutação , Proteínas não Estruturais Virais/química , Antivirais/farmacologia , Sítios de Ligação , Peptídeos Penetradores de Células/química , Cristalografia/métodos , Desenho de Fármacos , Farmacorresistência Viral/efeitos dos fármacos , Farmacorresistência Viral/genética , Humanos , Concentração Inibidora 50 , Peptídeos e Proteínas de Sinalização Intracelular , Cinética , Modelos Moleculares , Conformação Molecular , Biblioteca de Peptídeos , Peptídeos/química , Solventes/química
14.
Protein Sci ; 21(10): 1528-39, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22887347

RESUMO

The physiological relevance of contacts in crystal lattices often remains elusive. This was also the case for the complex between the invasion protein internalin B (InlB) from Listeria monocytogenes and its host cell receptor, the human receptor tyrosine kinase (RTK) MET. InlB is a MET agonist and induces bacterial host cell invasion. Activation of RTKs generally involves ligand-induced dimerization of the receptor ectodomain. The two currently available crystal structures of the InlB:MET complex show the same arrangement of InlB and MET in a 1:1 complex, but different dimeric 2:2 assemblies. Only one of these 2:2 assemblies is predicted to be stable by a computational procedure. This assembly is mainly stabilized by a contact between the Cap domain of InlB from one and the Sema domain of MET from another 1:1 complex. Here, we probe the physiological relevance of this interaction. We generated variants of the leucine-rich repeat (LRR) protein InlB by inserting an additional repeat between the first and the second LRR. This should allow formation of the 1:1 complex but disrupt the potential 2:2 complex involving the Cap-Sema contact due to steric distortions. A crystal structure of one of the engineered proteins showed that it folded properly. Binding affinity to MET was comparable to that of wild-type InlB. The InlB variant induced MET phosphorylation and cell scatter like wild-type InlB. These results suggest that the Cap-Sema interaction is not physiologically relevant and support the previously proposed assembly, in which a 2:2 InlB:MET complex is built around a ligand dimer.


Assuntos
Proteínas de Bactérias/química , Proteínas de Membrana/química , Proteínas Proto-Oncogênicas c-met/química , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cristalografia , Humanos , Leucina/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Fosforilação , Engenharia de Proteínas , Proteínas Proto-Oncogênicas c-met/metabolismo , Alinhamento de Sequência
16.
PLoS One ; 6(12): e27829, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22174749

RESUMO

Studying the biophysical characteristics of glycosylated proteins and solving their three-dimensional structures requires homogeneous recombinant protein of high quality.We introduce here a new approach to produce glycoproteins in homogenous form with the well-established, glycosylation mutant CHO Lec3.2.8.1 cells. Using preparative cell sorting, stable, high-expressing GFP 'master' cell lines were generated that can be converted fast and reliably by targeted integration via Flp recombinase-mediated cassette exchange (RMCE) to produce any glycoprotein. Small-scale transient transfection of HEK293 cells was used to identify genetically engineered constructs suitable for constructing stable cell lines. Stable cell lines expressing 10 different proteins were established. The system was validated by expression, purification, deglycosylation and crystallization of the heavily glycosylated luminal domains of lysosome-associated membrane proteins (LAMP).


Assuntos
Fenômenos Biofísicos , Técnicas de Cultura de Células/métodos , Linhagem Celular/citologia , Glicoproteínas/biossíntese , Glicoproteínas/química , Animais , Células CHO , Cricetinae , Cricetulus , Cristalização , DNA Nucleotidiltransferases/metabolismo , Vetores Genéticos/genética , Glicosilação , Proteínas de Fluorescência Verde/genética , Humanos , Proteínas Luminescentes/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes/biossíntese , Recombinação Genética/genética , Proteína Vermelha Fluorescente
17.
Nat Chem Biol ; 8(1): 117-24, 2011 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-22138621

RESUMO

Polyketides are structurally diverse and medically important natural products that have various biological activities. During biosynthesis, chain elongation uses activated dicarboxylic acid building blocks, and their availability therefore limits side chain variation in polyketides. Recently, the crotonyl-CoA carboxylase-reductase (CCR) class of enzymes was identified in primary metabolism and was found to be involved in extender-unit biosynthesis of polyketides. These enzymes are, in theory, capable of forming dicarboxylic acids that show any side chain from the respective unsaturated fatty acid precursor. To our knowledge, we here report the first crystal structure of a CCR, the hexylmalonyl-CoA synthase from Streptomyces sp. JS360, in complex with its substrate. Structural analysis and biochemical characterization of the enzyme, including active site mutations, are reported. Our analysis reveals how primary metabolic CCRs can evolve to produce various dicarboxylic acid building blocks, setting the stage to use CCRs for the production of unique extender units and, consequently, altered polyketides.


Assuntos
Acil-CoA Desidrogenases/química , Ciclo do Carbono , Policetídeos/química , Streptomyces/enzimologia , Acil-CoA Desidrogenases/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Biocatálise , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Policetídeos/metabolismo , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Alinhamento de Sequência , Especificidade por Substrato
18.
Structure ; 19(9): 1294-306, 2011 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-21893288

RESUMO

Actin assembly beneath enterohemorrhagic E. coli (EHEC) attached to its host cell is triggered by the intracellular interaction of its translocated effector proteins Tir and EspF(U) with human IRSp53 family proteins and N-WASP. Here, we report the structure of the N-terminal I-BAR domain of IRSp53 in complex with a Tir-derived peptide, in which the homodimeric I-BAR domain binds two Tir molecules aligned in parallel. This arrangement provides a protein scaffold linking the bacterium to the host cell's actin polymerization machinery. The structure uncovers a specific peptide-binding site on the I-BAR surface, conserved between IRSp53 and IRTKS. The Tir Asn-Pro-Tyr (NPY) motif, essential for pedestal formation, is specifically recognized by this binding site. The site was confirmed by mutagenesis and in vivo-binding assays. It is possible that IRSp53 utilizes the NPY-binding site for additional interactions with as yet unknown partners within the host cell.


Assuntos
Escherichia coli O157 , Proteínas de Escherichia coli/química , Proteínas do Tecido Nervoso/química , Fragmentos de Peptídeos/química , Receptores de Superfície Celular/química , Motivos de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Células COS , Calorimetria , Chlorocebus aethiops , Cristalografia por Raios X , Proteínas de Escherichia coli/genética , Interações Hospedeiro-Patógeno , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Imunoprecipitação , Modelos Moleculares , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fragmentos de Peptídeos/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptores de Superfície Celular/genética , Termodinâmica
19.
J Biol Chem ; 286(30): 26754-67, 2011 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-21632530

RESUMO

During the biosynthesis of heme d(1), the essential cofactor of cytochrome cd(1) nitrite reductase, the NirE protein catalyzes the methylation of uroporphyrinogen III to precorrin-2 using S-adenosyl-L-methionine (SAM) as the methyl group donor. The crystal structure of Pseudomonas aeruginosa NirE in complex with its substrate uroporphyrinogen III and the reaction by-product S-adenosyl-L-homocysteine (SAH) was solved to 2.0 Å resolution. This represents the first enzyme-substrate complex structure for a SAM-dependent uroporphyrinogen III methyltransferase. The large substrate binds on top of the SAH in a "puckered" conformation in which the two pyrrole rings facing each other point into the same direction either upward or downward. Three arginine residues, a histidine, and a methionine are involved in the coordination of uroporphyrinogen III. Through site-directed mutagenesis of the nirE gene and biochemical characterization of the corresponding NirE variants the amino acid residues Arg-111, Glu-114, and Arg-149 were identified to be involved in NirE catalysis. Based on our structural and biochemical findings, we propose a potential catalytic mechanism for NirE in which the methyl transfer reaction is initiated by an arginine catalyzed proton abstraction from the C-20 position of the substrate.


Assuntos
Proteínas de Bactérias/química , Metiltransferases/química , Pseudomonas aeruginosa/enzimologia , Uroporfirinogênios/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Catálise , Cristalografia por Raios X , Heme/análogos & derivados , Heme/biossíntese , Heme/química , Heme/metabolismo , Metiltransferases/genética , Metiltransferases/metabolismo , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Estrutura Terciária de Proteína , Pseudomonas aeruginosa/genética , Uroporfirinogênios/genética , Uroporfirinogênios/metabolismo
20.
J Biol Chem ; 286(20): 17593-600, 2011 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21454585

RESUMO

Despite the widespread biological function of carbohydrates, the polysaccharide synthesis mechanisms of glycosyltransferases remain largely unexplored. Bacterial levansucrases (glycoside hydrolase family 68) synthesize high molecular weight, ß-(2,6)-linked levan from sucrose by transfer of fructosyl units. The kinetic and biochemical characterization of Bacillus megaterium levansucrase SacB variants Y247A, Y247W, N252A, D257A, and K373A reveal novel surface motifs remote from the sucrose binding site with distinct influence on the polysaccharide product spectrum. The wild type activity (k(cat)) and substrate affinity (K(m)) are maintained. The structures of the SacB variants reveal clearly distinguishable subsites for polysaccharide synthesis as well as an intact active site architecture. These results lead to a new understanding of polysaccharide synthesis mechanisms. The identified surface motifs are discussed in the context of related glycosyltransferases.


Assuntos
Bacillus megaterium/enzimologia , Proteínas de Bactérias/metabolismo , Hexosiltransferases/metabolismo , Polissacarídeos Bacterianos/biossíntese , Motivos de Aminoácidos , Substituição de Aminoácidos , Bacillus megaterium/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Hexosiltransferases/química , Hexosiltransferases/genética , Mutação de Sentido Incorreto , Polissacarídeos Bacterianos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...