Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
PLoS Pathog ; 17(4): e1009477, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33857257

RESUMO

The lack of efficient methods to control the major diseases of crops most important to agriculture leads to huge economic losses and seriously threatens global food security. Many of the most important microbial plant pathogens, including bacteria, fungi, and oomycetes, secrete necrosis- and ethylene-inducing peptide 1 (Nep1)-like proteins (NLPs), which critically contribute to the virulence and spread of the disease. NLPs are cytotoxic to eudicot plants, as they disturb the plant plasma membrane by binding to specific plant membrane sphingolipid receptors. Their pivotal role in plant infection and broad taxonomic distribution makes NLPs a promising target for the development of novel phytopharmaceutical compounds. To identify compounds that bind to NLPs from the oomycetes Pythium aphanidermatum and Phytophthora parasitica, a library of 587 small molecules, most of which are commercially unavailable, was screened by surface plasmon resonance. Importantly, compounds that exhibited the highest affinity to NLPs were also found to inhibit NLP-mediated necrosis in tobacco leaves and Phytophthora infestans growth on potato leaves. Saturation transfer difference-nuclear magnetic resonance and molecular modelling of the most promising compound, anthranilic acid derivative, confirmed stable binding to the NLP protein, which resulted in decreased necrotic activity and reduced ion leakage from tobacco leaves. We, therefore, confirmed that NLPs are an appealing target for the development of novel phytopharmaceutical agents and strategies, which aim to directly interfere with the function of these major microbial virulence factors. The compounds identified in this study represent lead structures for further optimization and antimicrobial product development.


Assuntos
Phytophthora/patogenicidade , Doenças das Plantas/prevenção & controle , Pythium/patogenicidade , Solanum tuberosum/genética , Simulação de Dinâmica Molecular , Necrose , Phytophthora/genética , Doenças das Plantas/parasitologia , Folhas de Planta/genética , Folhas de Planta/parasitologia , Pythium/genética , Solanum tuberosum/parasitologia , Ressonância de Plasmônio de Superfície , Nicotiana/genética , Nicotiana/parasitologia
2.
PLoS Pathog ; 15(9): e1007951, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31479498

RESUMO

Necrosis and ethylene-inducing peptide 1 (Nep1)-like proteins (NLPs) are secreted by several phytopathogenic microorganisms. They trigger necrosis in various eudicot plants upon binding to plant sphingolipid glycosylinositol phosphorylceramides (GIPC). Interestingly, HaNLP3 from the obligate biotroph oomycete Hyaloperonospora arabidopsidis does not induce necrosis. We determined the crystal structure of HaNLP3 and showed that it adopts the NLP fold. However, the conformations of the loops surrounding the GIPC headgroup-binding cavity differ from those of cytotoxic Pythium aphanidermatum NLPPya. Essential dynamics extracted from µs-long molecular dynamics (MD) simulations reveals a limited conformational plasticity of the GIPC-binding cavity in HaNLP3 relative to toxic NLPs. This likely precludes HaNLP3 binding to GIPCs, which is the underlying reason for the lack of toxicity. This study reveals that mutations at key protein regions cause a switch between non-toxic and toxic phenotypes within the same protein scaffold. Altogether, these data provide evidence that protein flexibility is a distinguishing trait of toxic NLPs and highlight structural determinants for a potential functional diversification of non-toxic NLPs utilized by biotrophic plant pathogens.


Assuntos
Oomicetos/genética , Oomicetos/metabolismo , Doenças das Plantas/parasitologia , Sequência de Aminoácidos , Etilenos/metabolismo , Necrose/metabolismo , Peptídeos/metabolismo , Peronospora/genética , Proteínas/metabolismo , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
3.
FASEB J ; 34(8): 11068-11086, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32648626

RESUMO

NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome is a multiprotein complex which forms within cells in response to various microbial and self-derived triggers. Mutations in the gene encoding NLRP3 cause rare cryopyrin-associated periodic syndromes (CAPS) and growing evidence links NLRP3 inflammasome to common diseases such as Alzheimer´s disease. In order to modulate different stages of NLRP3 inflammasome assembly nine peptides whose sequences correspond to segments of inflammasome components NLRP3 and apoptosis-associated speck-like protein containing a CARD (ASC) were selected. Five peptides inhibited IL-1ß release, caspase-1 activation and ASC oligomerization in response to soluble and particulate NLRP3 triggers. Modulatory peptides also attenuated IL-1ß maturation induced by constitutive CAPS-associated NLRP3 mutants. Peptide corresponding to H2-H3 segment of ASC pyrin domain selectively inhibited NLRP3 inflammasome by binding to NLRP3 pyrin domain in the micromolar range. The peptide had no effect on AIM2 and NLRC4 inflammasomes as well as NF-κB pathway. The peptide effectively dampened neutrophil infiltration in the silica-induced peritonitis and when equipped with Antennapedia or Angiopep-2 motifs crossed the blood-brain barrier in a mouse model. Our study demonstrates that peptides represent an important tool for targeting multiprotein inflammatory complexes and can serve as the basis for the development of novel anti-inflammatory strategies for neurodegeneration.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Peptídeos/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Barreira Hematoencefálica/metabolismo , Caspase 1/metabolismo , Células Cultivadas , Síndromes Periódicas Associadas à Criopirina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Feminino , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Masculino , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Infiltração de Neutrófilos/fisiologia , Peritonite/metabolismo
4.
J Infect Dis ; 222(9): 1505-1516, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31970394

RESUMO

BACKGROUND: Bacterial toxins disrupt plasma membrane integrity with multitudinous effects on host cells. The secreted pore-forming toxin listeriolysin O (LLO) of the intracellular pathogen Listeria monocytogenes promotes egress of the bacteria from vacuolar compartments into the host cytosol often without overt destruction of the infected cell. Intracellular LLO activity is tightly controlled by host factors including compartmental pH, redox, proteolytic, and proteostatic factors, and inhibited by cholesterol. METHODS: Combining infection studies of L. monocytogenes wild type and isogenic mutants together with biochemical studies with purified phospholipases, we investigate the effect of their enzymatic activities on LLO. RESULTS: Here, we show that phosphocholine (ChoP), a reaction product of the phosphatidylcholine-specific phospholipase C (PC-PLC) of L. monocytogenes, is a potent inhibitor of intra- and extracellular LLO activities. Binding of ChoP to LLO is redox-independent and leads to the inhibition of LLO-dependent induction of calcium flux, mitochondrial damage, and apoptosis. ChoP also inhibits the hemolytic activities of the related cholesterol-dependent cytolysins (CDC), pneumolysin and streptolysin. CONCLUSIONS: Our study uncovers a strategy used by L. monocytogenes to modulate cytotoxic LLO activity through the enzymatic activity of its PC-PLC. This mechanism appears to be widespread and also used by other CDC pore-forming toxin-producing bacteria.


Assuntos
Toxinas Bacterianas/antagonistas & inibidores , Proteínas de Choque Térmico/antagonistas & inibidores , Proteínas Hemolisinas/antagonistas & inibidores , Listeria monocytogenes/efeitos dos fármacos , Fosforilcolina/farmacologia , Apoptose , Cálcio/metabolismo , Caspase 3/metabolismo , Células HeLa , Humanos , Listeria monocytogenes/enzimologia , Listeria monocytogenes/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo
5.
Mol Microbiol ; 112(4): 1253-1269, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31376198

RESUMO

Proteins of the aegerolysin family have a high abundance in Fungi. Due to their specific binding to membrane lipids, and their membrane-permeabilization potential in concert with protein partner(s) belonging to a membrane-attack-complex/perforin (MACPF) superfamily, they were proposed as useful tools in different biotechnological and biomedical applications. In this work, we performed functional studies on expression of the genes encoding aegerolysin and MACPF-like proteins in Aspergillus niger. Our results suggest the sporulation process being crucial for strong induction of the expression of all these genes. However, deletion of either of the aegerolysin genes did not influence the growth, development, sporulation efficiency and phenotype of the mutants, indicating that aegerolysins are not key factors in the sporulation process. In all our expression studies we noticed a strong correlation in the expression of one aegerolysin and MACPF-like gene. Aegerolysins were confirmed to be secreted from the fungus. We also showed the specific interaction of a recombinant A. niger aegerolysin with an invertebrate-specific membrane sphingolipid. Moreover, using this protein labelled with mCherry we successfully stained insect cells membranes containing this particular sphingolipid. Our combined results suggest, that aegerolysins in this species, and probably also in other aspergilli, could be involved in defence against predators.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas Hemolisinas/metabolismo , Perforina/metabolismo , Aspergillus niger/genética , Aspergillus niger/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/genética , Proteínas Fúngicas/fisiologia , Regulação Fúngica da Expressão Gênica/genética , Proteínas Hemolisinas/fisiologia , Proteínas de Membrana/metabolismo , Perforina/genética , Esfingolipídeos/metabolismo , Esporos Fúngicos/genética , Esporos Fúngicos/metabolismo
6.
Nucleic Acids Res ; 46(18): 9432-9443, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30053203

RESUMO

The GIL01 bacteriophage is a temperate phage that infects the insect pathogen Bacillus thuringiensis. During the lytic cycle, phage gene transcription is initiated from three promoters: P1 and P2, which control the expression of the early phage genes involved in genome replication and P3, which controls the expression of the late genes responsible for virion maturation and host lysis. Unlike most temperate phages, GIL01 lysogeny is not maintained by a dedicated phage repressor but rather by the host's regulator of the SOS response, LexA. Previously we showed that the lytic cycle was induced by DNA damage and that LexA, in conjunction with phage-encoded protein gp7, repressed P1. Here we examine the lytic/lysogenic switch in more detail and show that P3 is also repressed by a LexA-gp7 complex, binding to tandem LexA boxes within the promoter. We also demonstrate that expression from P3 is considerably delayed after DNA damage, requiring the phage-encoded DNA binding protein, gp6. Surprisingly, gp6 is homologous to LexA itself and, thus, is a rare example of a LexA homologue directly activating transcription. We propose that the interplay between these two LexA family members, with opposing functions, ensures the timely expression of GIL01 phage late genes.


Assuntos
Proteínas de Bactérias/genética , Bacteriófagos/genética , Lisogenia/genética , Serina Endopeptidases/genética , Transcrição Gênica/genética , Proteínas Virais/fisiologia , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Bacillus thuringiensis/genética , Bacillus thuringiensis/metabolismo , Proteínas de Bactérias/metabolismo , Bacteriófagos/metabolismo , Sequência de Bases , Citotoxinas/genética , Citotoxinas/metabolismo , Regulação Viral da Expressão Gênica , Regiões Promotoras Genéticas , Homologia de Sequência , Serina Endopeptidases/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
7.
Int J Mol Sci ; 20(3)2019 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-30736336

RESUMO

Diarrhea caused by enterotoxigenic Escherichia coli (ETEC) is one of the leading causes of mortality in children under five years of age and is a great burden on developing countries. The major virulence factor of the bacterium is the heat-labile enterotoxin (LT), a close homologue of the cholera toxin. The toxins bind to carbohydrate receptors in the gastrointestinal tract, leading to toxin uptake and, ultimately, to severe diarrhea. Previously, LT from human- and porcine-infecting ETEC (hLT and pLT, respectively) were shown to have different carbohydrate-binding specificities, in particular with respect to N-acetyllactosamine-terminating glycosphingolipids. Here, we probed 11 single-residue variants of the heat-labile enterotoxin with surface plasmon resonance spectroscopy and compared the data to the parent toxins. In addition we present a 1.45 Å crystal structure of pLTB in complex with branched lacto-N-neohexaose (Galß4GlcNAcß6[Galß4GlcNAcß3]Galß4Glc). The largest difference in binding specificity is caused by mutation of residue 94, which links the primary and secondary binding sites of the toxins. Residue 95 (and to a smaller extent also residues 7 and 18) also contribute, whereas residue 4 shows no effect on monovalent binding of the ligand and may rather be important for multivalent binding and avidity.


Assuntos
Escherichia coli Enterotoxigênica/genética , Enterotoxinas/química , Enterotoxinas/genética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Sítios de Ligação , Carboidratos/química , Cristalografia por Raios X , Humanos , Conformação Molecular , Ligação Proteica , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície
8.
PLoS Pathog ; 12(4): e1005567, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27082955

RESUMO

Cholera is the prime example of blood-group-dependent diseases, with individuals of blood group O experiencing the most severe symptoms. The cholera toxin is the main suspect to cause this relationship. We report the high-resolution crystal structures (1.1-1.6 Å) of the native cholera toxin B-pentamer for both classical and El Tor biotypes, in complexes with relevant blood group determinants and a fragment of its primary receptor, the GM1 ganglioside. The blood group A determinant binds in the opposite orientation compared to previously published structures of the cholera toxin, whereas the blood group H determinant, characteristic of blood group O, binds in both orientations. H-determinants bind with higher affinity than A-determinants, as shown by surface plasmon resonance. Together, these findings suggest why blood group O is a risk factor for severe cholera.


Assuntos
Sistema ABO de Grupos Sanguíneos/química , Sistema ABO de Grupos Sanguíneos/metabolismo , Toxina da Cólera/química , Toxina da Cólera/metabolismo , Cólera/sangue , Sequência de Bases , Cristalografia por Raios X , Humanos , Dados de Sequência Molecular , Conformação Proteica , Ressonância de Plasmônio de Superfície
9.
Bioconjug Chem ; 29(8): 2763-2775, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-30024737

RESUMO

Affinity chromatography based on bacterial immunoglobulin (Ig)-binding proteins represents the cornerstone of therapeutic antibody downstream processing. However, there is a pressing need for more robust affinity ligands that would withstand the harsh column sanitization conditions, while still displaying high selectivity for antibodies. Here, we report the development of linear peptide IgG ligands, identified from combinatorial phage-display library screens. The lead peptide was shown to compete with staphylococcal protein A for the IgG Fc region. Trimming analysis and alanine scanning revealed the minimal structural requirements of the peptide for Fc binding, and the minimized peptide GSYWYQVWF recognized all human IgG subtypes. Mutation of glutamine located at the nonessential position 6 to aspartate led to the optimized peptide GSYWYDVWF with 18-fold higher affinity ( KD app. 0.6 µM) compared to the parent peptide. When coupled to paramagnetic beads or a chromatographic matrix, the optimized ligand was shown to selectively enrich antibodies from complex protein mixtures.


Assuntos
Fragmentos Fc das Imunoglobulinas/química , Imunoglobulina G/química , Peptídeos/metabolismo , Sequência de Aminoácidos , Bacteriófagos/genética , Cromatografia de Afinidade/métodos , Ensaio de Imunoadsorção Enzimática , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Imunoglobulina G/genética , Ligantes , Peptídeos/química , Proteína Estafilocócica A/metabolismo , Ressonância de Plasmônio de Superfície
10.
Langmuir ; 34(30): 8983-8993, 2018 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-29983071

RESUMO

Understanding of the interactions between proteins and natural and artificially prepared lipid membrane surfaces and embedded nonpolar cores is important in studies of physiological processes and their pathologies and is applicable to nanotechnologies. In particular, rapidly growing interest in cellular droplets defines the need for simplified biomimetic lipid model systems to overcome in vivo complexity and variability. We present a protocol for the preparation of kinetically stable nanoemulsions with nanodroplets composed of sphingomyelin (SM) and cholesterol (Chol), as amphiphilic surfactants, and trioleoylglycerol (TOG), at various molar ratios. To prepare stable SM/Chol-coated monodisperse lipid nanodroplets, we modified a reverse phase evaporation method and combined it with ultrasonication. Lipid composition, ζ-potential, gyration and hydrodynamic radius, shape, and temporal stability of the lipid nanodroplets were characterized and compared to extruded SM/Chol large unilamellar vesicles. Lipid nanodroplets and large unilamellar vesicles with theoretical SM/Chol/TOG molar ratios of 1/1/4.7 and 4/1/11.7 were further investigated for the orientational order of their interfacial water molecules using a second harmonic scattering technique, and for interactions with the SM-binding and Chol-binding pore-forming toxins equinatoxin II and perfringolysin O, respectively. The surface characteristics (ζ-potential, orientational order of interfacial water molecules) and binding of these proteins to the nanodroplet SM/Chol monolayers were similar to those for the SM/Chol bilayers of the large unilamellar vesicles and SM/Chol Langmuir monolayers, in terms of their surface structures. We propose that such SM/Chol/TOG nanoparticles with the required lipid compositions can serve as experimental models for monolayer membrane to provide a system that imitates the natural lipid droplets.


Assuntos
Colesterol/química , Lipídeos/química , Nanoestruturas/química , Proteínas/metabolismo , Esfingomielinas/química , Ligação Proteica , Proteínas/química , Trioleína/química , Lipossomas Unilamelares/química , Água/química
11.
J Enzyme Inhib Med Chem ; 33(1): 1239-1247, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30141354

RESUMO

Autolysin E (AtlE) is a cell wall degrading enzyme that catalyzes the hydrolysis of the ß-1,4-glycosidic bond between the N-acetylglucosamine and N-acetylmuramic acid units of the bacterial peptidoglycan. Using our recently determined crystal structure of AtlE from Staphylococcus aureus and a combination of pharmacophore modeling, similarity search, and molecular docking, a series of (Phenylureido)piperidinyl benzamides were identified as potential binders and surface plasmon resonance (SPR) and saturation-transfer difference (STD) NMR experiments revealed that discovered compounds bind to AtlE in a lower micromolar range. (phenylureido)piperidinyl benzamides are the first reported non-substrate-like compounds that interact with this enzyme and enable further study of the interaction of small molecules with bacterial AtlE as potential inhibitors of this target.


Assuntos
Antibacterianos/farmacologia , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Piperidinas/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Antibacterianos/síntese química , Antibacterianos/química , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Testes de Sensibilidade Microbiana , Modelos Moleculares , Estrutura Molecular , N-Acetil-Muramil-L-Alanina Amidase/química , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Piperidinas/síntese química , Piperidinas/química , Staphylococcus aureus/enzimologia , Relação Estrutura-Atividade
12.
Drug Metab Dispos ; 45(8): 974-976, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28536098

RESUMO

Statins are well known lipid lowering agents that inhibit the enzyme 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase. They also activate drug metabolism but their exact receptor-mediated action has not been proven so far. We tested whether atorvastatin and rosuvastatin are direct ligands of human constitutive androstane receptor (CAR). We measured binding activities of atorvastatin and rosuvastatin to the human constitutive androstane receptor/retinoid X receptor α ligand-binding domain (CAR/RXRα-LBD) heterodimer with surface plasmon resonance (SPR). Additionally, three-dimensional models of CAR/RXRα-LBD were constructed by ligand-based and structure-based in silico modeling. Experiments and computational modeling show that atorvastatin and rosuvastatin bind to the human CAR/RXRα-LBD heterodimer, suggesting both can modulate the activity of CAR through direct interaction with the LBD of this receptor. We confirm that atorvastatin and rosuvastatin are direct ligands of CAR. The clinical consequences of CAR activation by statins are in their potential drug-drug interactions, and changes in glucose and energy metabolism.


Assuntos
Atorvastatina/metabolismo , Simulação por Computador , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptor X Retinoide alfa/metabolismo , Rosuvastatina Cálcica/metabolismo , Anticolesterolemiantes/metabolismo , Receptor Constitutivo de Androstano , Células Hep G2 , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/metabolismo , Ligantes , Ligação Proteica/fisiologia
13.
Nucleic Acids Res ; 43(15): 7315-29, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26138485

RESUMO

The SOS response in Eubacteria is a global response to DNA damage and its activation is increasingly associated with the movement of mobile genetic elements. The temperate phage GIL01 is induced into lytic growth using the host's SOS response to genomic stress. LexA, the SOS transcription factor, represses bacteriophage transcription by binding to a set of SOS boxes in the lysogenic promoter P1. However, LexA is unable to efficiently repress GIL01 transcription unless the small phage-encoded protein gp7 is also present. We found that gp7 forms a stable complex with LexA that enhances LexA binding to phage and cellular SOS sites and interferes with RecA-mediated auto-cleavage of LexA, the key step in the initiation of the SOS response. Gp7 did not bind DNA, alone or when complexed with LexA. Our findings suggest that gp7 induces a LexA conformation that favors DNA binding but disfavors LexA auto-cleavage, thereby altering the dynamics of the cellular SOS response. This is the first account of an accessory factor interacting with LexA to regulate transcription.


Assuntos
Fagos Bacilares/genética , Proteínas de Bactérias/metabolismo , Regulação Viral da Expressão Gênica , Recombinases Rec A/metabolismo , Proteínas Repressoras/metabolismo , Resposta SOS em Genética/genética , Serina Endopeptidases/metabolismo , Proteínas Virais/metabolismo , DNA/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Transcrição Gênica
14.
EMBO Rep ; 15(11): 1154-62, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25216944

RESUMO

Two mechanisms have emerged as major regulators of membrane shape: BAR domain-containing proteins, which induce invaginations and protrusions, and nuclear promoting factors, which cause generation of branched actin filaments that exert mechanical forces on membranes. While a large body of information exists on interactions of BAR proteins with membranes and regulatory proteins of the cytoskeleton, little is known about connections between these two processes. Here, we show that the F-BAR domain protein pacsin2 is able to associate with actin filaments using the same concave surface employed to bind to membranes, while some other tested N-BAR and F-BAR proteins (endophilin, CIP4 and FCHO2) do not associate with actin. This finding reveals a new level of complexity in membrane remodeling processes.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Citoesqueleto de Actina/química , Proteínas Adaptadoras de Transdução de Sinal/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Membrana Celular/metabolismo , Galinhas , Proteínas Associadas aos Microtúbulos/metabolismo , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Ligação Proteica
15.
Nucleic Acids Res ; 42(8): 5191-201, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24569352

RESUMO

Aminoacyl-tRNA synthetases (aaRS) are essential enzymes catalyzing the formation of aminoacyl-tRNAs, the immediate precursors for encoded peptides in ribosomal protein synthesis. Previous studies have suggested a link between tRNA aminoacylation and high-molecular-weight cellular complexes such as the cytoskeleton or ribosomes. However, the structural basis of these interactions and potential mechanistic implications are not well understood. To biochemically characterize these interactions we have used a system of two interacting archaeal aaRSs: an atypical methanogenic-type seryl-tRNA synthetase and an archaeal ArgRS. More specifically, we have shown by thermophoresis and surface plasmon resonance that these two aaRSs bind to the large ribosomal subunit with micromolar affinities. We have identified the L7/L12 stalk and the proteins located near the stalk base as the main sites for aaRS binding. Finally, we have performed a bioinformatics analysis of synonymous codons in the Methanothermobacter thermautotrophicus genome that supports a mechanism in which the deacylated tRNAs may be recharged by aaRSs bound to the ribosome and reused at the next occurrence of a codon encoding the same amino acid. These results suggest a mechanism of tRNA recycling in which aaRSs associate with the L7/L12 stalk region to recapture the tRNAs released from the preceding ribosome in polysomes.


Assuntos
Aminoacil-tRNA Sintetases/metabolismo , Archaea/enzimologia , Biossíntese de Proteínas , RNA de Transferência/metabolismo , Ribossomos/enzimologia , Arginina-tRNA Ligase/metabolismo , Genoma Arqueal , Methanobacteriaceae/genética , Proteínas Ribossômicas/metabolismo , Serina-tRNA Ligase/metabolismo
16.
Acta Chim Slov ; 63(3): 424-39, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27640371

RESUMO

Physical and functional interactions between molecules in living systems are central to all biological processes. Identification of protein complexes therefore is becoming increasingly important to gain a molecular understanding of cells and organisms. Several powerful methodologies and techniques have been developed to study molecular interactions and thus help elucidate their nature and role in biology as well as potential ways how to interfere with them. All different techniques used in these studies have their strengths and weaknesses and since they are mostly employed in in vitro conditions, a single approach can hardly accurately reproduce interactions that happen under physiological conditions. However, complementary usage of as many as possible available techniques can lead to relatively realistic picture of the biological process. Here we describe several proteomic, biophysical and structural tools that help us understand the nature and mechanism of these interactions.


Assuntos
Proteínas/metabolismo , Fenômenos Biofísicos , Calorimetria , Cromatografia de Afinidade , Microscopia Crioeletrônica , Cristalografia por Raios X , Transferência Ressonante de Energia de Fluorescência , Bicamadas Lipídicas , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Proteômica , Técnicas de Microbalança de Cristal de Quartzo , Ressonância de Plasmônio de Superfície , Técnicas do Sistema de Duplo-Híbrido
17.
Nucleic Acids Res ; 41(21): 9901-10, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23965307

RESUMO

RecA protein is a hallmark for the bacterial response to insults inflicted on DNA. It catalyzes the strand exchange step of homologous recombination and stimulates self-inactivation of the LexA transcriptional repressor. Importantly, by these activities, RecA contributes to the antibiotic resistance of bacteria. An original way to decrease the acquisition of antibiotic resistance would be to block RecA association with LexA. To engineer inhibitors of LexA-RecA complex formation, we have mapped the interaction area between LexA and active RecA-ssDNA filament (RecA*) and generated a three-dimensional model of the complex. The model revealed that one subunit of the LexA dimer wedges into a deep helical groove of RecA*, forming multiple interaction sites along seven consecutive RecA protomers. Based on the model, we predicted that LexA in its DNA-binding conformation also forms a complex with RecA* and that the operator DNA sterically precludes interaction with RecA*, which guides the induction of SOS gene expression. Moreover, the model shows that besides the catalytic C-terminal domain of LexA, its N-terminal DNA-binding domain also interacts with RecA*. Because all the model-based predictions have been confirmed experimentally, the presented model offers a validated insight into the critical step of the bacterial DNA damage response.


Assuntos
Proteínas de Bactérias/química , Recombinases Rec A/química , Proteínas Repressoras/química , Serina Endopeptidases/química , Sequência de Aminoácidos , Proteínas de Bactérias/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Regiões Operadoras Genéticas , Recombinases Rec A/metabolismo , Proteínas Repressoras/metabolismo , Resposta SOS em Genética , Serina Endopeptidases/metabolismo
18.
BMC Microbiol ; 14: 88, 2014 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-24713082

RESUMO

BACKGROUND: The SOS response including two main proteins LexA and RecA, maintains the integrity of bacterial genomes after DNA damage due to metabolic or environmental assaults. Additionally, derepression of LexA-regulated genes can result in mutations, genetic exchange and expression of virulence factors. Here we describe the first comprehensive description of the in silico LexA regulon in Clostridium difficile, an important human pathogen. RESULTS: We grouped thirty C. difficile strains from different ribotypes and toxinotypes into three clusters according to lexA gene/protein variability. We applied in silico analysis coupled to surface plasmon resonance spectroscopy (SPR) and determined 16 LexA binding sites in C. difficile. Our data indicate that strains within the cluster, as defined by LexA variability, harbour several specific LexA regulon genes. In addition to core SOS genes: lexA, recA, ruvCA and uvrBA, we identified a LexA binding site on the pathogenicity locus (PaLoc) and in the putative promoter region of several genes involved in housekeeping, sporulation and antibiotic resistance. CONCLUSIONS: Results presented here suggest that in C. difficile LexA is not merely a regulator of the DNA damage response genes but also controls the expression of dozen genes involved in various other biological functions. Our in vitro results indicate that in C. difficile inactivation of LexA repressor depends on repressor`s dissociation from the operators. We report that the repressor`s dissociation rates from operators differentiate, thus the determined LexA-DNA dissociation constants imply on the timing of SOS gene expression in C. difficile.


Assuntos
Proteínas de Bactérias/metabolismo , Clostridioides difficile/genética , Regulação Bacteriana da Expressão Gênica , Regulon , Serina Endopeptidases/metabolismo , Proteínas de Bactérias/genética , Sítios de Ligação , Simulação por Computador , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Serina Endopeptidases/genética , Ressonância de Plasmônio de Superfície
19.
Anal Biochem ; 447: 74-81, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24220292

RESUMO

Surface plasmon resonance (SPR)-based biosensors have been widely utilized for measuring interactions of a variety of molecules. Fewer examples include higher biological entities such as bacteria and viruses, and even fewer deal with plant viruses. Here, we describe the optimization of an SPR sensor chip for evaluation of the interaction of the economically relevant filamentous Potato virus Y (PVY) with monoclonal antibodies. Different virus isolates were efficiently and stably bound to a previously immobilized polyclonal antibody surface, which remained stable over subsequent injection regeneration steps. The ability of the biosensor to detect and quantify PVY particles was compared with ELISA and RT-qPCR. Stably captured virus surfaces were successfully used to explore kinetic parameters of the interaction of a panel of monoclonal antibodies with two PVY isolates representing the main viral serotypes N and O. In addition, the optimized biosensor proved to be suitable for evaluating whether two given monoclonal antibodies compete for the same epitope within the viral particle surface. The strategy proposed in this work can help to improve existing serologic diagnostic tools that target PVY and will allow investigation of the inherent serological variability of the virus and exploration for new interactions of PVY particles with other proteins.


Assuntos
Anticorpos Monoclonais/imunologia , Potyvirus/imunologia , Potyvirus/isolamento & purificação , Ressonância de Plasmônio de Superfície/métodos , Ligação Competitiva , Epitopos/imunologia , Potyvirus/química
20.
Nucleic Acids Res ; 40(4): 1879-89, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22021385

RESUMO

Synthetic scaffolds that permit spatial and temporal organization of enzymes in living cells are a promising post-translational strategy for controlling the flow of information in both metabolic and signaling pathways. Here, we describe the use of plasmid DNA as a stable, robust and configurable scaffold for arranging biosynthetic enzymes in the cytoplasm of Escherichia coli. This involved conversion of individual enzymes into custom DNA-binding proteins by genetic fusion to zinc-finger domains that specifically bind unique DNA sequences. When expressed in cells that carried a rationally designed DNA scaffold comprising corresponding zinc finger binding sites, the titers of diverse metabolic products, including resveratrol, 1,2-propanediol and mevalonate were increased as a function of the scaffold architecture. These results highlight the utility of DNA scaffolds for assembling biosynthetic enzymes into functional metabolic structures. Beyond metabolism, we anticipate that DNA scaffolds may be useful in sequestering different types of enzymes for specifying the output of biological signaling pathways or for coordinating other assembly-line processes such as protein folding, degradation and post-translational modifications.


Assuntos
Vias Biossintéticas , DNA/química , Engenharia Metabólica , Sítios de Ligação , Biocatálise , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Enzimas/genética , Enzimas/metabolismo , Escherichia coli/metabolismo , Ácido Mevalônico/metabolismo , Plasmídeos/genética , Propilenoglicol/metabolismo , Resveratrol , Estilbenos/metabolismo , Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA