Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Proteomics ; 269: 104719, 2022 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-36089190

RESUMO

Streptomycetes are multicellular gram-positive bacteria that produce many bioactive compounds, including antibiotics, antitumorals and immunosuppressors. The Streptomyces phosphoproteome remains largely uncharted even though protein phosphorylation at Ser/Thr/Tyr is known to modulate morphological differentiation and specialized metabolic processes. We here expand the S. coelicolor phosphoproteome by optimised immobilized zirconium (IV) affinity chromatography and mass spectrometry to identify phosphoproteins at the vegetative and sporulating stages. We mapped 361 phosphorylation sites (41% pSer, 56.2% pThr, 2.8% pTyr) and discovered four novel Thr phosphorylation motifs ("Kxxxx(pT)xxxxK", "DxE(pT)", "D(pT)" and "Exxxxx(pT)") in 351 phosphopeptides derived from 187 phosphoproteins. We identified 154 novel phosphoproteins, thereby almost doubling the number of experimentally verified Streptomyces phosphoproteins. Novel phosphoproteins included cell division proteins (FtsK, CrgA) and specialized metabolism regulators (ArgR, AfsR, CutR and HrcA) that were differentially phosphorylated in the vegetative and in the antibiotic producing sporulating stages. Phosphoproteins involved in primary metabolism included 27 novel ribosomal proteins that were phosphorylated during the vegetative stage. Phosphorylation of these proteins likely participate in the intricate and incompletely understood regulation of Streptomyces development and secondary metabolism. We conclude that Zr(IV)-IMAC is an efficient and sensitive method to study protein phosphorylation and regulation in bacteria and enhance our understanding of bacterial signalling. SIGNIFICANCE: Two thirds of the secondary metabolites used in clinic, especially antibiotics, were discovered in Streptomyces strains. Antibiotic resistance became one of the major challenges in clinic, and new antibiotics are urgently required in clinic. Next-generation sequencing analyses revealed that streptomycetes harbour many cryptic secondary metabolite pathways, i.e. pathways not expressed in the laboratory. Secondary metabolism is tightly connected with hypha differentiation and sporulation, and understanding Streptomyces differentiation is one of the main challenges in industrial microbiology, in order to activate the expression of cryptic pathways in the laboratory. Protein phosphorylation at Ser/Thr/Tyr modulates development and secondary metabolism, but the Streptomyces phosphoproteome is still largely uncharted. Previous S. coelicolor phosphoproteomic studies used TiO2 affinity enrichment and LC-MS/MS identifying a total of 184 Streptomyces phosphoproteins. Here, we used by first time zirconium (IV) affinity chromatography and mass spectrometry, identifying 186 S. coelicolor phosphoproteins. Most of these phosphoproteins (154) were not identified in previous phosphoproteomic studies using TiO2 affinity enrichment. Thereby we almost doubling the number of experimentally verified Streptomyces phosphoproteins. Zr(IV)-IMAC affinity chromatography also worked in E. coli, allowing the identification of phosphoproteins that were not identified by TiO2 affinity chromatography. We conclude that Zr(IV)-IMAC is an efficient and sensitive method for studies of protein phosphorylation and regulation in bacteria to enhance our understanding of bacterial signalling networks. Moreover, the new Streptomyces phosphoproteins identified will contribute to design further works to understand and modulate Streptomyces secondary metabolism activation.


Assuntos
Streptomyces coelicolor , Antibacterianos , Cromatografia de Afinidade , Cromatografia Líquida , Escherichia coli/metabolismo , Proteínas de Escherichia coli , Proteínas de Membrana , Fosfopeptídeos/análise , Fosfoproteínas/análise , Fosforilação , Proteoma/metabolismo , Proteômica/métodos , Proteínas Ribossômicas/metabolismo , Streptomyces coelicolor/química , Streptomyces coelicolor/metabolismo , Espectrometria de Massas em Tandem/métodos , Titânio , Zircônio/química , Zircônio/metabolismo
2.
Nature ; 590(7846): 463-467, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33536618

RESUMO

Actinobacteria produce numerous antibiotics and other specialized metabolites that have important applications in medicine and agriculture1. Diffusible hormones frequently control the production of such metabolites by binding TetR family transcriptional repressors (TFTRs), but the molecular basis for this remains unclear2. The production of methylenomycin antibiotics in Streptomyces coelicolor A3(2) is initiated by the binding of 2-alkyl-4-hydroxymethylfuran-3-carboxylic acid (AHFCA) hormones to the TFTR MmfR3. Here we report the X-ray crystal structure of an MmfR-AHFCA complex, establishing the structural basis for hormone recognition. We also elucidate the mechanism for DNA release upon hormone binding through the single-particle cryo-electron microscopy structure of an MmfR-operator complex. DNA binding and release assays with MmfR mutants and synthetic AHFCA analogues define the role of individual amino acid residues and hormone functional groups in ligand recognition and DNA release. These findings will facilitate the exploitation of actinobacterial hormones and their associated TFTRs in synthetic biology and in the discovery of new antibiotics.


Assuntos
Antibacterianos/biossíntese , Furanos/metabolismo , Streptomyces coelicolor/metabolismo , Apoproteínas/química , Apoproteínas/metabolismo , Apoproteínas/ultraestrutura , Proteínas de Bactérias/química , Proteínas de Bactérias/classificação , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/ultraestrutura , Microscopia Crioeletrônica , Cristalografia por Raios X , DNA/química , DNA/genética , DNA/metabolismo , DNA/ultraestrutura , Furanos/química , Hormônios/química , Hormônios/classificação , Hormônios/metabolismo , Ligantes , Modelos Moleculares , Peptídeos/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/classificação , Proteínas Repressoras/metabolismo , Proteínas Repressoras/ultraestrutura , Transdução de Sinais , Streptomyces coelicolor/química , Streptomyces coelicolor/genética , Relação Estrutura-Atividade
3.
Chemistry ; 25(14): 3675-3684, 2019 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-30600851

RESUMO

Nitric oxide (NO) can function as both a cytotoxin and a signalling molecule. In both cases, reaction with iron-sulfur (Fe-S) cluster proteins plays an important role because Fe-S clusters are reactive towards NO and so are a primary site of general NO-induced damage (toxicity). This sensitivity to nitrosylation is harnessed in the growing group of regulatory proteins that function in sensing of NO via an Fe-S cluster. Although information about the products of cluster nitrosylation is now emerging, detection and identification of intermediates remains a major challenge, due to their transient nature and the difficulty in distinguishing spectroscopically similar iron-NO species. Here we report studies of the NO-sensing Fe-S cluster regulators NsrR and WhiD using non-denaturing mass spectrometry, in which non-covalent interactions between the protein and Fe/S/NO species are preserved. The data provide remarkable insight into the nitrosylation reactions, permitting identification, for the first time, of protein-bound mono-, di- and tetranitrosyl [4Fe-4S] cluster complexes ([4Fe-4S](NO), [4Fe-4S])(NO)2 and [4Fe-4S](NO)4 ) as intermediates along pathways to formation of product Roussin's red ester (RRE) and Roussin's black salt (RBS)-like species. The data allow the nitrosylation mechanisms of NsrR and WhiD to be elucidated and clearly distinguished.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Mycobacterium tuberculosis/metabolismo , Óxido Nítrico/metabolismo , Streptomyces coelicolor/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Bactérias/química , Proteínas Ferro-Enxofre/química , Modelos Moleculares , Mycobacterium tuberculosis/química , Conformação Proteica , Espectrometria de Massas por Ionização por Electrospray , Streptomyces coelicolor/química , Fatores de Transcrição/química
5.
Sci Rep ; 8(1): 16524, 2018 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-30410115

RESUMO

Coiled-coil domains of intermediate filaments (IF) and prokaryotic IF-like proteins enable oligomerisation and filamentation, and no additional function is ascribed to these coiled-coil domains. However, an IF-like protein from Streptomyces reticuli was reported to display cellulose affinity. We demonstrate that cellulose affinity is an intrinsic property of the IF-like proteins FilP and Scy and the coiled-coil protein DivIVA from the genus Streptomyces. Furthermore, IF-like proteins and DivIVA from other prokaryotic species and metazoan IF display cellulose affinity despite having little sequence homology. Cellulose affinity-based purification is utilised to isolate native FilP protein from the whole cell lysate of S. coelicolor. Moreover, cellulose affinity allowed for the isolation of IF and IF-like protein from the whole cell lysate of C. crescentus and a mouse macrophage cell line. The binding to cellulose is mediated by certain combinations of coiled-coil domains, as demornstrated for FilP and lamin. Fusions of target proteins to cellulose-binding coiled-coil domains allowed for cellulose-based protein purification. The data presented show that cellulose affinity is a novel function of certain coiled-coil domains of IF and IF-like proteins from evolutionary diverse species.


Assuntos
Bactérias/metabolismo , Celulose/metabolismo , Filamentos Intermediários/química , Filamentos Intermediários/metabolismo , Macrófagos/metabolismo , Animais , Bactérias/química , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Escherichia coli/química , Escherichia coli/metabolismo , Macrófagos/citologia , Espectrometria de Massas , Camundongos , Ligação Proteica , Domínios Proteicos , Homologia de Sequência , Streptomyces coelicolor/química , Streptomyces coelicolor/metabolismo
6.
Arch Biochem Biophys ; 639: 26-37, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29288053

RESUMO

The phosphotransferase system (PTS) controls the preferential use of sugars in bacteria and it is also involved in other processes, such as chemotaxis. It is formed by a protein cascade in which the first two proteins are general (namely, EI and HPr) and the others are sugar-specific permeases. The Rsd protein binds specifically to the RNA polymerase (RNAP) σ70 factor. We first characterized the conformational stability of Escherichia coli Rsd. And second, we delineated the binding regions of Streptomyces coelicolor, HPrsc, and E. coli Rsd, by using fragments derived from each protein. To that end, we used several biophysical probes, namely, fluorescence, CD, NMR, ITC and BLI. Rsd had a free energy of unfolding of 15 kcal mol-1 at 25 °C, and a thermal denaturation midpoint of 103 °C at pH 6.5. The affinity between Rsd and HPrsc was 2 µM. Interestingly enough, the isolated helical-peptides, comprising the third (RsdH3) and fourth (RsdH4) Rsd helices, also interacted with HPrsc in a specific manner, and with affinities similar to that of the whole Rsd. Moreover, the isolated peptide of HPrsc, HPr9-30, comprising the active site, His15, also was bound to intact Rsd with similar affinity. Therefore, binding between Rsd and HPrsc was modulated by the two helices H3 and H4 of Rsd, and the regions around the active site of HPrsc. This implies that specific fragments of Rsd and HPrsc can be used to interfere with other protein-protein interactions (PPIs) of each other protein.


Assuntos
Proteínas de Bactérias/química , Proteínas de Escherichia coli/química , Escherichia coli/química , Peptídeos/química , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/química , Proteínas Repressoras/química , Streptomyces coelicolor/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/genética , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/metabolismo , Estrutura Secundária de Proteína , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Streptomyces coelicolor/genética , Streptomyces coelicolor/metabolismo
7.
PLoS One ; 11(12): e0167763, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27936128

RESUMO

Many species of Gram-positive bacteria use sortase transpeptidases to covalently affix proteins to their cell wall or to assemble pili. Sortase-displayed proteins perform critical and diverse functions for cell survival, including cell adhesion, nutrient acquisition, and morphological development, among others. Based on their amino acid sequences, there are at least six types of sortases (class A to F enzymes); however, class E enzymes have not been extensively studied. Class E sortases are used by soil and freshwater-dwelling Actinobacteria to display proteins that contain a non-canonical LAXTG sorting signal, which differs from 90% of known sorting signals by substitution of alanine for proline. Here we report the first crystal structure of a class E sortase, the 1.93 Å resolution structure of the SrtE1 enzyme from Streptomyces coelicolor. The active site is bound to a tripeptide, providing insight into the mechanism of substrate binding. SrtE1 possesses ß3/ß4 and ß6/ß7 active site loops that contact the LAXTG substrate and are structurally distinct from other classes. We propose that SrtE1 and other class E sortases employ a conserved tyrosine residue within their ß3/ß4 loop to recognize the amide nitrogen of alanine at position P3 of the sorting signal through a hydrogen bond, as seen here. Incapability of hydrogen-bonding with canonical proline-containing sorting signals likely contributes to class E substrate specificity. Furthermore, we demonstrate that surface anchoring of proteins involved in aerial hyphae formation requires an N-terminal segment in SrtE1 that is presumably positioned within the cytoplasm. Combined, our results reveal unique features within class E enzymes that enable them to recognize distinct sorting signals, and could facilitate the development of substrate-based inhibitors of this important enzyme family.


Assuntos
Aminoaciltransferases/química , Proteínas de Bactérias/química , Cisteína Endopeptidases/química , Streptomyces coelicolor/enzimologia , Sequência de Aminoácidos , Aminoaciltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Cisteína Endopeptidases/metabolismo , Modelos Moleculares , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Conformação Proteica , Sinais Direcionadores de Proteínas , Streptomyces coelicolor/química , Streptomyces coelicolor/metabolismo , Especificidade por Substrato
8.
J Biol Chem ; 291(44): 23175-23187, 2016 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-27634042

RESUMO

ADP-ribosylation is a post-translational modification that can alter the physical and chemical properties of target proteins and that controls many important cellular processes. Macrodomains are evolutionarily conserved structural domains that bind ADP-ribose derivatives and are found in proteins with diverse cellular functions. Some proteins from the macrodomain family can hydrolyze ADP-ribosylated substrates and therefore reverse this post-translational modification. Bacteria and Streptomyces, in particular, are known to utilize protein ADP-ribosylation, yet very little is known about their enzymes that synthesize and remove this modification. We have determined the crystal structure and characterized, both biochemically and functionally, the macrodomain protein SCO6735 from Streptomyces coelicolor This protein is a member of an uncharacterized subfamily of macrodomain proteins. Its crystal structure revealed a highly conserved macrodomain fold. We showed that SCO6735 possesses the ability to hydrolyze PARP-dependent protein ADP-ribosylation. Furthermore, we showed that expression of this protein is induced upon DNA damage and that deletion of this protein in S. coelicolor increases antibiotic production. Our results provide the first insights into the molecular basis of its action and impact on Streptomyces metabolism.


Assuntos
Antibacterianos/biossíntese , Proteínas de Bactérias/metabolismo , Streptomyces coelicolor/metabolismo , Adenosina Difosfato Ribose/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Dano ao DNA , Processamento de Proteína Pós-Traducional , Streptomyces coelicolor/química , Streptomyces coelicolor/genética
9.
J Microbiol ; 54(9): 626-631, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27572512

RESUMO

The sco0765 gene was annotated as a glycosyl hydrolase family 5 endoglucanase from the genomic sequence of Streptomyces coelicolor A3(2) and consisted of 2,241 bp encoding a polypeptide of 747 amino acids (molecular weight of 80.5 kDa) with a 29-amino acid signal peptide for secretion. The SCO0765 recombinant protein was heterogeneously over-expressed in Streptomyces lividans TK24 under the control of a strong ermE* promoter. The purified SCO0765 protein showed the expected molecular weight of the mature form (718 aa, 77.6 kDa) on sodium dodecyl sulfate-polyacryl amide gel electrophoresis. SCO0765 showed high activity toward ß-glucan and carboxymethyl cellulose (CMC) and negligible activity to Avicel, xylan, and xyloglucan. The SCO0765 cellulase had a maximum activity at pH 6.0 and 40°C toward CMC and at pH 9.0 and 50-60°C toward ß-glucan. Thin layer chromatography of the hydrolyzed products of CMC and ß-glucan by SCO0765 gave cellotriose as the major product and cellotetraose, cellopentaose, and longer oligosaccharides as the minor products. These results clearly demonstrate that SCO0765 is an endo-ß-1,4-cellulase, hydrolyzing the ß-1,4 glycosidic bond of cellulose into cellotriose.


Assuntos
Celulase/química , Celulase/metabolismo , Celulose/análogos & derivados , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Streptomyces coelicolor/enzimologia , Tetroses/metabolismo , Celulase/genética , Celulose/metabolismo , Estabilidade Enzimática , Proteínas Fúngicas/genética , Concentração de Íons de Hidrogênio , Cinética , Peso Molecular , Streptomyces coelicolor/química , Streptomyces coelicolor/genética , Streptomyces coelicolor/metabolismo , Especificidade por Substrato , beta-Glucanas/metabolismo
10.
J Biol Chem ; 290(11): 7280-90, 2015 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-25631049

RESUMO

Lipoate-protein ligases are used to scavenge lipoic acid from the environment and attach the coenzyme to its cognate proteins, which are generally the E2 components of the 2-oxoacid dehydrogenases. The enzymes use ATP to activate lipoate to its adenylate, lipoyl-AMP, which remains tightly bound in the active site. This mixed anhydride is attacked by the ϵ-amino group of a specific lysine present on a highly conserved acceptor protein domain, resulting in the amide-linked coenzyme. The Streptomyces coelicolor genome encodes only a single putative lipoate ligase. However, this protein had only low sequence identity (<25%) to the lipoate ligases of demonstrated activity and appears to be a circularly permuted version of the known lipoate ligase proteins in that the canonical C-terminal domain seems to have been transposed to the N terminus. We tested the activity of this protein both by in vivo complementation of an Escherichia coli ligase-deficient strain and by in vitro assays. Moreover, when the domains were rearranged into a protein that mimicked the arrangement found in the canonical lipoate ligases, the enzyme retained complementation activity. Finally, when the two domains were separated into two proteins, both domain-containing proteins were required for complementation and catalysis of the overall ligase reaction in vitro. However, only the large domain-containing protein was required for transfer of lipoate from the lipoyl-AMP intermediate to the acceptor proteins, whereas both domain-containing proteins were required to form lipoyl-AMP.


Assuntos
Escherichia coli/enzimologia , Peptídeo Sintases/química , Peptídeo Sintases/genética , Streptomyces coelicolor/enzimologia , Ácido Tióctico/metabolismo , Monofosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/metabolismo , Evolução Molecular , Modelos Moleculares , Dados de Sequência Molecular , Peptídeo Sintases/metabolismo , Filogenia , Estrutura Terciária de Proteína , Alinhamento de Sequência , Streptomyces coelicolor/química , Streptomyces coelicolor/genética , Streptomyces coelicolor/metabolismo
11.
J Mol Graph Model ; 56: 10-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25541526

RESUMO

All-trans-retinoic acid (ATRA), the biologically most active metabolite of vitamin A, plays a major role in the regulation of cellular differentiation and proliferation, and it is also an important pharmacological agent particularly used in the treatment of cancer, skin, neurodegenerative and autoimmune diseases. However, ATRA is very easy to be metabolized into 4-hydroxyl-RA in vivo by CYP26A1, an inducible cytochrome P450 enzyme, eventually into more polar metabolites. Therefore, it is vital to develop specific retinoic acid metabolism blocking agents (RAMBAs) to inhibit the metabolic enzyme CYP26A1 in the treatment of relevant diseases aforementioned. In this study, CYP26A1 and its interactions with retinoic acid-competitive metabolism blocking agents were investigated by a combined ligand- and structure-based approach. First, since the crystal structure of CYP26A1 protein has not been determined, we constructed the 3D structure of CYP26A1 using homology modeling. In order to achieve a deeper insight into the mode of action of RAMBAs in the active site, the molecular superimposition model and the common feature pharmacophore model were constructed, and molecular docking was performed. The molecular superimposition model is composed of three features: the main chain groups, side chain groups, and azole groups. The common feature pharmacophore model consists of five chemical features: four hydrophobic groups and one hydrogen acceptor (HHHHA). The results of molecular docking show that the characteristic groups of RAMBAs were mapped into three different active pockets, respectively. A structure-activity relationship (SAR) was obtained by a combination of the molecular superimposition and docking results with the pharmacophore model. This study gives more insight into the interaction model inside the CYP26A1 active site and provides guidance for the design of more potent and possibly more selective RAMBAs.


Assuntos
Azóis/química , Inibidores das Enzimas do Citocromo P-450/química , Sistema Enzimático do Citocromo P-450/química , Tretinoína/química , Sequência de Aminoácidos , Azóis/metabolismo , Bacillus megaterium/química , Bacillus megaterium/enzimologia , Inibidores das Enzimas do Citocromo P-450/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Desenho de Fármacos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Cinética , Ligantes , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/enzimologia , Ligação Proteica , Ácido Retinoico 4 Hidroxilase , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Eletricidade Estática , Streptomyces coelicolor/química , Streptomyces coelicolor/enzimologia , Homologia Estrutural de Proteína , Relação Estrutura-Atividade , Synechocystis/química , Synechocystis/enzimologia , Termodinâmica , Tretinoína/metabolismo
12.
Chembiochem ; 15(14): 2156-61, 2014 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-25155017

RESUMO

Wild-type Streptomyces coelicolor A3(2) produces aminobacteriohopanetriol as the only elongated C35 hopanoid. The hopanoid phenotype of two mutants bearing a deletion of genes from a previously identified hopanoid biosynthesis gene cluster provides clues to the formation of C35 bacteriohopanepolyols. orf14 encodes a putative nucleosidase; its deletion induces the accumulation of adenosylhopane as it cannot be converted into ribosylhopane. orf18 encodes a putative transaminase; its deletion results in the accumulation of adenosylhopane, ribosylhopane, and bacteriohopanetetrol. Ribosylhopane was postulated twenty years ago as a precursor for bacterial hopanoids but was never identified in a bacterium. Absence of the transaminase encoded by orf18 prevents the reductive amination of ribosylhopane into aminobacteriohopanetriol and induces its accumulation. Its reduction by an aldose-reductase-like enzyme produces bacteriohopanetetrol, which is normally not present in S. coelicolor.


Assuntos
Streptomyces coelicolor/metabolismo , Triterpenos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Vias Biossintéticas , Deleção de Genes , Genes Bacterianos , Família Multigênica , N-Glicosil Hidrolases/genética , N-Glicosil Hidrolases/metabolismo , Streptomyces coelicolor/química , Streptomyces coelicolor/genética , Transaminases/genética , Transaminases/metabolismo , Triterpenos/química
13.
mBio ; 4(4)2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-23963177

RESUMO

UNLABELLED: Soils host diverse microbial communities that include filamentous actinobacteria (actinomycetes). These bacteria have been a rich source of useful metabolites, including antimicrobials, antifungals, anticancer agents, siderophores, and immunosuppressants. While humans have long exploited these compounds for therapeutic purposes, the role these natural products may play in mediating interactions between actinomycetes has been difficult to ascertain. As an initial step toward understanding these chemical interactions at a systems level, we employed the emerging techniques of nanospray desorption electrospray ionization (NanoDESI) and matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) imaging mass spectrometry to gain a global chemical view of the model bacterium Streptomyces coelicolor interacting with five other actinomycetes. In each interaction, the majority of secreted compounds associated with S. coelicolor colonies were unique, suggesting an idiosyncratic response from S. coelicolor. Spectral networking revealed a family of unknown compounds produced by S. coelicolor during several interactions. These compounds constitute an extended suite of at least 12 different desferrioxamines with acyl side chains of various lengths; their production was triggered by siderophores made by neighboring strains. Taken together, these results illustrate that chemical interactions between actinomycete bacteria exhibit high complexity and specificity and can drive differential secondary metabolite production. IMPORTANCE: Actinomycetes, filamentous actinobacteria from the soil, are the deepest natural source of useful medicinal compounds, including antibiotics, antifungals, and anticancer agents. There is great interest in developing new strategies that increase the diversity of metabolites secreted by actinomycetes in the laboratory. Here we used several metabolomic approaches to examine the chemicals made by these bacteria when grown in pairwise coculture. We found that these interspecies interactions stimulated production of numerous chemical compounds that were not made when they grew alone. Among these compounds were at least 12 different versions of a molecule called desferrioxamine, a siderophore used by the bacteria to gather iron. Many other compounds of unknown identity were also observed, and the pattern of compound production varied greatly among the interaction sets. These findings suggest that chemical interactions between actinomycetes are surprisingly complex and that coculture may be a promising strategy for finding new molecules from actinomycetes.


Assuntos
Metaboloma , Interações Microbianas , Streptomyces coelicolor/química , Streptomyces coelicolor/metabolismo , Actinobacteria , Bactérias , Microbiologia do Solo , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Streptomyces coelicolor/isolamento & purificação
14.
J Microbiol Methods ; 93(2): 138-43, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23517679

RESUMO

Prodigiosin-like pigments or prodiginines (PdGs) are promising drugs owing to their reported antitumor, antibiotic, and immunosuppressive activities. These natural compounds are produced by several bacteria, including Streptomyces coelicolor and Serratia marcescens as most commonly studied models. The bright red color of these tripyrrole pigments made them excellent reporter molecules for studies aimed at understanding the molecular mechanisms that control secondary metabolite production in microorganisms. However, the natural red fluorescence of PdGs has only been rarely used as a biophysical parameter for detection and assessment of PdG biosynthesis. In this work, we used S. coelicolor in order to exemplify how intrinsic red fluorescence could be utilized for rapid, low-cost, sensitive, specific and accurate semi-quantitative analyses of PdG biosynthesis. Additionally, and contrary to the colorimetric-based approach, the fluorescence-based method allows in situ spatio-temporal visualization of PdG synthesis throughout a solid culture of S. coelicolor. As PdG production is related to cell differentiation, their red autofluorescence could be exploited, by means of confocal microscopy, as a natural marker of the entrance into a crucial developmental stage in the course of the S. coelicolor life cycle.


Assuntos
Produtos Biológicos/análise , Prodigiosina/análogos & derivados , Streptomyces coelicolor/metabolismo , Prodigiosina/análise , Sensibilidade e Especificidade , Streptomyces coelicolor/química
15.
Biochemistry ; 52(1): 4-18, 2013 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-23240653

RESUMO

Nickel-containing superoxide dismutases (NiSODs) represent a novel approach to the detoxification of superoxide in biology and thus contribute to the biodiversity of mechanisms for the removal of reactive oxygen species (ROS). While Ni ions play critical roles in anaerobic microbial redox (hydrogenases and CO dehydrogenase/acetyl coenzyme A synthase), they have never been associated with oxygen metabolism. Several SODs have been characterized from numerous sources and are classified by their catalytic metal as Cu/ZnSOD, MnSOD, or FeSOD. Whereas aqueous solutions of Cu(II), Mn(II), and Fe(II) ions are capable of catalyzing the dismutation of superoxide, solutions of Ni(II) are not. Nonetheless, NiSOD catalyzes the reaction at the diffusion-controlled limit (~10(9) M(-1) s(-1)). To do this, nature has created a Ni coordination unit with the appropriate Ni(III/II) redox potential (~0.090 V vs Ag/AgCl). This potential is achieved by a unique ligand set comprised of residues from the N-terminus of the protein: Cys2 and Cys6 thiolates, the amino terminus and imidazole side chain of His1, and a peptide N-donor from Cys2. Over the past several years, synthetic modeling efforts by several groups have provided insight into understanding the intrinsic properties of this unusual Ni coordination site. Such analogues have revealed information regarding the (i) electrochemical properties that support Ni-based redox, (ii) oxidative protection and/or stability of the coordinated CysS ligands, (iii) probable H(+) sources for H(2)O(2) formation, and (iv) nature of the Ni coordination geometry throughout catalysis. This review includes the results and implications of such biomimetic work as it pertains to the structure and function of NiSOD.


Assuntos
Materiais Biomiméticos/química , Complexos de Coordenação/química , Níquel/química , Superóxido Dismutase/química , Animais , Materiais Biomiméticos/metabolismo , Complexos de Coordenação/metabolismo , Humanos , Modelos Moleculares , Níquel/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo , Streptomyces coelicolor/química , Streptomyces coelicolor/enzimologia , Superóxido Dismutase/metabolismo
16.
Biochemistry ; 51(37): 7393-402, 2012 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-22909257

RESUMO

The phosphotransferase system (PTS) controls the use of sugars in bacteria. The PTS is ubiquitous in bacteria, but it does not occur in plants and animals; it modulates catabolite repression, intermediate metabolism, gene expression, and chemotaxis. Its uniqueness and pleiotropic function make the PTS an attractive target for new antibacterial drugs. The PTS is constituted of two general proteins, namely, enzyme I (EI) and the histidine phosphocarrier (HPr), and various sugar-specific permeases. EI has two domains: the N-terminal domain (EIN), which binds to HPr, and the C-terminal domain (EIC), which contains the dimerization interface. In this work, we determined the binding affinities of peptides derived from EIN of Streptomyces coelicolor (EIN(sc)) against HPr of the same organism (HPr(sc)), by using nuclear magnetic resonance and isothermal titration calorimetry techniques. Furthermore, we measured the affinity of EIN(sc) for (i) a peptide derived from HPr(sc), containing the active-site histidine, and (ii) other peptides identified previously by phage display and combinatorial chemistry in Escherichia coli [Mukhija, S. L., et al (1998) Eur. J. Biochem. 254, 433-438; Mukhija, S., and Erni, B. (1997) Mol. Microbiol. 25, 1159-1166]. The affinities were in the range of ~10 µM, being slightly higher for the binding of EIN(sc) with peptides derived from HPr(sc), phage display, or combinatorial chemistry (K(D) ~ 5 µM). Because the affinity of intact EIN(sc) for the whole HPr(sc) is 12 µM, we suggest that the assayed peptides might be considered as good hit compounds for inhibiting the interaction between HPr(sc) and EIN(sc).


Assuntos
Sistemas de Transporte de Aminoácidos/antagonistas & inibidores , Antibacterianos/química , Proteínas de Bactérias/antagonistas & inibidores , Inibidores Enzimáticos/química , Peptídeos/química , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Nitrogenado)/antagonistas & inibidores , Streptomyces coelicolor/enzimologia , Sistemas de Transporte de Aminoácidos/química , Sistemas de Transporte de Aminoácidos/metabolismo , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Técnicas de Química Combinatória , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Peptídeos/metabolismo , Peptídeos/farmacologia , Fosfoenolpiruvato/química , Fosfoenolpiruvato/metabolismo , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/química , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Fosfotransferases (Aceptor do Grupo Nitrogenado)/química , Fosfotransferases (Aceptor do Grupo Nitrogenado)/metabolismo , Estrutura Terciária de Proteína , Streptomyces coelicolor/química
17.
Electron. j. biotechnol ; 15(1): 8-8, Jan. 2012. ilus, tab
Artigo em Inglês | LILACS | ID: lil-640534

RESUMO

Background: The growing problem of environmental pollution caused by synthetic plastics has led to the search for alternative materials such as biodegradable plastics. Of the biopolymers presently under development, starch/natural rubber is one promising alternative. Several species of bacteria and fungi are capable of degrading natural rubber and many can degrade starch. Results: Streptomyces coelicolor CH13 was isolated from soil according to its ability to produce translucent halos on a mineral salts medium, MSM, supplemented with natural rubber and to degrade starch. Scanning electron microscope studies showed that it colonized the surfaces of strips of a new starch/natural rubber biopolymer and rubber gloves and caused degradation by forming holes, and surface degradation. Starch was completely removed and polyisoprene chains were broken down to produce aldehyde and/or carbonyl groups. After 6 weeks of cultivation with strips of the polymers in MSM, S. coelicolor CH13 reduced the weight of the starch/NR biopolymer by 92 percent and that of the rubber gloves by 14.3 percent. Conclusions: This study indicated that this bacterium causes the biodegradation of the new biopolymer and natural rubber and confirms that this new biopolymer can be degraded in the environment and would be suitable as a ‘green plastic’ derived from natural sources.


Assuntos
Amido/metabolismo , Biopolímeros/metabolismo , Borracha/metabolismo , Streptomyces coelicolor/metabolismo , Streptomyces coelicolor/química , Biodegradação Ambiental , Biopolímeros/química , Borracha/química
18.
Mol Microbiol ; 77(4): 943-57, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20572939

RESUMO

Lipoproteins are a distinct class of bacterial membrane proteins that are translocated across the cytoplasmic membrane primarily by the Sec general secretory pathway and then lipidated on a conserved cysteine by the enzyme lipoprotein diacylglycerol transferase (Lgt). The signal peptide is cleaved by lipoprotein signal peptidase (Lsp) to leave the lipid-modified cysteine at the N-terminus of the mature lipoprotein. In all Gram-positive bacteria tested to date this pathway is non-essential and the lipid attaches the protein to the outer leaflet of the cytoplasmic membrane. Here we identify lipoproteins in the model Gram-positive bacterium Streptomyces coelicolor using bioinformatics coupled with proteomic and downstream analysis. We report that Streptomyces species translocate large numbers of lipoproteins out via the Tat (twin arginine translocase) pathway and we present evidence that lipoprotein biogenesis might be an essential pathway in S. coelicolor. This is the first analysis of lipoproteins and lipoprotein biogenesis in Streptomyces and provides the first evidence that lipoprotein biogenesis could be essential in a Gram-positive bacterium. This report also provides the first experimental evidence that Tat plays a major role in the translocation of lipoproteins in a specific bacterium.


Assuntos
Lipoproteínas/metabolismo , Streptomyces coelicolor/metabolismo , Biologia Computacional , Lipoproteínas/genética , Transporte Proteico , Proteoma/análise , Streptomyces coelicolor/química , Streptomyces coelicolor/genética
19.
Biochemistry ; 48(51): 12252-64, 2009 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-19954209

RESUMO

WhiD, a member of the WhiB-like (Wbl) family of iron-sulfur proteins found exclusively within the actinomycetes, is required for the late stages of sporulation in Streptomyces coelicolor. Like all other Wbl proteins, WhiD has not so far been purified in a soluble form that contains a significant amount of cluster, and characterization has relied on cluster-reconstituted protein. Thus, a major goal in Wbl research is to obtain and characterize native protein containing iron-sulfur clusters. Here we report the analysis of S. coelicolor WhiD purified anaerobically from Escherichia coli as a soluble protein containing a single [4Fe-4S](2+) cluster ligated by four cysteines. Upon exposure to oxygen, spectral features associated with the [4Fe-4S] cluster were lost in a slow reaction that unusually yielded apo-WhiD directly without significant concentrations of cluster intermediates. This process was found to be highly pH dependent with an optimal stability observed between pH 7.0 and pH 8.0. Low molecular weight thiols, including a mycothiol analogue and thioredoxin, exerted a small but significant protective effect against WhiD cluster loss, an activity that could be of physiological importance. [4Fe-4S](2+) WhiD was found to react much more rapidly with superoxide than with either oxygen or hydrogen peroxide, which may also be of physiological significance. Loss of the [4Fe-4S] cluster to form apoprotein destabilized the protein fold significantly but did not lead to complete unfolding. Finally, apo-WhiD exhibited negligible activity in an insulin-based disulfide reductase assay, demonstrating that it does not function as a general protein disulfide reductase.


Assuntos
Proteínas Ferro-Enxofre/química , Streptomyces coelicolor/química , Fatores de Transcrição/química , Sequência de Aminoácidos , Proteínas Ferro-Enxofre/genética , Proteínas Ferro-Enxofre/isolamento & purificação , Proteínas Ferro-Enxofre/metabolismo , Dados de Sequência Molecular , Estabilidade Proteica , Solubilidade , Streptomyces coelicolor/genética , Streptomyces coelicolor/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/isolamento & purificação , Fatores de Transcrição/metabolismo
20.
J Microbiol ; 47(5): 549-56, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19851727

RESUMO

Mono-ADP-ribosylation is the enzymatic transfer of ADP-ribose from NAD(+) to acceptor proteins catalyzed by ADP-ribosyltransferases. Using m-aminophenylboronate affinity chromatography, 2D-gel electrophoresis, in-gel digestion and MALDI-TOF analysis we have identified eight in vitro ADP-ribosylated proteins in Streptomyces coelicolor, which can be classified into three categories: (i) secreted proteins; (ii) metabolic enzymes using NAD(+)/NADH or NADP(+)/NADPH as coenzymes; and (iii) other proteins. The secreted proteins could be classified into two functional categories: SCO2008 and SC05477 encode members of the family of periplasmic extracellular solute-binding proteins, and SCO6108 and SC01968 are secreted hydrolases. Dehydrogenases are encoded by SC04824 and SC04771. The other targets are GlnA (glutamine synthetase I., SC02198) and SpaA (starvation-sensing protein encoded by SC07629). SCO2008 protein and GlnA had been identified as ADP-ribosylated proteins in previous studies. With these results we provided experimental support for a previous suggestion that ADP-ribosylation may regulate membrane transport and localization of periplasmic proteins. Since ADP-ribosylation results in inactivation of the target protein, ADP-ribosylation of dehydrogenases might modulate crucial primary metabolic pathways in Streptomyces. Several of the proteins identified here could provide a strong connection between protein ADP-ribosylation and the regulation of morphological differentiation in S. coelicolor.


Assuntos
Adenosina Difosfato Ribose/análise , Proteínas de Bactérias/química , Proteínas de Bactérias/isolamento & purificação , Streptomyces coelicolor/química , Sequência de Aminoácidos , Cromatografia de Afinidade , Eletroforese em Gel Bidimensional , Dados de Sequência Molecular , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA