Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.617
Filtrer
1.
Protein Sci ; 33(10): e5130, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39302095

RÉSUMÉ

Type III polyketide synthases (PKSs) catalyze the formation of a variety of polyketide natural products with remarkable structural diversity and biological activities. Despite significant progress in structural and mechanistic studies of type III PKSs in bacteria, fungi, and plants, research on type III PKSs in cyanobacteria is lacking. Here, we report structural and mechanistic insights into CylI, a type III PKS that catalyzes the formation of the alkylresorcinol intermediate in cylindrocyclophane biosynthesis. The crystal structure of apo-CylI reveals a distinct arrangement of structural elements that are proximal to the active site. We further solved the crystal structures of CylI in complexes with two substrate analogues at resolutions of 1.9 Å. The complex structures indicate that N259 is the key residue that determines the substrate preference of CylI. We also solved the crystal structure of CylI complexed with the alkylresorcinol product at a resolution of 2.0 Å. Structural analysis and mutagenesis experiments suggested that S170 functions as a key residue that determines cyclization specificity. On the basis of this result, a double mutant was engineered to completely switch the cyclization of CylI from aldol condensation to lactonization. This work elucidates the molecular basis of type III PKS in cyanobacteria and lays the foundation for engineering CylI-like enzymes to generate new products.


Sujet(s)
Cyanobactéries , Polyketide synthases , Polyketide synthases/composition chimique , Polyketide synthases/métabolisme , Polyketide synthases/génétique , Cristallographie aux rayons X , Cyanobactéries/enzymologie , Cyanobactéries/métabolisme , Cyanobactéries/génétique , Modèles moléculaires , Protéines bactériennes/composition chimique , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Domaine catalytique , Spécificité du substrat , Conformation des protéines
2.
Curr Microbiol ; 81(11): 379, 2024 Sep 28.
Article de Anglais | MEDLINE | ID: mdl-39340701

RÉSUMÉ

Streptomyces bikiniensis HD-087 is capable of synthesizing various antimicrobial substances to counter the detrimental effects of hazardous microorganisms. To elucidate whether it produces polyketide antibiotics and the synthesis mechanism of antibiotic substances, the metabolites and related genes of S. bikiniensis HD-087 were analyzed through LC-MS, anti-Magnaporthe oryzae activity detection, and bioinformatics approaches. The result indicated that the strain HD-087 could produce erythromycin, a polyketide antibiotic. The inhibitory zones of the fermentation supernatant of strain HD-087 and methanol solution of erythromycin extract against M. oryzae were 40.84 ± 0.68 mm and 33.18 ± 0.81 mm, respectively. The IC50 value of erythromycin extract for inhibiting spore germination of erythromycin extract was 220.43 µg/mL. There are two polyketide synthesis gene clusters in the genome of strain HD-087, namely t1pks-nrps and t3pks-lantipeptide-t1pks-nrps. The key gene pksL in the t3pks-lantipeptide-t1pks-nrps gene cluster was predicted. The results suggested that it encodes a stable, hydrophilic, and acidic protein, mainly composed of α-helix and random coil. The PksL protein contains dehydrogenase (DH), ketone reductase (KR), acyl carrier protein (ACP), and ketone synthase (KS) domains. Moreover, it can form interaction networks with 11 proteins containing domains, such as polyketide synthase and ACP synthase. The molecular docking between PksL and acetyl-CoA is stable and strong, suggesting that PksL protein could catalyze the synthesis of polyketides with CoA as a substrate. This study provides a theoretical basis for further exploring the polyketides synthesis mechanism and developing antifungal metabolites in S. bikiniensis HD-087.


Sujet(s)
Biologie informatique , Famille multigénique , Polyketide synthases , Streptomyces , Polyketide synthases/génétique , Polyketide synthases/métabolisme , Streptomyces/génétique , Streptomyces/métabolisme , Streptomyces/enzymologie , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Érythromycine/pharmacologie , Antifongiques/pharmacologie , Antifongiques/métabolisme , Polycétides/métabolisme , Polycétides/pharmacologie , Antibactériens/pharmacologie , Simulation de docking moléculaire , Ascomycota
3.
Food Res Int ; 195: 114961, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39277234

RÉSUMÉ

Monacolin K (MK), also known as lovastatin, is a polyketide compound with the ability to reduce plasma cholesterol levels and many other bio-activities. Red yeast rice (also named Hongqu) rich in MK derived from Monascus fermentation has attracted widespread attention due to its excellent performance in reducing blood lipids. However, industrial Monascus fermentation suffers from the limitations such as low yield of MK, long fermentation period, and susceptibility to contamination. In this study, we firstly blocked the competitive pathway of MK biosynthesis to create polyketide synthase gene pigA (the key gene responsible for the biosynthesis of Monascus azaphilone pigments) deficient strain A1. Then, based on the strategies to increase precursor supply for MK biosynthesis, acetyl-CoA carboxylase gene acc overexpression strains C1 and C2 were constructed with WT and A1 as the parent, respectively. Finally, histone deacetylase gene hos2 overexpression strain H1 was constructed by perturbation of histone acetylation modification. HPLC detection revealed all these four strains significantly increased their abilities to produce MK. After 14 days of solid-state fermentation, the MK yields of strains A1, C1, C2, and H1 reached 2.03 g/100 g, 1.81 g/100 g, 2.45 g/100 g and 2.52 g/100 g, which increased by 28.5 %, 14.7 %, 43.9 % and 36.1 % compared to WT, respectively. RT-qPCR results showed that overexpression of hos2 significantly increased the expression level of almost all genes responsible for MK biosynthesis after 5-day growth. Overall, the abilities of these strains to produce MK has been greatly improved, and MK production period has been shortened to 14 days from 20 days, providing new approaches for efficient production of Hongqu rich in MK.


Sujet(s)
Fermentation , Histone , Lovastatine , Monascus , Monascus/métabolisme , Monascus/génétique , Acétylation , Histone/métabolisme , Acetyl-coA carboxylase/métabolisme , Acetyl-coA carboxylase/génétique , Polyketide synthases/génétique , Polyketide synthases/métabolisme , Hypolipémiants/pharmacologie , Produits biologiques/métabolisme , Histone deacetylases/métabolisme , Histone deacetylases/génétique
4.
J Agric Food Chem ; 72(39): 21318-21343, 2024 Oct 02.
Article de Anglais | MEDLINE | ID: mdl-39302874

RÉSUMÉ

Bacterial trans-acyltransferase polyketide synthases (trans-AT PKSs) are among the most complex enzymes, which are responsible for generating a wide range of natural products, identified as trans-AT polyketides. These polyketides have received significant attention in drug development due to their structural diversity and potent bioactivities. With approximately 300 synthesized molecules discovered so far, trans-AT PKSs are found widespread in bacteria. Their biosynthesis pathways exhibit considerable genetic diversity, leading to the emergence of numerous enzymes with novel mechanisms, serving as a valuable resource for genetic engineering aimed at modifying small molecules' structures and creating new engineered enzymes. Despite the systematic discussions on trans-AT polyketides and their biosynthesis in earlier studies, the continuous advancements in tools, methods, compound identification, and biosynthetic pathways require a fresh update on accumulated knowledge. This review seeks to provide a comprehensive discussion for the 27 types of trans-AT polyketides discovered within the last seven years, detailing their sources, structures, biological activities, and biosynthetic pathways. By reviewing this new knowledge, a more profound understanding of the trans-AT polyketide family can be achieved.


Sujet(s)
Bactéries , Voies de biosynthèse , Polyketide synthases , Polycétides , Polycétides/métabolisme , Polycétides/composition chimique , Polyketide synthases/métabolisme , Polyketide synthases/génétique , Polyketide synthases/composition chimique , Bactéries/métabolisme , Bactéries/génétique , Bactéries/enzymologie , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/composition chimique , Découverte de médicament , Humains
5.
Gut Microbes ; 16(1): 2397874, 2024.
Article de Anglais | MEDLINE | ID: mdl-39229962

RÉSUMÉ

Recently, the mounting integration of probiotics into human health strategies has gathered considerable attention. Although the benefits of probiotics have been widely recognized in patients with gastrointestinal disorders, immune system modulation, and chronic-degenerative diseases, there is a growing need to evaluate their potential risks. In this context, new concerns have arisen regarding the safety of probiotics as some strains may have adverse effects in humans. Among these strains, Escherichia coli Nissle 1917 (EcN) exhibited traits of concern due to a pathogenic locus in its genome that produces potentially genotoxic metabolites. As the use of probiotics for therapeutic purposes is increasing, the effects of potentially harmful probiotics must be carefully evaluated. To this end, in this narrative review article, we reported the findings of the most relevant in vitro and in vivo studies investigating the expanding applications of probiotics and their impact on human well-being addressing concerns arising from the presence of antibiotic resistance and pathogenic elements, with a focus on the polyketide synthase (pks) pathogenic island of EcN. In this context, the literature data here discussed encourages a thorough profiling of probiotics to identify potential harmful elements as done for EcN where potential genotoxic effects of colibactin, a secondary metabolite, were observed. Specifically, while some studies suggest EcN is safe for gastrointestinal health, conflicting findings highlight the need for further research to clarify its safety and optimize its use in therapy. Overall, the data here presented suggest that a comprehensive assessment of the evolving landscape of probiotics is essential to make evidence-based decisions and ensure their correct use in humans.


Sujet(s)
Escherichia coli , Peptides , Polycétides , Probiotiques , Escherichia coli/génétique , Escherichia coli/métabolisme , Humains , Polycétides/métabolisme , Peptides/métabolisme , Peptides/génétique , Animaux , Mutagènes/métabolisme , Mutagènes/toxicité , Altération de l'ADN , Polyketide synthases/génétique , Polyketide synthases/métabolisme
6.
J Nat Prod ; 87(9): 2139-2147, 2024 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-39194958

RÉSUMÉ

ortho-Quinone methides (o-QMs) are a class of highly reactive intermediates that serve as important nonisolable building blocks (NBBs) in organic synthesis and small-molecule library construction. Because of their instability and nonisolability, most reported o-QMs are generated through in situ chemical synthesis, and only a few natural o-QMs have been reported due to the lack of directed discovery strategies. Herein, a new natural o-QM precursor (trichophenol A, 2) was identified from the fungal strain of Trichoderma sp. AT0167 through genome mining, which was generated by trilA (nonreducing polyketide synthase) and trilB (2-oxoglutarate dependent dioxygenase). Combinatorial biosynthesis via two other known NRPKS genes with trilA and trilB was performed, leading to the generation of five new trichophenol o-QM oligomers (trichophenols D-H, 5-9). The strategy combining genome mining with combinatorial biosynthesis not only targetedly uncovered a new natural o-QM precursor but also produced various new molecules through oligomerization of the new o-QM and its designated o-QM acceptors without chemical synthesis and isolation of intermediates, which was named NBB genome mining-combinatorial biosynthesis strategy for o-QM molecule library construction. This study provides a new strategy for the targeted discovery of natural o-QMs and small-molecule library construction with natural o-QMs.


Sujet(s)
Indolequinones , Indolequinones/composition chimique , Structure moléculaire , Polyketide synthases/métabolisme
7.
Science ; 385(6709): 671-678, 2024 08 09.
Article de Anglais | MEDLINE | ID: mdl-39116217

RÉSUMÉ

Prymnesium parvum are harmful haptophyte algae that cause massive environmental fish kills. Their polyketide polyether toxins, the prymnesins, are among the largest nonpolymeric compounds in nature and have biosynthetic origins that have remained enigmatic for more than 40 years. In this work, we report the "PKZILLAs," massive P. parvum polyketide synthase (PKS) genes that have evaded previous detection. PKZILLA-1 and -2 encode giant protein products of 4.7 and 3.2 megadaltons that have 140 and 99 enzyme domains. Their predicted polyene product matches the proposed pre-prymnesin precursor of the 90-carbon-backbone A-type prymnesins. We further characterize the variant PKZILLA-B1, which is responsible for the shorter B-type analog prymnesin-B1, from P. parvum RCC3426 and thus establish a general model of haptophyte polyether biosynthetic logic. This work expands expectations of genetic and enzymatic size limits in biology.


Sujet(s)
Haptophyta , , Polyketide synthases , Haptophyta/enzymologie , Haptophyta/génétique , Polyènes/métabolisme , Polyènes/composition chimique , /biosynthèse , Polyketide synthases/génétique , Polyketide synthases/métabolisme , Polycétides/métabolisme , Domaines protéiques
8.
Biochemistry ; 63(18): 2240-2244, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39186058

RÉSUMÉ

Engineered type I polyketide synthases (type I PKSs) can enable access to diverse polyketide pharmacophores and generate non-natural natural products. However, the promise of type I PKS engineering remains modestly realized at best. Here, we report that ketosynthase (KS) domains, the key carbon-carbon bond-forming catalysts, control which intermediates are allowed to progress along the PKS assembly lines and which intermediates are excluded. Using bimodular PKSs, we demonstrate that KSs can be exquisitely selective for the upstream polyketide substrate while retaining promiscuity for the extender unit that they incorporate. It is then the downstream KS that acts as a gatekeeper to ensure the fidelity of the extender unit incorporation by the upstream KS. We also demonstrate that these findings are not universally applicable; substrate-tolerant KSs do allow engineered polyketide intermediates to be extended. Our results demonstrate the utility for evaluating the KS-induced bottlenecks to gauge the feasibility of engineering PKS assembly lines.


Sujet(s)
Polyketide synthases , Ingénierie des protéines , Polyketide synthases/métabolisme , Polyketide synthases/composition chimique , Polyketide synthases/génétique , Ingénierie des protéines/méthodes , Polycétides/métabolisme , Polycétides/composition chimique , Spécificité du substrat , Domaines protéiques
9.
Curr Opin Chem Biol ; 81: 102507, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39098210

RÉSUMÉ

Polyketides represent an important class of natural products, renowned for their intricate structures and diverse biological activities. In contrast to common fatty acids, polyketides possess relatively more rigid carbon skeletons, more complex ring systems, and chiral centers. These structural features are primarily achieved through distinctive enzymatic cyclizations and oxidations as tailoring steps. In this opinion, we discuss the recent progress in deciphering the mechanisms of cyclization and oxidation within polyketide biosynthesis. By shedding light on these enzymatic processes, this article seeks to motivate the community to unravel the remaining mysteries surrounding cyclase and oxidase functionalities and to explore novel polyketide natural products through genome mining.


Sujet(s)
Oxydoréduction , Polycétides , Polycétides/métabolisme , Polycétides/composition chimique , Cyclisation , Produits biologiques/métabolisme , Produits biologiques/composition chimique , Polyketide synthases/métabolisme
10.
Nat Commun ; 15(1): 6485, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090122

RÉSUMÉ

The modular nature of polyketide assembly lines and the significance of their products make them prime targets for combinatorial engineering. The recently updated module boundary has been successful for engineering short synthases, yet larger synthases constructed using the updated boundary have not been investigated. Here we describe our design and implementation of a BioBricks-like platform to rapidly construct 5 triketide, 25 tetraketide, and 125 pentaketide synthases to test every module combination of the pikromycin synthase. Anticipated products are detected from 60% of the triketide synthases, 32% of the tetraketide synthases, and 6.4% of the pentaketide synthases. We determine ketosynthase gatekeeping and module-skipping are the principal impediments to obtaining functional synthases. The platform is also employed to construct active hybrid synthases by incorporating modules from the erythromycin, spinosyn, and rapamycin assembly lines. The relaxed gatekeeping of a ketosynthase in the rapamycin synthase is especially encouraging in the quest to produce designer polyketides.


Sujet(s)
Macrolides , Polyketide synthases , Polyketide synthases/métabolisme , Polyketide synthases/génétique , Macrolides/métabolisme , Ingénierie des protéines/méthodes , Érythromycine , Polycétides/métabolisme , Polycétides/composition chimique , Streptomyces/enzymologie , Streptomyces/génétique , Sirolimus , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique
11.
Curr Opin Chem Biol ; 82: 102510, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39128325

RÉSUMÉ

Mushroom-forming fungi exhibit a distinctive ecology, which is unsurprisingly also reflected in unique and divergent biosynthetic pathways. We review this phenomenon through the lens of the polyketide metabolism, where mushrooms often deviate from established principles and challenge conventional paradigms. This is evident not only by non-canonical enzyme architectures and functions but also by their propensity for multi-product synthases rather than single-product pathways. Nevertheless, mushrooms also feature many polyketides familiar from plants, bacteria, and fungi of their sister division Ascomycota, which, however, are the result of an independent evolution. In this regard, the captivating biosynthetic pathways of mushrooms might even help us understand the biological pressures that led to the simultaneous production of the same natural products (via convergent evolution, co-evolution, and/or metaevolution) and thus address the question of their raison d'être.


Sujet(s)
Agaricales , Polyketide synthases , Polyketide synthases/métabolisme , Polyketide synthases/génétique , Agaricales/enzymologie , Agaricales/métabolisme , Polycétides/métabolisme , Polycétides/composition chimique , Voies de biosynthèse , Produits biologiques/métabolisme , Produits biologiques/composition chimique
12.
Adv Sci (Weinh) ; 11(35): e2401708, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38995095

RÉSUMÉ

Modular polyketide synthases (PKSs) are capable of synthesizing diverse natural products with fascinating bioactivities. Canonical enoyl-CoA hydratases (ECHs) are components of the ß-branching cassette that modifies the polyketide chain by adding a ß-methyl branch. Herein, it is demonstrated that the deletion of an atypical ECHQ domain (featuring a Q280 residue) of Art21, a didomain protein contains an ECHQ domain and a thioesterase (TE) domain, reprograms the polyketide assembly line from synthesizing tetracyclic aurantinins (ARTs) to bicyclic auritriacids (ATAs) with much lower antibacterial activities. Genes encoding the ECHQ-TE didomain proteins distribute in many PKS gene clusters from different bacteria. Significantly, the ART PKS machinery can be directed to make ARTs, ATAs, or both of them by employing appropriate ECHQ-TE proteins, implying a great potential for using this reprogramming strategy in polyketide structure diversification.


Sujet(s)
Énoyl-CoA hydratases , Polyketide synthases , Polycétides , Polycétides/métabolisme , Polyketide synthases/métabolisme , Polyketide synthases/génétique , Polyketide synthases/composition chimique , Énoyl-CoA hydratases/métabolisme , Énoyl-CoA hydratases/génétique , Famille multigénique
13.
Nat Commun ; 15(1): 5687, 2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-38971862

RÉSUMÉ

Base editing (BE) faces protospacer adjacent motif (PAM) constraints and off-target effects in both eukaryotes and prokaryotes. For Streptomyces, renowned as one of the most prolific bacterial producers of antibiotics, the challenges are more pronounced due to its diverse genomic content and high GC content. Here, we develop a base editor named eSCBE3-NG-Hypa, tailored with both high efficiency and -fidelity for Streptomyces. Of note, eSCBE3-NG-Hypa recognizes NG PAM and exhibits high activity at challenging sites with high GC content or GC motifs, while displaying minimal off-target effects. To illustrate its practicability, we employ eSCBE3-NG-Hypa to achieve precise key amino acid conversion of the dehydratase (DH) domains within the modular polyketide synthase (PKS) responsible for the insecticide avermectins biosynthesis, achieving domains inactivation. The resulting DH-inactivated mutants, while ceasing avermectins production, produce a high yield of oligomycin, indicating competitive relationships among multiple biosynthetic gene clusters (BGCs) in Streptomyces avermitilis. Leveraging this insight, we use eSCBE3-NG-Hypa to introduce premature stop codons into competitor gene cluster of ave in an industrial S. avermitilis, with the mutant Δolm exhibiting the highest 4.45-fold increase in avermectin B1a compared to the control. This work provides a potent tool for modifying biosynthetic pathways and advancing metabolic engineering in Streptomyces.


Sujet(s)
Systèmes CRISPR-Cas , Cytosine , Édition de gène , Polyketide synthases , Streptomyces , Streptomyces/génétique , Streptomyces/métabolisme , Édition de gène/méthodes , Polyketide synthases/génétique , Polyketide synthases/métabolisme , Cytosine/métabolisme , Ivermectine/analogues et dérivés , Ivermectine/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Oligomycines
14.
Microb Cell Fact ; 23(1): 202, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39026365

RÉSUMÉ

BACKGROUND: Microbial genome sequencing and analysis revealed the presence of abundant silent secondary metabolites biosynthetic gene clusters (BGCs) in streptomycetes. Activating these BGCs has great significance for discovering new compounds and novel biosynthetic pathways. RESULTS: In this study, we found that ovmZ and ovmW homologs, a pair of interdependent transcriptional regulators coding genes, are widespread in actinobacteria and closely associated with the biosynthesis of secondary metabolites. Through co-overexpression of native ovmZ and ovmW in Streptomyces neyagawaensis NRRL B-3092, a silent type II polyketide synthase (PKS) gene cluster was activated to produce gephyromycin A, tetrangomycin and fridamycin E with the yields of 22.3 ± 8.0 mg/L, 4.8 ± 0.5 mg/L and 20.3 ± 4.1 mg/L respectively in the recombinant strain of S.ne/pZnWn. However, expression of either ovmZ or ovmW failed to activate this gene cluster. Interestingly, overexpression of the heterologous ovmZ and ovmW pair from oviedomycin BGC of S. ansochromogenes 7100 also led to awakening of this silent angucyclinone BGC in S. neyagawaensis. CONCLUSION: A silent angucyclinone BGC was activated by overexpressing both ovmZ and ovmW in S. neyagawaensis. Due to the wide distribution of ovmZ and ovmW in the BGCs of actinobacteria, co-overexpression of ovmZ and ovmW could be a strategy for activating silent BGCs, thus stimulating the biosynthesis of secondary metabolites.


Sujet(s)
Anthraquinones , Antibactériens , Famille multigénique , Streptomyces , Streptomyces/génétique , Streptomyces/métabolisme , Antibactériens/biosynthèse , Anthraquinones/métabolisme , Régulation de l'expression des gènes bactériens , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Voies de biosynthèse/génétique , Polyketide synthases/génétique , Polyketide synthases/métabolisme , Métabolisme secondaire/génétique ,
15.
J Agric Food Chem ; 72(31): 17499-17509, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39045837

RÉSUMÉ

The natural compound (R)-(-)-mellein exhibits antiseptic and fungicidal activities. We investigated its biosynthesis using the polyketide synthase encoded by SACE_5532 (pks8) from Saccharopolyspora erythraea heterologously expressed in Streptomyces albus B4, a chassis chosen for its fast growth, genetic manipulability, and ample large short-chain acyl-CoA precursor supply. High-level heterologous (R)-(-)-mellein yield was achieved by pks8 overexpression and duplication. The precursor supply pathways were strengthened by overexpression of SACE_0028 (encoding acetyl-CoA carboxylase) and four genes involved in ß-oxidation (fadD, fadE, fadB, and fadA). Cell growth inhibition by (R)-(-)-mellein production at high concentration was relieved by in situ adsorption using Amberlite XAD16 resin. The final strain, B4mel12, produced (R)-(-)-mellein at 6395.2 mg/L in shake-flask fermentation. Overall, this is the first report of heterologous (R)-(-)-mellein synthesis in microorganism with a high titer. (R)-(-)-mellein prototype in this study opens a possibility for the overproduction of valuable melleins in S. albus B4.


Sujet(s)
Protéines bactériennes , Génie métabolique , Polyketide synthases , Streptomyces , Streptomyces/génétique , Streptomyces/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Polyketide synthases/génétique , Polyketide synthases/métabolisme , Fermentation , Saccharopolyspora/génétique , Saccharopolyspora/métabolisme , Acetyl-coA carboxylase/génétique , Acetyl-coA carboxylase/métabolisme
16.
Chem Commun (Camb) ; 60(66): 8712-8715, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39056119

RÉSUMÉ

A VMYH motif was determined to help ketosynthases in polyketide assembly lines select α,ß-unsaturated intermediates from an equilibrium mediated by an upstream dehydratase. Alterations of this motif decreased ketosynthase selectivity within a model tetraketide synthase, most significantly when replaced by the TNGQ motif of ketosynthases that accept D-ß-hydroxy intermediates.


Sujet(s)
Hydro-lyases , Polyketide synthases , Polyketide synthases/métabolisme , Polyketide synthases/composition chimique , Hydro-lyases/métabolisme , Hydro-lyases/composition chimique
17.
Curr Biol ; 34(16): 3698-3706.e4, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-38986615

RÉSUMÉ

The catastrophic loss of aquatic life in the Central European Oder River in 2022, caused by a toxic bloom of the haptophyte microalga Prymnesium parvum (in a wide sense, s.l.), underscores the need to improve our understanding of the genomic basis of the toxin. Previous morphological, phylogenetic, and genomic studies have revealed cryptic diversity within P. parvum s.l. and uncovered three clade-specific (types A, B, and C) prymnesin toxins. Here, we used state-of-the-art long-read sequencing and assembled the first haplotype-resolved diploid genome of a P. parvum type B from the strain responsible for the Oder disaster. Comparative analyses with type A genomes uncovered a genome-size expansion driven by repetitive elements in type B. We also found conserved synteny but divergent evolution in several polyketide synthase (PKS) genes, which are known to underlie toxin production in combination with environmental cues. We identified an approximately 20-kbp deletion in the largest PKS gene of type B that we link to differences in the chemical structure of types A and B prymnesins. Flow cytometry and electron microscopy analyses confirmed diploidy in the Oder River strain and revealed differences to closely related strains in both ploidy and morphology. Our results provide unprecedented resolution of strain diversity in P. parvum s.l. and a better understanding of the genomic basis of toxin variability in haptophytes. The reference-quality genome will enable us to better understand changes in microbial diversity in the face of increasing environmental pressures and provides a basis for strain-level monitoring of invasive Prymnesium in the future.


Sujet(s)
Haptophyta , Haptophyta/génétique , Haplotypes , Microalgues/génétique , Toxines de la flore et de la faune marines/génétique , Animaux , Phylogenèse , Poissons/génétique , Polyketide synthases/génétique , Polyketide synthases/métabolisme
18.
Appl Microbiol Biotechnol ; 108(1): 427, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39046587

RÉSUMÉ

Filamentous fungi are prolific producers of bioactive natural products and play a vital role in drug discovery. Yet, their potential cannot be fully exploited since many biosynthetic genes are silent or cryptic under laboratory culture conditions. Several strategies have been applied to activate these genes, with heterologous expression as one of the most promising approaches. However, successful expression and identification of new products are often hindered by host-dependent factors, such as low gene targeting efficiencies, a high metabolite background, or a lack of selection markers. To overcome these challenges, we have constructed a Penicillium crustosum expression host in a pyrG deficient strain by combining the split-marker strategy and CRISPR-Cas9 technology. Deletion of ligD and pcribo improved gene targeting efficiencies and enabled the use of an additional selection marker in P. crustosum. Furthermore, we reduced the secondary metabolite background by inactivation of two highly expressed gene clusters and abolished the formation of the reactive ortho-quinone methide. Finally, we replaced the P. crustosum pigment gene pcr4401 with the commonly used Aspergillus nidulans wA expression site for convenient use of constructs originally designed for A. nidulans in our P. crustosum host strain. As proof of concept, we successfully expressed a single polyketide synthase gene and an entire gene cluster at the P. crustosum wA locus. Resulting transformants were easily detected by their albino phenotype. With this study, we provide a highly efficient platform for heterologous expression of fungal genes. KEY POINTS: Construction of a highly efficient Penicillium crustosum heterologous expression host Reduction of secondary metabolite background by genetic dereplication strategy Integration of wA site to provide an alternative host besides Aspergillus nidulans.


Sujet(s)
Systèmes CRISPR-Cas , Penicillium , Métabolisme secondaire , Penicillium/génétique , Penicillium/métabolisme , Métabolisme secondaire/génétique , Aspergillus nidulans/génétique , Aspergillus nidulans/métabolisme , Polyketide synthases/génétique , Polyketide synthases/métabolisme , Famille multigénique , Ciblage de gène/méthodes , Régulation de l'expression des gènes fongiques , Protéines fongiques/génétique , Protéines fongiques/métabolisme , Voies de biosynthèse/génétique , Génie métabolique/méthodes , Expression des gènes
19.
Fungal Genet Biol ; 174: 103912, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39004163

RÉSUMÉ

The Fusarium solani species complex (FSSC) is comprised of important pathogens of plants and humans. A distinctive feature of FSSC species is perithecial pigmentation. While the dark perithecial pigments of other Fusarium species are derived from fusarubins synthesized by polyketide synthase 3 (PKS3), the perithecial pigments of FSSC are derived from an unknown metabolite synthesized by PKS35. Here, we confirm in FSSC species Fusarium vanettenii that PKS35 (fsnI) is required for perithecial pigment synthesis by deletion analysis and that fsnI is closely related to phnA from Penicillium herquei, as well as duxI from Talaromyces stipentatus, which produce prephenalenone as an early intermediate in herqueinone and duclauxin synthesis respectively. The production of prephenalenone by expression of fsnI in Saccharomyces cerevisiae indicates that it is also an early intermediate in perithecial pigment synthesis. We next identified a conserved cluster of 10 genes flanking fsnI in F. vanettenii that when expressed in F. graminearum led to the production of a novel corymbiferan lactone F as a likely end product of the phenalenone biosynthetic pathway in FSSC.


Sujet(s)
Voies de biosynthèse , Fusarium , Phénalènes , Pigmentation , Polyketide synthases , Fusarium/génétique , Fusarium/métabolisme , Phénalènes/métabolisme , Voies de biosynthèse/génétique , Polyketide synthases/génétique , Polyketide synthases/métabolisme , Pigmentation/génétique , Protéines fongiques/génétique , Protéines fongiques/métabolisme , Pigments biologiques/métabolisme , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Talaromyces/génétique , Talaromyces/métabolisme , Penicillium/génétique , Penicillium/métabolisme
20.
J Am Soc Mass Spectrom ; 35(9): 2136-2142, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39038158

RÉSUMÉ

The type 1 polyketide synthase (PKS) assembly line uses its modular structure to produce polyketide natural products that form the basis of many pharmaceuticals. Currently, several cryoelectron microscopy (cryo-EM) structures of a multidomain PKS module have been constructed, but much remains to be learned. Here we utilize ion-mobility mass spectrometry (IM-MS) to record size and shape information and detect different conformational states of a 207 kDa didomain dimer comprised of ketosynthase (KS) and acyl transferase (AT), excised from full-length module. Furthermore, gas-phase stability differences between these different conformations are captured by collision induced unfolding (CIU) technology. Additionally, through tracking these forms as a function of time, we elucidate a detailed disassembly pathway for KS-AT dimers for the first time.


Sujet(s)
Spectrométrie de mobilité ionique , Polyketide synthases , Polycétides , Spectrométrie de mobilité ionique/méthodes , Polyketide synthases/composition chimique , Polyketide synthases/métabolisme , Polycétides/composition chimique , Polycétides/métabolisme , Spectrométrie de masse/méthodes , Multimérisation de protéines , Modèles moléculaires , Conformation des protéines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE