Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.949
Filtrar
1.
ACS Chem Biol ; 19(9): 1896-1903, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39248435

RESUMEN

The class III lanthipeptide synthetase (LanKC) installs unusual amino acids, such as lanthionine and labionin, in lanthipeptides. Through genome mining, we discovered a new class III lanthipeptide synthetase coding gene (nptKC) and precursor peptide coding genes (nptA1, nptA2, and nptA3) in the genome of the actinobacterium Nocardiopsis alba. Coexpression experiments of the biosynthetic genes in Escherichia coli resulted in the production of new lanthipeptides named nocardiopeptins A1-A3. Analysis of two-dimensional NMR spectra after enzymatic degradation and partial basic hydrolysis of nocardiopeptin A2 revealed that labionin was located in lanthionine with opposite orientations, forming a nesting structure in nocardiopeptin A2. To the best of our knowledge, this bridging pattern in the lanthipeptides was unprecedented, indicating a novel reaction characteristic of the class III lanthipeptide synthetase NptKC.


Asunto(s)
Alanina , Alanina/análogos & derivados , Alanina/metabolismo , Alanina/química , Escherichia coli/genética , Escherichia coli/metabolismo , Péptido Sintasas/metabolismo , Péptido Sintasas/genética , Sulfuros/química , Sulfuros/metabolismo , Péptidos/química , Péptidos/metabolismo , Actinobacteria/metabolismo , Actinobacteria/genética , Actinobacteria/química
2.
Mar Drugs ; 22(8)2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39195464

RESUMEN

Nonribosomal peptides (NRPs) are biosynthesized by nonribosomal peptide synthetases (NRPSs) and are widely distributed in both terrestrial and marine organisms. Many NRPs and their analogs are biologically active and serve as therapeutic agents. The adenylation (A) domain is a key catalytic domain that primarily controls the sequence of a product during the assembling of NRPs and thus plays a predominant role in the structural diversity of NRPs. Engineering of the A domain to alter substrate specificity is a potential strategy for obtaining novel NRPs for pharmaceutical studies. On the basis of introducing the catalytic mechanism and multiple functions of the A domains, this article systematically describes several representative NRPS engineering strategies targeting the A domain, including mutagenesis of substrate-specificity codes, substitution of condensation-adenylation bidomains, the entire A domain or its subdomains, domain insertion, and whole-module rearrangements.


Asunto(s)
Péptido Sintasas , Ingeniería de Proteínas , Péptido Sintasas/genética , Péptido Sintasas/metabolismo , Péptido Sintasas/química , Especificidad por Sustrato , Organismos Acuáticos , Dominio Catalítico , Animales
3.
Methods Enzymol ; 702: 89-119, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39155122

RESUMEN

Non-ribosomal peptide synthesis produces a wide range of bioactive peptide natural products and is reliant on a modular architecture based on repeating catalytic domains able to generate diverse peptide sequences. In this chapter we detail an in vitro biochemical assay to explore the substrate specificity of condensation domains, which are responsible for peptide elongation, from the biosynthetic machinery that produces from the siderophore fuscachelin. This assay removes the requirement to utilise the specificity of adjacent adenylation domains and allows the acceptance of a wide range of synthetic substrates to be explored.


Asunto(s)
Sideróforos , Especificidad por Sustrato , Sideróforos/química , Sideróforos/biosíntesis , Péptido Sintasas/metabolismo , Péptido Sintasas/química , Péptido Sintasas/genética , Péptidos/química , Péptidos/metabolismo , Biosíntesis de Péptidos Independientes de Ácidos Nucleicos , Dominio Catalítico
4.
Nat Commun ; 15(1): 5787, 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39025839

RESUMEN

Coevolutionary antagonism generates relentless selection that can favour genetic exchange, including transfer of antibiotic synthesis and resistance genes among bacteria, and sexual recombination of disease resistance alleles in eukaryotes. We report an unusual link between biological conflict and DNA transfer in bdelloid rotifers, microscopic animals whose genomes show elevated levels of horizontal gene transfer from non-metazoan taxa. When rotifers were challenged with a fungal pathogen, horizontally acquired genes were over twice as likely to be upregulated as other genes - a stronger enrichment than observed for abiotic stressors. Among hundreds of upregulated genes, the most markedly overrepresented were clusters resembling bacterial polyketide and nonribosomal peptide synthetases that produce antibiotics. Upregulation of these clusters in a pathogen-resistant rotifer species was nearly ten times stronger than in a susceptible species. By acquiring, domesticating, and expressing non-metazoan biosynthetic pathways, bdelloids may have evolved to resist natural enemies using antimicrobial mechanisms absent from other animals.


Asunto(s)
Transferencia de Gen Horizontal , Rotíferos , Animales , Rotíferos/genética , Rotíferos/metabolismo , Vías Biosintéticas/genética , Péptido Sintasas/genética , Péptido Sintasas/metabolismo , Policétidos/metabolismo , Filogenia , Familia de Multigenes
5.
Microb Cell Fact ; 23(1): 207, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39044227

RESUMEN

The engineering of non ribosomal peptide synthetases (NRPS) for new substrate specificity is a potent strategy to incorporate non-canonical amino acids into peptide sequences, thereby creating peptide diversity and broadening applications. The non-ribosomal peptide pyoverdine is the primary siderophore produced by Pseudomonas aeruginosa and holds biomedical promise in diagnosis, bio-imaging and antibiotic vectorization. We engineered the adenylation domain of PvdD, the terminal NRPS in pyoverdine biosynthesis, to accept a functionalized amino acid. Guided by molecular modeling, we rationally designed mutants of P. aeruginosa with mutations at two positions in the active site. A single amino acid change results in the successful incorporation of an azido-L-homoalanine leading to the synthesis of a new pyoverdine analog, functionalized with an azide function. We further demonstrated that copper free click chemistry is efficient on the functionalized pyoverdine and that the conjugated siderophore retains the iron chelation properties and its capacity to be recognized and transported by P. aeruginosa. The production of clickable pyoverdine holds substantial biotechnological significance, paving the way for numerous downstream applications.


Asunto(s)
Química Clic , Oligopéptidos , Péptido Sintasas , Ingeniería de Proteínas , Pseudomonas aeruginosa , Oligopéptidos/biosíntesis , Oligopéptidos/metabolismo , Pseudomonas aeruginosa/enzimología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Péptido Sintasas/metabolismo , Péptido Sintasas/genética , Ingeniería de Proteínas/métodos , Sideróforos/biosíntesis , Sideróforos/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Dominio Catalítico , Especificidad por Sustrato
6.
Enzyme Microb Technol ; 180: 110481, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39047348

RESUMEN

The thermal instability of γ-glutamylmethylamide synthetase (GMAS) from Methylovorus mays has imposed limitations on its industrial applications, affecting both stability and activity at reaction temperatures. In this study, disulfide bridges were introduced through a combination of directed evolution and rational design to enhance GMAS stability. Among the variants that we generated, M12 exhibited a 1.46-fold improvement in relative enzyme activity and a 6.23-fold increase in half-life at 40℃ compared to the wild-type GMAS. Employing variant M12 under optimal conditions, we achieved the production of 645.7 mM (112.49 g/L) L-theanine with a productivity of 29.3 mM/h, from 800 mM substrate in an ATP regeneration system. Our strategy significantly enhances the biosynthesis efficiency of L-theanine by preserving the structural stability of the enzyme during the catalysis process.


Asunto(s)
Estabilidad de Enzimas , Glutamatos , Péptido Sintasas , Glutamatos/metabolismo , Glutamatos/biosíntesis , Péptido Sintasas/metabolismo , Péptido Sintasas/genética , Péptido Sintasas/química , Evolución Molecular Dirigida , Cinética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Temperatura , Ligasas de Carbono-Nitrógeno
7.
Curr Microbiol ; 81(9): 275, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39020143

RESUMEN

In this study, the toxigenic characteristics of 14 strains of Microcystis were analyzed, and single nucleotide polymorphism (SNP) and insertion/deletion (InDel) loci in microcystin synthetase (mcy) gene clusters were screened. Based on SNP and InDel loci associated with the toxigenic characteristics, primers and TaqMan or Cycling fluorescent probes were designed to develop duplex real-time fluorescent quantitative PCR (FQ-PCR) assays. After evaluating specificity and sensitivity, these assays were applied to detect the toxigenic Microcystis genotypes in a shrimp pond where Microcystis blooms occurred. The results showed a total of 2155 SNP loci and 66 InDel loci were obtained, of which 12 SNP loci and 5 InDel loci were associated with the toxigenic characteristics. Three duplex real-time FQ-PCR assays were developed, each of which could quantify two genotypes of toxigenic Microcystis. These FQ-PCR assays were highly specific, and two Cycling assays were more sensitive than TaqMan assay. In the shrimp pond, six genotypes of toxigenic Microcystis were detected using the developed FQ-PCR assays, indicating that above genotyping assays have the potential for quantitative analysis of the toxigenic Microcystis genotypes in natural water.


Asunto(s)
Genotipo , Microcystis , Familia de Multigenes , Polimorfismo de Nucleótido Simple , Reacción en Cadena en Tiempo Real de la Polimerasa , Microcystis/genética , Microcystis/clasificación , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Microcistinas/genética , Mutación INDEL , Proteínas Bacterianas/genética , Sensibilidad y Especificidad , Estanques/microbiología , Péptido Sintasas/genética
8.
J Nat Prod ; 87(7): 1704-1713, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-38990199

RESUMEN

Fungal secondary metabolite (SM) biosynthetic gene clusters (BGCs) containing dimethylallyltryptophan synthases (DMATSs) produce structurally diverse prenylated indole alkaloids with wide-ranging activities that have vast potential as human therapeutics. To discover new natural products produced by DMATSs, we mined the Department of Energy Joint Genome Institute's MycoCosm database for DMATS-containing BGCs. We found a DMATS BGC in Aspergillus homomorphus CBS 101889, which also contains a nonribosomal peptide synthetase (NRPS). This BGC appeared to have a previously unreported combination of genes, which suggested the cluster might make novel SMs. We refactored this BGC with highly inducible promoters into the model fungus Aspergillus nidulans. The expression of this refactored BGC in A. nidulans resulted in the production of eight tryptophan-containing diketopiperazines, six of which are new to science. We have named them homomorphins A-F (2, 4-8). Perhaps even more intriguingly, to our knowledge, this is the first discovery of C4-prenylated tryptophan-containing diketopiperazines and their derivatives. In addition, the NRPS from this BGC is the first described that has the ability to promiscuously combine tryptophan with either of two different amino acids, in this case, l-valine or l-allo-isoleucine.


Asunto(s)
Aspergillus nidulans , Aspergillus , Dicetopiperazinas , Péptido Sintasas , Triptófano , Triptófano/metabolismo , Triptófano/química , Dicetopiperazinas/química , Aspergillus nidulans/genética , Aspergillus nidulans/metabolismo , Aspergillus/química , Péptido Sintasas/metabolismo , Péptido Sintasas/genética , Estructura Molecular , Familia de Multigenes , Alcaloides Indólicos/química , Alcaloides Indólicos/metabolismo , Transferasas Alquil y Aril/metabolismo , Transferasas Alquil y Aril/genética
9.
Proc Natl Acad Sci U S A ; 121(26): e2321349121, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38889152

RESUMEN

Germ cells are regulated by local microenvironments (niches), which secrete instructive cues. Conserved developmental signaling molecules act as niche-derived regulatory factors, yet other types of niche signals remain to be identified. Single-cell RNA-sequencing of sexual planarians revealed niche cells expressing a nonribosomal peptide synthetase (nrps). Inhibiting nrps led to loss of female reproductive organs and testis hyperplasia. Mass spectrometry detected the dipeptide ß-alanyl-tryptamine (BATT), which is associated with reproductive system development and requires nrps and a monoamine-transmitter-synthetic enzyme Aromatic L-amino acid decarboxylase (AADC) for its production. Exogenous BATT rescued the reproductive defects after nrps or aadc inhibition, restoring fertility. Thus, a nonribosomal, monoamine-derived peptide provided by niche cells acts as a critical signal to trigger planarian reproductive development. These findings reveal an unexpected function for monoamines in niche-germ cell signaling. Furthermore, given the recently reported role for BATT as a male-derived factor required for reproductive maturation of female schistosomes, these results have important implications for the evolution of parasitic flatworms and suggest a potential role for nonribosomal peptides as signaling molecules in other organisms.


Asunto(s)
Planarias , Animales , Planarias/metabolismo , Femenino , Masculino , Péptido Sintasas/metabolismo , Péptido Sintasas/genética , Desarrollo Sexual , Péptidos/metabolismo , Reproducción/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
10.
Int J Mol Sci ; 25(11)2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38892200

RESUMEN

The pyoverdine siderophore is produced by Pseudomonas aeruginosa to access iron. Its synthesis involves the complex coordination of four nonribosomal peptide synthetases (NRPSs), which are responsible for assembling the pyoverdine peptide backbone. The precise cellular organization of these NRPSs and their mechanisms of interaction remain unclear. Here, we used a combination of several single-molecule microscopy techniques to elucidate the spatial arrangement of NRPSs within pyoverdine-producing cells. Our findings reveal that PvdL differs from the three other NRPSs in terms of localization and mobility patterns. PvdL is predominantly located in the inner membrane, while the others also explore the cytoplasmic compartment. Leveraging the power of multicolor single-molecule localization, we further reveal co-localization between PvdL and the other NRPSs, suggesting a pivotal role for PvdL in orchestrating the intricate biosynthetic pathway. Our observations strongly indicates that PvdL serves as a central orchestrator in the assembly of NRPSs involved in pyoverdine biosynthesis, assuming a critical regulatory function.


Asunto(s)
Oligopéptidos , Péptido Sintasas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/enzimología , Oligopéptidos/biosíntesis , Oligopéptidos/metabolismo , Péptido Sintasas/metabolismo , Péptido Sintasas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Sideróforos/biosíntesis , Sideróforos/metabolismo
11.
J Biol Chem ; 300(8): 107489, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38908753

RESUMEN

Nonribosomal peptide synthetases (NRPSs) are responsible for the production of important biologically active peptides. The large, multidomain NRPSs operate through an assembly line strategy in which the growing peptide is tethered to carrier domains that deliver the intermediates to neighboring catalytic domains. While most NRPS domains catalyze standard chemistry of amino acid activation, peptide bond formation, and product release, some canonical NRPS catalytic domains promote unexpected chemistry. The paradigm monobactam antibiotic sulfazecin is produced through the activity of a terminal thioesterase domain of SulM, which catalyzes an unusual ß-lactam-forming reaction in which the nitrogen of the C-terminal N-sulfo-2,3-diaminopropionate residue attacks its thioester tether to release the monobactam product. We have determined the structure of the thioesterase domain as both a free-standing domain and a didomain complex with the upstream holo peptidyl-carrier domain. The position of variant lid helices results in an active site pocket that is quite constrained, a feature that is likely necessary to orient the substrate properly for ß-lactam formation. Modeling of a sulfazecin tripeptide into the active site identifies a plausible binding mode identifying potential interactions for the sulfamate and the peptide backbone with Arg2849 and Asn2819, respectively. The overall structure is similar to the ß-lactone-forming thioesterase domain that is responsible for similar ring closure in the production of obafluorin. We further use these insights to enable bioinformatic analysis to identify additional, uncharacterized ß-lactam-forming biosynthetic gene clusters by genome mining.


Asunto(s)
Proteínas Bacterianas , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dominios Proteicos , Dominio Catalítico , Tioléster Hidrolasas/química , Tioléster Hidrolasas/metabolismo , Tioléster Hidrolasas/genética , Péptido Sintasas/química , Péptido Sintasas/metabolismo , Péptido Sintasas/genética , Cristalografía por Rayos X , Modelos Moleculares
12.
BMC Microbiol ; 24(1): 226, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937695

RESUMEN

BACKGROUND: Bacterial antimicrobial resistance poses a severe threat to humanity, necessitating the urgent development of new antibiotics. Recent advances in genome sequencing offer new avenues for antibiotic discovery. Paenibacillus genomes encompass a considerable array of antibiotic biosynthetic gene clusters (BGCs), rendering these species as good candidates for genome-driven novel antibiotic exploration. Nevertheless, BGCs within Paenibacillus genomes have not been extensively studied. RESULTS: We conducted an analysis of 554 Paenibacillus genome sequences, sourced from the National Center for Biotechnology Information database, with a focused investigation involving 89 of these genomes via antiSMASH. Our analysis unearthed a total of 848 BGCs, of which 716 (84.4%) were classified as unknown. From the initial pool of 554 Paenibacillus strains, we selected 26 available in culture collections for an in-depth evaluation. Genomic scrutiny of these selected strains unveiled 255 BGCs, encoding non-ribosomal peptide synthetases, polyketide synthases, and bacteriocins, with 221 (86.7%) classified as unknown. Among these strains, 20 exhibited antimicrobial activity against the gram-positive bacterium Micrococcus luteus, yet only six strains displayed activity against the gram-negative bacterium Escherichia coli. We proceeded to focus on Paenibacillus brasilensis, which featured five new BGCs for further investigation. To facilitate detailed characterization, we constructed a mutant in which a single BGC encoding a novel antibiotic was activated while simultaneously inactivating multiple BGCs using a cytosine base editor (CBE). The novel antibiotic was found to be localized to the cell wall and demonstrated activity against both gram-positive bacteria and fungi. The chemical structure of the new antibiotic was elucidated on the basis of ESIMS, 1D and 2D NMR spectroscopic data. The novel compound, with a molecular weight of 926, was named bracidin. CONCLUSIONS: This study outcome highlights the potential of Paenibacillus species as valuable sources for novel antibiotics. In addition, CBE-mediated dereplication of antibiotics proved to be a rapid and efficient method for characterizing novel antibiotics from Paenibacillus species, suggesting that it will greatly accelerate the genome-based development of new antibiotics.


Asunto(s)
Antibacterianos , Genoma Bacteriano , Familia de Multigenes , Paenibacillus , Paenibacillus/genética , Paenibacillus/metabolismo , Antibacterianos/farmacología , Antibacterianos/biosíntesis , Péptido Sintasas/genética , Sintasas Poliquetidas/genética , Bacteriocinas/genética , Bacteriocinas/farmacología , Bacteriocinas/biosíntesis , Vías Biosintéticas/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Descubrimiento de Drogas/métodos
13.
Angew Chem Int Ed Engl ; 63(33): e202406360, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-38822735

RESUMEN

Unnatural product (uNP) nonribosomal peptides promise to be a valuable source of pharmacophores for drug discovery. However, the extremely large size and complexity of the nonribosomal peptide synthetase (NRPS) enzymes pose formidable challenges to the production of such uNPs by combinatorial biosynthesis and synthetic biology. Here we report a new NRPS dissection strategy that facilitates the engineering and heterologous production of these NRPSs. This strategy divides NRPSs into "splitting units", each forming an enzyme subunit that contains catalytically independent modules. Functional collaboration between the subunits is then facilitated by artificially duplicating, at the N-terminus of the downstream subunit, the linker - thiolation domain - linker fragment that is resident at the C-terminus of the upstream subunit. Using the suggested split site that follows a conserved motif in the linker connecting the adenylation and the thiolation domains allows cognate or chimeric splitting unit pairs to achieve productivities that match, and in many cases surpass those of hybrid chimeric enzymes, and even those of intact NRPSs, upon production in a heterologous chassis. Our strategy provides facile options for the rational engineering of fungal NRPSs and for the combinatorial reprogramming of nonribosomal peptide production.


Asunto(s)
Péptido Sintasas , Ingeniería de Proteínas , Péptido Sintasas/metabolismo , Péptido Sintasas/química , Péptido Sintasas/genética , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/metabolismo
14.
Int J Mol Sci ; 25(11)2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38892087

RESUMEN

Utilizing bioinformatics tools, this study expands our understanding of secondary metabolism in Botrytis cinerea, identifying novel genes within polyketide synthase (PKS), non-ribosomal peptide synthetase (NRPS), sesquiterpene cyclase (STC), diterpene cyclase (DTC), and dimethylallyltryptophan synthase (DMATS) families. These findings enrich the genetic framework associated with B. cinerea's pathogenicity and ecological adaptation, offering insights into uncharted metabolic pathways. Significantly, the discovery of previously unannotated genes provides new molecular targets for developing targeted antifungal strategies, promising to enhance crop protection and advance our understanding of fungal biochemistry. This research not only broadens the scope of known secondary metabolites but also opens avenues for future exploration into B. cinerea's biosynthetic capabilities, potentially leading to novel antifungal compounds. Our work underscores the importance of integrating bioinformatics and genomics for fungal research, paving the way for sustainable agricultural practices by pinpointing precise molecular interventions against B. cinerea. This study sets a foundation for further investigations into the fungus's secondary metabolism, with implications for biotechnology and crop disease management.


Asunto(s)
Botrytis , Péptido Sintasas , Sintasas Poliquetidas , Metabolismo Secundario , Botrytis/genética , Botrytis/patogenicidad , Metabolismo Secundario/genética , Péptido Sintasas/genética , Péptido Sintasas/metabolismo , Sintasas Poliquetidas/genética , Sintasas Poliquetidas/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Biología Computacional/métodos , Familia de Multigenes , Genes Fúngicos
15.
Int J Mol Sci ; 25(10)2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38791129

RESUMEN

Next-generation sequencing has transformed the acquisition of vast amounts of genomic information, including the rapid identification of target gene sequences in metagenomic databases. However, dominant species can sometimes hinder the detection of rare bacterial species. Therefore, a highly sensitive amplification technique that can selectively amplify bacterial genomes containing target genes of interest was developed in this study. The rolling circle amplification (RCA) method can initiate amplification from a single locus using a specific single primer to amplify a specific whole genome. A mixed cell suspension was prepared using Pseudomonas fluorescens ATCC17400 (targeting nonribosomal peptide synthetase [NRPS]) and Escherichia coli (non-target), and a specific primer designed for the NRPS was used for the RCA reaction. The resulting RCA product (RCP) amplified only the Pseudomonas genome. The NRPS was successfully amplified using RCP as a template from even five cells, indicating that the single-priming RCA technique can specifically enrich the target genome using gene-specific primers. Ultimately, this specific genome RCA technique was applied to metagenomes extracted from sponge-associated bacteria, and NRPS sequences were successfully obtained from an unknown sponge-associated bacterium. Therefore, this method could be effective for accessing species-specific sequences of NRPS in unknown bacteria, including viable but non-culturable bacteria.


Asunto(s)
Genoma Bacteriano , Técnicas de Amplificación de Ácido Nucleico , Péptido Sintasas , Péptido Sintasas/genética , Técnicas de Amplificación de Ácido Nucleico/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Escherichia coli/genética , Pseudomonas fluorescens/genética , Análisis de Secuencia de ADN/métodos , Metagenoma/genética
16.
Molecules ; 29(9)2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38731473

RESUMEN

Chalkophomycin is a novel chalkophore with antibiotic activities isolated from Streptomyces sp. CB00271, while its potential in studying cellular copper homeostasis makes it an important probe and drug lead. The constellation of N-hydroxylpyrrole, 2H-oxazoline, diazeniumdiolate, and methoxypyrrolinone functional groups into one compact molecular architecture capable of coordinating cupric ions draws interest to unprecedented enzymology responsible for chalkophomycin biosynthesis. To elucidate the biosynthetic machinery for chalkophomycin production, the chm biosynthetic gene cluster from S. sp. CB00271 was identified, and its involvement in chalkophomycin biosynthesis was confirmed by gene replacement. The chm cluster was localized to a ~31 kb DNA region, consisting of 19 open reading frames that encode five nonribosomal peptide synthetases (ChmHIJLO), one modular polyketide synthase (ChmP), six tailoring enzymes (ChmFGMNQR), two regulatory proteins (ChmAB), and four resistance proteins (ChmA'CDE). A model for chalkophomycin biosynthesis is proposed based on functional assignments from sequence analysis and structure modelling, and is further supported by analogy to over 100 chm-type gene clusters in public databases. Our studies thus set the stage to fully investigate chalkophomycin biosynthesis and to engineer chalkophomycin analogues through a synthetic biology approach.


Asunto(s)
Familia de Multigenes , Péptido Sintasas , Sintasas Poliquetidas , Streptomyces , Streptomyces/genética , Streptomyces/enzimología , Streptomyces/metabolismo , Sintasas Poliquetidas/genética , Sintasas Poliquetidas/metabolismo , Sintasas Poliquetidas/química , Péptido Sintasas/metabolismo , Péptido Sintasas/genética , Péptido Sintasas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química
17.
Appl Microbiol Biotechnol ; 108(1): 325, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38717668

RESUMEN

Actinomycetota have been widely described as valuable sources for the acquisition of secondary metabolites. Most microbial metabolites are produced via metabolic pathways encoded by biosynthetic gene clusters (BGCs). Although many secondary metabolites are not essential for the survival of bacteria, they play an important role in their adaptation and interactions within microbial communities. This is how bacteria isolated from extreme environments such as Antarctica could facilitate the discovery of new BGCs with biotechnological potential. This study aimed to isolate rare Actinomycetota strains from Antarctic soil and sediment samples and identify their metabolic potential based on genome mining and exploration of biosynthetic gene clusters. To this end, the strains were sequenced using Illumina and Oxford Nanopore Technologies platforms. The assemblies were annotated and subjected to phylogenetic analysis. Finally, the BGCs present in each genome were identified using the antiSMASH tool, and the biosynthetic diversity of the Micrococcaceae family was evaluated. Taxonomic annotation revealed that seven strains were new and two were previously reported in the NCBI database. Additionally, BGCs encoding type III polyketide synthases (T3PKS), beta-lactones, siderophores, and non-ribosomal peptide synthetases (NRPS) have been identified, among others. In addition, the sequence similarity network showed a predominant type of BGCs in the family Micrococcaceae, and some genera were distinctly grouped. The BGCs identified in the isolated strains could be associated with applications such as antimicrobials, anticancer agents, and plant growth promoters, among others, positioning them as excellent candidates for future biotechnological applications and innovations. KEY POINTS: • Novel Antarctic rare Actinomycetota strains were isolated from soil and sediments • Genome-based taxonomic affiliation revealed seven potentially novel species • Genome mining showed metabolic potential for novel natural products.


Asunto(s)
Sedimentos Geológicos , Familia de Multigenes , Filogenia , Microbiología del Suelo , Regiones Antárticas , Sedimentos Geológicos/microbiología , Metabolismo Secundario/genética , Actinobacteria/genética , Actinobacteria/metabolismo , Actinobacteria/clasificación , Genoma Bacteriano , Biotecnología/métodos , Vías Biosintéticas/genética , Péptido Sintasas/genética , Péptido Sintasas/metabolismo , Sintasas Poliquetidas/genética , Sintasas Poliquetidas/metabolismo
18.
Structure ; 32(5): 520-522, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38701750

RESUMEN

In a recent issue of Nature Chemical Biology, Folger et al. demonstrated a high-throughput approach for engineering peptide bond forming domains from non-ribosomal peptide synthesis. A non-ribosomal peptide synthetase module from surfactin biosynthesis was reprogrammed to accept a fatty acid substrate into peptide biosynthesis, thus illustrating the potential of this approach for generating novel bioactive peptides.


Asunto(s)
Péptido Sintasas , Ingeniería de Proteínas , Péptido Sintasas/metabolismo , Péptido Sintasas/química , Péptido Sintasas/genética , Ingeniería de Proteínas/métodos , Péptidos/metabolismo , Péptidos/química
19.
Mol Plant Microbe Interact ; 37(5): 467-476, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38805410

RESUMEN

The soil-borne phytopathogenic gram-negative bacterium Ralstonia solanacearum species complex (RSSC) produces staphyloferrin B and micacocidin as siderophores that scavenge for trivalent iron (Fe3+) in the environment, depending on the intracellular divalent iron (Fe2+) concentration. The staphyloferrin B-deficient mutant reportedly retains its virulence, but the relationship between micacocidin and virulence remains unconfirmed. To elucidate the effect of micacocidin on RSSC virulence, we generated the micacocidin productivity-deficient mutant (ΔRSc1806) that lacks RSc1806, which encodes a putative polyketide synthase/non-ribosomal peptide synthetase, using the RSSC phylotype I Ralstonia pseudosolanacearum strain OE1-1. When incubated in the condition without Fe2+, ΔRSc1806 showed significantly lower Fe3+-scavenging activity, compared with OE1-1. Until 8 days after inoculation on tomato plants, ΔRSc1806 was not virulent, similar to the mutant (ΔphcA) missing phcA, which encodes the LysR-type transcriptional regulator PhcA that regulates the expression of the genes responsible for quorum sensing (QS)-dependent phenotypes including virulence. The transcriptome analysis revealed that RSc1806 deletion significantly altered the expression of more than 80% of the PhcA-regulated genes in the mutant grown in medium with or without Fe2+. Among the PhcA-regulated genes, the transcript levels of the genes whose expression was affected by the deletion of RSc1806 were strongly and positively correlated between the ΔRSc1806 and the phcA-deletion mutant. Furthermore, the deletion of RSc1806 significantly modified QS-dependent phenotypes, similar to the effects of the deletion of phcA. Collectively, our findings suggest that the deletion of micacocidin production-related RSc1806 alters the regulation of PhcA-regulated genes responsible for QS-dependent phenotypes including virulence as well as Fe3+-scavenging activity. [Formula: see text] Copyright © 2024 The Author(s). This is an open access article distributed under the CC BY-NC-ND 4.0 International license.


Asunto(s)
Proteínas Bacterianas , Regulación Bacteriana de la Expresión Génica , Enfermedades de las Plantas , Percepción de Quorum , Solanum lycopersicum , Percepción de Quorum/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Virulencia , Solanum lycopersicum/microbiología , Enfermedades de las Plantas/microbiología , Hierro/metabolismo , Ralstonia/genética , Ralstonia/patogenicidad , Sideróforos/metabolismo , Eliminación de Gen , Péptido Sintasas/genética , Péptido Sintasas/metabolismo
20.
Microb Cell Fact ; 23(1): 144, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38773450

RESUMEN

Fengycin is an important member of the lipopeptide family with a wide range of applications in the agricultural, food, medical and cosmetic industries. However, its commercial application is severely hindered by low productivity and high cost. Therefore, numerous studies have been devoted to improving the production of fengycin. We summarize these studies in this review with the aim of providing a reference and guidance for future researchers. This review begins with an overview of the synthesis mechanism of fengycin via the non-ribosomal peptide synthetases (NRPS), and then delves into the strategies for improving the fengycin production in recent years. These strategies mainly include fermentation optimization and metabolic engineering, and the metabolic engineering encompasses enhancement of precursor supply, application of regulatory factors, promoter engineering, and application of genome-engineering (genome shuffling and genome-scale metabolic network model). Finally, we conclude this review with a prospect of fengycin production.


Asunto(s)
Lipopéptidos , Ingeniería Metabólica , Ingeniería Metabólica/métodos , Lipopéptidos/biosíntesis , Lipopéptidos/metabolismo , Fermentación , Péptido Sintasas/genética , Péptido Sintasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA