Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.632
Filtrer
1.
Microb Cell Fact ; 23(1): 194, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970033

RÉSUMÉ

BACKGROUND: Biotransformation of CO2 into high-value-added carbon-based products is a promising process for reducing greenhouse gas emissions. To realize the green transformation of CO2, we use fatty acids as carbon source to drive CO2 fixation to produce succinate through a portion of the 3-hydroxypropionate (3HP) cycle in Cupriavidus necator H16. RESULTS: This work can achieve the production of a single succinate molecule from one acetyl-CoA molecule and two CO2 molecules. It was verified using an isotope labeling experiment utilizing NaH13CO3. This implies that 50% of the carbon atoms present in succinate are derived from CO2, resulting in a twofold increase in efficiency compared to prior methods of succinate biosynthesis that relied on the carboxylation of phosphoenolpyruvate or pyruvate. Meanwhile, using fatty acid as a carbon source has a higher theoretical yield than other feedstocks and also avoids carbon loss during acetyl-CoA and succinate production. To further optimize succinate production, different approaches including the optimization of ATP and NADPH supply, optimization of metabolic burden, and optimization of carbon sources were used. The resulting strain was capable of producing succinate to a level of 3.6 g/L, an increase of 159% from the starting strain. CONCLUSIONS: This investigation established a new method for the production of succinate by the implementation of two CO2 fixation reactions and demonstrated the feasibility of ATP, NADPH, and metabolic burden regulation strategies in biological carbon fixation.


Sujet(s)
Dioxyde de carbone , Cupriavidus necator , Acides gras , Acide succinique , Dioxyde de carbone/métabolisme , Cupriavidus necator/métabolisme , Acides gras/métabolisme , Acide succinique/métabolisme , Acétyl coenzyme A/métabolisme , NADP/métabolisme
2.
Nat Commun ; 15(1): 5388, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918376

RÉSUMÉ

Heparan sulfate (HS) is degraded in lysosome by a series of glycosidases. Before the glycosidases can act, the terminal glucosamine of HS must be acetylated by the integral lysosomal membrane enzyme heparan-α-glucosaminide N-acetyltransferase (HGSNAT). Mutations of HGSNAT cause HS accumulation and consequently mucopolysaccharidosis IIIC, a devastating lysosomal storage disease characterized by progressive neurological deterioration and early death where no treatment is available. HGSNAT catalyzes a unique transmembrane acetylation reaction where the acetyl group of cytosolic acetyl-CoA is transported across the lysosomal membrane and attached to HS in one reaction. However, the reaction mechanism remains elusive. Here we report six cryo-EM structures of HGSNAT along the reaction pathway. These structures reveal a dimer arrangement and a unique structural fold, which enables the elucidation of the reaction mechanism. We find that a central pore within each monomer traverses the membrane and controls access of cytosolic acetyl-CoA to the active site at its luminal mouth where glucosamine binds. A histidine-aspartic acid catalytic dyad catalyzes the transfer reaction via a ternary complex mechanism. Furthermore, the structures allow the mapping of disease-causing variants and reveal their potential impact on the function, thus creating a framework to guide structure-based drug discovery efforts.


Sujet(s)
Acetyltransferases , Cryomicroscopie électronique , Lysosomes , Mucopolysaccharidose de type III , Mucopolysaccharidose de type III/génétique , Mucopolysaccharidose de type III/métabolisme , Mucopolysaccharidose de type III/enzymologie , Humains , Lysosomes/métabolisme , Lysosomes/enzymologie , Acetyltransferases/métabolisme , Acetyltransferases/composition chimique , Acetyltransferases/génétique , Domaine catalytique , Mutation , Héparitine sulfate/métabolisme , Acétyl coenzyme A/métabolisme , Acétyl coenzyme A/composition chimique , Modèles moléculaires , Glucosamine/métabolisme , Glucosamine/composition chimique , Acétylation , Membranes intracellulaires/métabolisme
3.
Microb Cell Fact ; 23(1): 173, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38867236

RÉSUMÉ

BACKGROUND: The microbial chiral product (R)-3-hydroxybutyrate (3-HB) is a gateway to several industrial and medical compounds. Acetyl-CoA is the key precursor for 3-HB, and several native pathways compete with 3-HB production. The principal competing pathway in wild-type Escherichia coli for acetyl-CoA is mediated by citrate synthase (coded by gltA), which directs over 60% of the acetyl-CoA into the tricarboxylic acid cycle. Eliminating citrate synthase activity (deletion of gltA) prevents growth on glucose as the sole carbon source. In this study, an alternative approach is used to generate an increased yield of 3-HB: citrate synthase activity is reduced but not eliminated by targeted substitutions in the chromosomally expressed enzyme. RESULTS: Five E. coli GltA variants were examined for 3-HB production via heterologous overexpression of a thiolase (phaA) and NADPH-dependent acetoacetyl-CoA reductase (phaB) from Cupriavidus necator. In shake flask studies, four variants showed nearly 5-fold greater 3-HB yield compared to the wild-type, although pyruvate accumulated. Overexpression of either native thioesterases TesB or YciA eliminated pyruvate formation, but diverted acetyl-CoA towards acetate formation. Overexpression of pantothenate kinase similarly decreased pyruvate formation but did not improve 3-HB yield. Controlled batch studies at the 1.25 L scale demonstrated that the GltA[A267T] variant produced the greatest 3-HB titer of 4.9 g/L with a yield of 0.17 g/g. In a phosphate-starved repeated batch process, E. coli ldhA poxB pta-ackA gltA::gltA[A267T] generated 15.9 g/L 3-HB (effective concentration of 21.3 g/L with dilution) with yield of 0.16 g/g from glucose as the sole carbon source. CONCLUSIONS: This study demonstrates that GltA variants offer a means to affect the generation of acetyl-CoA derived products. This approach should benefit a wide range of acetyl-CoA derived biochemical products in E. coli and other microbes. Enhancing substrate affinity of the introduced pathway genes like thiolase towards acetyl-CoA will likely further increase the flux towards 3-HB while reducing pyruvate and acetate accumulation.


Sujet(s)
Acide 3-hydroxy-butyrique , Acétyl coenzyme A , Citrate (si)-synthase , Escherichia coli , Escherichia coli/génétique , Escherichia coli/métabolisme , Acétyl coenzyme A/métabolisme , Citrate (si)-synthase/métabolisme , Citrate (si)-synthase/génétique , Acide 3-hydroxy-butyrique/métabolisme , Acide 3-hydroxy-butyrique/biosynthèse , Génie métabolique/méthodes , Protéines Escherichia coli/métabolisme , Protéines Escherichia coli/génétique , Cetone oxidoreductases/métabolisme , Cetone oxidoreductases/génétique , Alcohol oxidoreductases
4.
Science ; 384(6701): eadj4301, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38870309

RÉSUMÉ

Mitochondria are critical for proper organ function and mechanisms to promote mitochondrial health during regeneration would benefit tissue homeostasis. We report that during liver regeneration, proliferation is suppressed in electron transport chain (ETC)-dysfunctional hepatocytes due to an inability to generate acetyl-CoA from peripheral fatty acids through mitochondrial ß-oxidation. Alternative modes for acetyl-CoA production from pyruvate or acetate are suppressed in the setting of ETC dysfunction. This metabolic inflexibility forces a dependence on ETC-functional mitochondria and restoring acetyl-CoA production from pyruvate is sufficient to allow ETC-dysfunctional hepatocytes to proliferate. We propose that metabolic inflexibility within hepatocytes can be advantageous by limiting the expansion of ETC-dysfunctional cells.


Sujet(s)
Acétyl coenzyme A , Hépatocytes , Régénération hépatique , Mitochondries du foie , Acide pyruvique , Animaux , Hépatocytes/métabolisme , Acétyl coenzyme A/métabolisme , Souris , Acide pyruvique/métabolisme , Mitochondries du foie/métabolisme , Oxydoréduction , Prolifération cellulaire , Acides gras/métabolisme , Foie/métabolisme , Transport d'électrons , Souris de lignée C57BL , Mitochondries/métabolisme , Mâle
5.
Cancer Lett ; 595: 217006, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-38823763

RÉSUMÉ

Driver genomic mutations in tumors define specific molecular subtypes that display distinct malignancy competence, therapeutic resistance and clinical outcome. Although TP53 mutation has been identified as the most common mutation in hepatocellular carcinoma (HCC), current understanding on the biological traits and therapeutic strategies of this subtype has been largely unknown. Here, we reveal that fatty acid ß oxidation (FAO) is remarkable repressed in TP53 mutant HCC and which links to poor prognosis in HCC patients. We further demonstrate that carnitine palmitoyltransferase 1 (CPT1A), the rate-limiting enzyme of FAO, is universally downregulated in liver tumor tissues, and which correlates with poor prognosis in HCC and promotes HCC progression in the de novo liver tumor and xenograft tumor models. Mechanically, hepatic Cpt1a loss disrupts lipid metabolism and acetyl-CoA production. Such reduction in acetyl-CoA reduced histone acetylation and epigenetically reprograms branched-chain amino acids (BCAA) catabolism, and leads to the accumulation of cellular BCAAs and hyperactivation of mTOR signaling. Importantly, we reveal that genetic ablation of CPT1A renders TP53 mutant liver cancer mTOR-addicted and sensitivity to mTOR inhibitor AZD-8055 treatment. Consistently, Cpt1a loss in HCC directs tumor cell therapeutic response to AZD-8055. CONCLUSION: Our results show genetic evidence for CPT1A as a metabolic tumor suppressor in HCC and provide a therapeutic approach for TP53 mutant HCC patients.


Sujet(s)
Carcinome hépatocellulaire , Carnitine O-palmitoyltransferase , Tumeurs du foie , Mutation , Protéine p53 suppresseur de tumeur , Humains , Carnitine O-palmitoyltransferase/génétique , Carnitine O-palmitoyltransferase/métabolisme , Carnitine O-palmitoyltransferase/antagonistes et inhibiteurs , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Animaux , Souris , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Acides aminés à chaine ramifiée/métabolisme , Lignée cellulaire tumorale , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Métabolisme lipidique/génétique , Transduction du signal , Acétyl coenzyme A/métabolisme , Régulation de l'expression des gènes tumoraux , Mâle
6.
Cell Death Dis ; 15(6): 392, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38834617

RÉSUMÉ

Keratinocyte proliferation and differentiation in epidermis are well-controlled and essential for reacting to stimuli such as ultraviolet light. Imbalance between proliferation and differentiation is a characteristic feature of major human skin diseases such as psoriasis and squamous cell carcinoma. However, the effect of keratinocyte metabolism on proliferation and differentiation remains largely elusive. We show here that the gluconeogenic enzyme fructose-1,6-bisphosphatase 1 (FBP1) promotes differentiation while inhibits proliferation of keratinocyte and suppresses psoriasis development. FBP1 is identified among the most upregulated genes induced by UVB using transcriptome sequencing and is elevated especially in upper epidermis. Fbp1 heterozygous mice exhibit aberrant epidermis phenotypes with local hyperplasia and dedifferentiation. Loss of FBP1 promotes proliferation and inhibits differentiation of keratinocytes in vitro. Mechanistically, FBP1 loss facilitates glycolysis-mediated acetyl-CoA production, which increases histone H3 acetylation at lysine 9, resulting in enhanced transcription of proliferation genes. We further find that the expression of FBP1 is dramatically reduced in human psoriatic lesions and in skin of mouse imiquimod psoriasis model. Fbp1 deficiency in mice facilitates psoriasis-like skin lesions development through glycolysis and acetyl-CoA production. Collectively, our findings reveal a previously unrecognized role of FBP1 in epidermal homeostasis and provide evidence for FBP1 as a metabolic psoriasis suppressor.


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Fructose-1,6-diphosphatase , Histone , Kératinocytes , Psoriasis , Animaux , Humains , Souris , Acétyl coenzyme A/métabolisme , Acétylation , Modèles animaux de maladie humaine , Fructose-1,6-diphosphatase/métabolisme , Fructose-1,6-diphosphatase/génétique , Glycolyse , Histone/métabolisme , Kératinocytes/métabolisme , Kératinocytes/anatomopathologie , Souris de lignée C57BL , Psoriasis/anatomopathologie , Psoriasis/métabolisme , Psoriasis/génétique
7.
Nature ; 630(8016): 466-474, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38839952

RÉSUMÉ

Histone acetylation regulates gene expression, cell function and cell fate1. Here we study the pattern of histone acetylation in the epithelial tissue of the Drosophila wing disc. H3K18ac, H4K8ac and total lysine acetylation are increased in the outer rim of the disc. This acetylation pattern is controlled by nuclear position, whereby nuclei continuously move from apical to basal locations within the epithelium and exhibit high levels of H3K18ac when they are in proximity to the tissue surface. These surface nuclei have increased levels of acetyl-CoA synthase, which generates the acetyl-CoA for histone acetylation. The carbon source for histone acetylation in the rim is fatty acid ß-oxidation, which is also increased in the rim. Inhibition of fatty acid ß-oxidation causes H3K18ac levels to decrease in the genomic proximity of genes involved in disc development. In summary, there is a physical mark of the outer rim of the wing and other imaginal epithelia in Drosophila that affects gene expression.


Sujet(s)
Acétyl coenzyme A , Noyau de la cellule , Chromatine , Drosophila melanogaster , Animaux , Acetate coA-ligase/métabolisme , Acétyl coenzyme A/métabolisme , Acétylation , Transport biologique , Noyau de la cellule/génétique , Noyau de la cellule/métabolisme , Chromatine/métabolisme , Chromatine/génétique , Drosophila melanogaster/enzymologie , Drosophila melanogaster/génétique , Drosophila melanogaster/croissance et développement , Drosophila melanogaster/métabolisme , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Acides gras/composition chimique , Acides gras/métabolisme , Régulation de l'expression des gènes , Histone/composition chimique , Histone/métabolisme , Disques imaginaux/cytologie , Disques imaginaux/croissance et développement , Disques imaginaux/métabolisme , Lysine/métabolisme , Oxydoréduction , Ailes d'animaux/cytologie , Ailes d'animaux/croissance et développement , Ailes d'animaux/métabolisme
8.
Nat Commun ; 15(1): 4858, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38871712

RÉSUMÉ

Serpentinization, a geochemical process found on modern and ancient Earth, provides an ultra-reducing environment that can support microbial methanogenesis and acetogenesis. Several groups of archaea, such as the order Methanocellales, are characterized by their ability to produce methane. Here, we generate metagenomic sequences from serpentinized springs in The Cedars, California, and construct a circularized metagenome-assembled genome of a Methanocellales archaeon, termed Met12, that lacks essential methanogenesis genes. The genome includes genes for an acetyl-CoA pathway, but lacks genes encoding methanogenesis enzymes such as methyl-coenzyme M reductase, heterodisulfide reductases and hydrogenases. In situ transcriptomic analyses reveal high expression of a multi-heme c-type cytochrome, and heterologous expression of this protein in a model bacterium demonstrates that it is capable of accepting electrons. Our results suggest that Met12, within the order Methanocellales, is not a methanogen but a CO2-reducing, electron-fueled acetogen without electron bifurcation.


Sujet(s)
Méthane , Méthane/métabolisme , Génome d'archéobactérie , Protéines d'archée/métabolisme , Protéines d'archée/génétique , Oxidoreductases/génétique , Oxidoreductases/métabolisme , Métagénome/génétique , Phylogenèse , Acétyl coenzyme A/métabolisme , Dioxyde de carbone/métabolisme , Métagénomique
9.
Microb Biotechnol ; 17(6): e14514, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38923400

RÉSUMÉ

Pyruvate dehydrogenase (PDH) catalyses the irreversible decarboxylation of pyruvate to acetyl-CoA, which feeds the tricarboxylic acid cycle. We investigated how the loss of PDH affects metabolism in Pseudomonas putida. PDH inactivation resulted in a strain unable to utilize compounds whose assimilation converges at pyruvate, including sugars and several amino acids, whereas compounds that generate acetyl-CoA supported growth. PDH inactivation also resulted in the loss of carbon catabolite repression (CCR), which inhibits the assimilation of non-preferred compounds in the presence of other preferred compounds. Pseudomonas putida can degrade many aromatic compounds, most of which produce acetyl-CoA, making it useful for biotransformation and bioremediation. However, the genes involved in these metabolic pathways are often inhibited by CCR when glucose or amino acids are also present. Our results demonstrate that the PDH-null strain can efficiently degrade aromatic compounds even in the presence of other preferred substrates, which the wild-type strain does inefficiently, or not at all. As the loss of PDH limits the assimilation of many sugars and amino acids and relieves the CCR, the PDH-null strain could be useful in biotransformation or bioremediation processes that require growth with mixtures of preferred substrates and aromatic compounds.


Sujet(s)
Répression catabolique , Pseudomonas putida , Complexe du pyruvate déshydrogénase , Pseudomonas putida/génétique , Pseudomonas putida/métabolisme , Pseudomonas putida/enzymologie , Complexe du pyruvate déshydrogénase/métabolisme , Complexe du pyruvate déshydrogénase/génétique , Hydrocarbures aromatiques/métabolisme , Dépollution biologique de l'environnement , Acétyl coenzyme A/métabolisme , Acide pyruvique/métabolisme , Délétion de gène , Voies et réseaux métaboliques/génétique
10.
ACS Synth Biol ; 13(6): 1798-1808, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38748665

RÉSUMÉ

Betulinic acid (BA) is a lupane-type triterpenoid with potent anticancer and anti-HIV activities. Its great potential in clinical applications necessitates the development of an efficient strategy for BA synthesis. This study attempted to achieve efficient BA biosynthesis in Saccharomyces cerevisiae using systematic metabolic engineering strategies. First, a de novo BA biosynthesis pathway in S. cerevisiae was constructed, which yielded a titer of 14.01 ± 0.21 mg/L. Then, by enhancing the BA synthesis pathway and dynamic inhibition of the competitive pathway, a greater proportion of the metabolic flow was directed toward BA synthesis, achieving a titer of 88.07 ± 5.83 mg/L. Next, acetyl-CoA and NADPH supply was enhanced, which increased the BA titer to 166.43 ± 1.83 mg/L. Finally, another BA synthesis pathway in the peroxisome was constructed. Dual regulation of the peroxisome and cytoplasmic metabolism increased the BA titer to 210.88 ± 4.76 mg/L. Following fed-batch fermentation process modification, the BA titer reached 682.29 ± 8.16 mg/L. Overall, this work offers a guide for building microbial cell factories that are capable of producing terpenoids with efficiency.


Sujet(s)
Acide bétulinique , Génie métabolique , NADP , Triterpènes pentacycliques , Saccharomyces cerevisiae , Triterpènes , Génie métabolique/méthodes , Saccharomyces cerevisiae/métabolisme , Saccharomyces cerevisiae/génétique , Triterpènes pentacycliques/métabolisme , Triterpènes/métabolisme , NADP/métabolisme , Acétyl coenzyme A/métabolisme , Fermentation , Voies de biosynthèse/génétique
11.
Nat Commun ; 15(1): 4094, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750017

RÉSUMÉ

tRNA modifications affect ribosomal elongation speed and co-translational folding dynamics. The Elongator complex is responsible for introducing 5-carboxymethyl at wobble uridine bases (cm5U34) in eukaryotic tRNAs. However, the structure and function of human Elongator remain poorly understood. In this study, we present a series of cryo-EM structures of human ELP123 in complex with tRNA and cofactors at four different stages of the reaction. The structures at resolutions of up to 2.9 Å together with complementary functional analyses reveal the molecular mechanism of the modification reaction. Our results show that tRNA binding exposes a universally conserved uridine at position 33 (U33), which triggers acetyl-CoA hydrolysis. We identify a series of conserved residues that are crucial for the radical-based acetylation of U34 and profile the molecular effects of patient-derived mutations. Together, we provide the high-resolution view of human Elongator and reveal its detailed mechanism of action.


Sujet(s)
Cryomicroscopie électronique , ARN de transfert , Humains , ARN de transfert/métabolisme , ARN de transfert/composition chimique , ARN de transfert/génétique , Uridine/composition chimique , Uridine/métabolisme , Mutation , Acétyl coenzyme A/métabolisme , Acétyl coenzyme A/composition chimique , Modèles moléculaires , Acétylation , Histone acetyltransferases/métabolisme , Histone acetyltransferases/composition chimique , Histone acetyltransferases/génétique , Liaison aux protéines
12.
Nat Metab ; 6(5): 914-932, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38702440

RÉSUMÉ

Acetate, a precursor of acetyl-CoA, is instrumental in energy production, lipid synthesis and protein acetylation. However, whether acetate reprogrammes tumour metabolism and plays a role in tumour immune evasion remains unclear. Here, we show that acetate is the most abundant short-chain fatty acid in human non-small cell lung cancer tissues, with increased tumour-enriched acetate uptake. Acetate-derived acetyl-CoA induces c-Myc acetylation, which is mediated by the moonlighting function of the metabolic enzyme dihydrolipoamide S-acetyltransferase. Acetylated c-Myc increases its stability and subsequent transcription of the genes encoding programmed death-ligand 1, glycolytic enzymes, monocarboxylate transporter 1 and cell cycle accelerators. Dietary acetate supplementation promotes tumour growth and inhibits CD8+ T cell infiltration, whereas disruption of acetate uptake inhibits immune evasion, which increases the efficacy of anti-PD-1-based therapy. These findings highlight a critical role of acetate promoting tumour growth beyond its metabolic role as a carbon source by reprogramming tumour metabolism and immune evasion, and underscore the potential of controlling acetate metabolism to curb tumour growth and improve the response to immune checkpoint blockade therapy.


Sujet(s)
Acétates , Antigène CD274 , Protéines proto-oncogènes c-myc , Antigène CD274/métabolisme , Humains , Acétates/métabolisme , Acétates/pharmacologie , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Animaux , Souris , Échappement immunitaire , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/immunologie , Régulation positive , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Acétylation , Tumeurs du poumon/métabolisme , Tumeurs du poumon/immunologie , Acétyl coenzyme A/métabolisme , Échappement de la tumeur à la surveillance immunitaire
13.
J Agric Food Chem ; 72(15): 8704-8714, 2024 Apr 17.
Article de Anglais | MEDLINE | ID: mdl-38572931

RÉSUMÉ

Miltiradiene serves as a crucial precursor in the synthesis of various high-value abietane-type diterpenes, exhibiting diverse pharmacological activities. Previous efforts to enhance miltiradiene production have primarily focused on the mevalonate acetate (MVA) pathway. However, limited emphasis has been placed on optimizing the supply of acetyl-CoA and NADPH. In this study, we constructed a platform yeast strain for miltiradiene production by reinforcing the biosynthetic pathway of geranylgeranyl diphosphate (GGPP) and acetyl-CoA, and addressing the imbalance between the supply and demand of the redox cofactor NADPH within the cytoplasm, resulting in an increase in miltiradiene yield to 1.31 g/L. Furthermore, we conducted modifications to the miltiradiene synthase fusion protein tSmKSL1-CfTPS1. Finally, the comprehensive engineering strategies and protein modification strategies culminated in 1.43 g/L miltiradiene in the engineered yeast under shake flask culture conditions. Overall, our work established efficient yeast cell factories for miltiradiene production, providing a foothold for heterologous biosynthesis of abietane-type diterpenes.


Sujet(s)
Diterpènes , Saccharomyces cerevisiae , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Abiétanes , Acétyl coenzyme A/métabolisme , NADP/métabolisme , Diterpènes/métabolisme , Génie métabolique/méthodes
14.
Microb Cell Fact ; 23(1): 97, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38561811

RÉSUMÉ

BACKGROUND: Biotransformation of waste oil into value-added nutraceuticals provides a sustainable strategy. Thraustochytrids are heterotrophic marine protists and promising producers of omega (ω) fatty acids. Although the metabolic routes for the assimilation of hydrophilic carbon substrates such as glucose are known for these microbes, the mechanisms employed for the conversion of hydrophobic substrates are not well established. Here, thraustochytrid Schizochytrium limacinum SR21 was investigated for its ability to convert oils (commercial oils with varying fatty acid composition and waste cooking oil) into ω-3 fatty acid; docosahexaenoic acid (DHA). RESULTS: Within 72 h SR21 consumed ~ 90% of the oils resulting in enhanced biomass (7.5 g L- 1) which was 2-fold higher as compared to glucose. Statistical analysis highlights C16 fatty acids as important precursors of DHA biosynthesis. Transcriptomic data indicated the upregulation of multiple lipases, predicted to possess signal peptides for secretory, membrane-anchored and cytoplasmic localization. Additionally, transcripts encoding for mitochondrial and peroxisomal ß-oxidation along with acyl-carnitine transporters were abundant for oil substrates that allowed complete degradation of fatty acids to acetyl CoA. Further, low levels of oxidative biomarkers (H2O2, malondialdehyde) and antioxidants were determined for hydrophobic substrates, suggesting that SR21 efficiently mitigates the metabolic load and diverts the acetyl CoA towards energy generation and DHA accumulation. CONCLUSIONS: The findings of this study contribute to uncovering the route of assimilation of oil substrates by SR21. The thraustochytrid employs an intricate crosstalk among the extracellular and intracellular molecular machinery favoring energy generation. The conversion of hydrophobic substrates to DHA can be further improved using synthetic biology tools, thereby providing a unique platform for the sustainable recycling of waste oil substrates.


Sujet(s)
Acide docosahexaénoïque , Straménopiles , Acide docosahexaénoïque/métabolisme , Acétyl coenzyme A/métabolisme , Peroxyde d'hydrogène/métabolisme , Straménopiles/génétique , Acides gras/métabolisme , Biotransformation , Analyse de profil d'expression de gènes , Glucose/métabolisme
15.
Int Immunopharmacol ; 133: 112124, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38663312

RÉSUMÉ

The impaired osteogenic capability of bone marrow mesenchymal stem cells (BMSCs) caused by persistent inflammation is the main pathogenesis of inflammatory bone diseases. Recent studies show that metabolism is disturbed in osteogenically differentiated BMSCs in response to Lipopolysaccharide (LPS) treatment, while the mechanism involved remains incompletely revealed. Herein, we demonstrated that BMSCs adapted their metabolism to regulate acetyl-coenzyme A (acetyl-CoA) availability and RNA acetylation level, ultimately affecting osteogenic differentiation. The mitochondrial dysfunction and impaired osteogenic potential upon inflammatory conditions accompanied by the reduced acetyl-CoA content, which in turn suppressed N4-acetylation (ac4C) level. Supplying acetyl-CoA by sodium citrate (SC) addition rescued ac4C level and promoted the osteogenic capacity of LPS-treated cells through the ATP citrate lyase (ACLY) pathway. N-acetyltransferase 10 (NAT10) inhibitor remodelin reduced ac4C level and consequently impeded osteogenic capacity. Meanwhile, the osteo-promotive effect of acetyl-CoA-dependent ac4C might be attributed to fatty acid oxidation (FAO), as evidenced by activating FAO by L-carnitine supplementation counteracted remodelin-induced inhibition of osteogenesis. Further in vivo experiments confirmed the promotive role of acetyl-CoA in the endogenous bone regeneration in rat inflammatory mandibular defects. Our study uncovered a metabolic-epigenetic axis comprising acetyl-CoA and ac4C modification in the process of inflammatory osteogenesis of BMSCs and suggested a new target for bone tissue repair in the context of inflammatory bone diseases.


Sujet(s)
Acétyl coenzyme A , Différenciation cellulaire , Lipopolysaccharides , Cellules souches mésenchymateuses , Ostéogenèse , Animaux , Ostéogenèse/effets des médicaments et des substances chimiques , Acétyl coenzyme A/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Acétylation , Cellules cultivées , Rats , Mâle , Rat Sprague-Dawley , ATP citrate (pro-S)-lyase/métabolisme , Acetyltransferases/métabolisme , Acetyltransferases/génétique
16.
Cancer Lett ; 592: 216903, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38670307

RÉSUMÉ

High levels of acetyl-CoA are considered a key metabolic feature of metastatic cancers. However, the impacts of acetyl-CoA metabolic accumulation on cancer microenvironment remodeling are poorly understood. In this study, using human hepatocellular carcinoma (HCC) tissues and orthotopic xenograft models, we found a close association between high acetyl-CoA levels in HCCs, increased infiltration of tumor-associated neutrophils (TANs) in the cancer microenvironment and HCC metastasis. Cytokine microarray and enzyme-linked immunosorbent assays (ELISA) revealed the crucial role of the chemokine (C-X-C motif) ligand 1(CXCL1). Mechanistically, acetyl-CoA accumulation induces H3 acetylation-dependent upregulation of CXCL1 gene expression. CXCL1 recruits TANs, leads to neutrophil extracellular traps (NETs) formation and promotes HCC metastasis. Collectively, our work linked the accumulation of acetyl-CoA in HCC cells and TANs infiltration, and revealed that the CXCL1-CXC receptor 2 (CXCR2)-TANs-NETs axis is a potential target for HCCs with high acetyl-CoA levels.


Sujet(s)
Acétyl coenzyme A , Carcinome hépatocellulaire , Chimiokine CXCL1 , Tumeurs du foie , Granulocytes neutrophiles , Microenvironnement tumoral , Animaux , Femelle , Humains , Mâle , Souris , Acétyl coenzyme A/métabolisme , Acétylation , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/génétique , Lignée cellulaire tumorale , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Pièges extracellulaires/métabolisme , Régulation de l'expression des gènes tumoraux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Souris nude , Infiltration par les neutrophiles , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/anatomopathologie , Récepteurs à l'interleukine-8B/métabolisme , Récepteurs à l'interleukine-8B/génétique , Adulte , Adulte d'âge moyen , Sujet âgé , Souris de lignée BALB C
17.
mBio ; 15(5): e0341423, 2024 May 08.
Article de Anglais | MEDLINE | ID: mdl-38572988

RÉSUMÉ

Acetyl-CoA carboxylases (ACCs) convert acetyl-CoA to malonyl-CoA, a key step in fatty acid biosynthesis and autotrophic carbon fixation pathways. Three functionally distinct components, biotin carboxylase (BC), biotin carboxyl carrier protein (BCCP), and carboxyltransferase (CT), are either separated or partially fused in different combinations, forming heteromeric ACCs. However, an ACC with fused BC-BCCP and separate CT has not been identified, leaving its catalytic mechanism unclear. Here, we identify two BC isoforms (BC1 and BC2) from Chloroflexus aurantiacus, a filamentous anoxygenic phototroph that employs 3-hydroxypropionate (3-HP) bi-cycle rather than Calvin cycle for autotrophic carbon fixation. We reveal that BC1 possesses fused BC and BCCP domains, where BCCP could be biotinylated by E. coli or C. aurantiacus BirA on Lys553 residue. Crystal structures of BC1 and BC2 at 3.2 Å and 3.0 Å resolutions, respectively, further reveal a tetramer of two BC1-BC homodimers, and a BC2 homodimer, all exhibiting similar BC architectures. The two BC1-BC homodimers are connected by an eight-stranded ß-barrel of the partially resolved BCCP domain. Disruption of ß-barrel results in dissociation of the tetramer into dimers in solution and decreased biotin carboxylase activity. Biotinylation of the BCCP domain further promotes BC1 and CTß-CTα interactions to form an enzymatically active ACC, which converts acetyl-CoA to malonyl-CoA in vitro and produces 3-HP via co-expression with a recombinant malonyl-CoA reductase in E. coli cells. This study revealed a heteromeric ACC that evolves fused BC-BCCP but separate CTα and CTß to complete ACC activity.IMPORTANCEAcetyl-CoA carboxylase (ACC) catalyzes the rate-limiting step in fatty acid biosynthesis and autotrophic carbon fixation pathways across a wide range of organisms, making them attractive targets for drug discovery against various infections and diseases. Although structural studies on homomeric ACCs, which consist of a single protein with three subunits, have revealed the "swing domain model" where the biotin carboxyl carrier protein (BCCP) domain translocates between biotin carboxylase (BC) and carboxyltransferase (CT) active sites to facilitate the reaction, our understanding of the subunit composition and catalytic mechanism in heteromeric ACCs remains limited. Here, we identify a novel ACC from an ancient anoxygenic photosynthetic bacterium Chloroflexus aurantiacus, it evolves fused BC and BCCP domain, but separate CT components to form an enzymatically active ACC, which converts acetyl-CoA to malonyl-CoA in vitro and produces 3-hydroxypropionate (3-HP) via co-expression with recombinant malonyl-CoA reductase in E. coli cells. These findings expand the diversity and molecular evolution of heteromeric ACCs and provide a structural basis for potential applications in 3-HP biosynthesis.


Sujet(s)
Acetyl-coA carboxylase , Carbon-nitrogen ligases , Chloroflexus , Acetyl-coA carboxylase/métabolisme , Acetyl-coA carboxylase/génétique , Acetyl-coA carboxylase/composition chimique , Carbon-nitrogen ligases/métabolisme , Carbon-nitrogen ligases/génétique , Carbon-nitrogen ligases/composition chimique , Chloroflexus/génétique , Chloroflexus/métabolisme , Chloroflexus/enzymologie , Escherichia coli/génétique , Escherichia coli/métabolisme , Escherichia coli/enzymologie , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/composition chimique , Biotine/métabolisme , Biotine/biosynthèse , Malonyl coenzyme A/métabolisme , Acétyl coenzyme A/métabolisme , Protéines Escherichia coli/métabolisme , Protéines Escherichia coli/génétique , Protéines Escherichia coli/composition chimique , Fatty acid synthase type II
18.
Article de Anglais | MEDLINE | ID: mdl-38621758

RÉSUMÉ

Lycopene has been widely used in the food industry and medical field due to its antioxidant, anti-cancer, and anti-inflammatory properties. However, achieving efficient manufacture of lycopene using chassis cells on an industrial scale remains a major challenge. Herein, we attempted to integrate multiple metabolic engineering strategies to establish an efficient and balanced lycopene biosynthetic system in Saccharomyces cerevisiae. First, the lycopene synthesis pathway was modularized to sequentially enhance the metabolic flux of the mevalonate pathway, the acetyl-CoA supply module, and lycopene exogenous enzymatic module. The modular operation enabled the efficient conversion of acetyl-CoA to downstream pathway of lycopene synthesis, resulting in a 3.1-fold increase of lycopene yield. Second, we introduced acetate as an exogenous carbon source and utilized an acetate-repressible promoter to replace the natural ERG9 promoter. This approach not only enhanced the supply of acetyl-CoA but also concurrently diminished the flux toward the competitive ergosterol pathway. As a result, a further 42.3% increase in lycopene production was observed. Third, we optimized NADPH supply and mitigated cytotoxicity by overexpressing ABC transporters to promote lycopene efflux. The obtained strain YLY-PDR11 showed a 12.7-fold increase in extracellular lycopene level compared to the control strain. Finally, the total lycopene yield reached 343.7 mg/L, which was 4.3 times higher than that of the initial strain YLY-04. Our results demonstrate that combining multi-modular metabolic engineering with efflux engineering is an effective approach to improve the production of lycopene. This strategy can also be applied to the overproduction of other desirable isoprenoid compounds with similar synthesis and storage patterns in S. cerevisiae. ONE-SENTENCE SUMMARY: In this research, lycopene production in yeast was markedly enhanced by integrating a multi-modular approach, acetate signaling-based down-regulation of competitive pathways, and an efflux optimization strategy.


Sujet(s)
Acétyl coenzyme A , Caroténoïdes , Lycopène , Génie métabolique , Saccharomyces cerevisiae , Lycopène/métabolisme , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Génie métabolique/méthodes , Caroténoïdes/métabolisme , Acétyl coenzyme A/métabolisme , Acide mévalonique/métabolisme , Voies de biosynthèse , Régions promotrices (génétique) , NADP/métabolisme , Voies et réseaux métaboliques/génétique , Acétates/métabolisme
19.
Nat Commun ; 15(1): 3267, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38627361

RÉSUMÉ

In vitro biotransformation (ivBT) facilitated by in vitro synthetic enzymatic biosystems (ivSEBs) has emerged as a highly promising biosynthetic platform. Several ivSEBs have been constructed to produce poly-3-hydroxybutyrate (PHB) via acetyl-coenzyme A (acetyl-CoA). However, some systems are hindered by their reliance on costly ATP, limiting their practicality. This study presents the design of an ATP-free ivSEB for one-pot PHB biosynthesis via acetyl-CoA utilizing starch-derived maltodextrin as the sole substrate. Stoichiometric analysis indicates this ivSEB can self-maintain NADP+/NADPH balance and achieve a theoretical molar yield of 133.3%. Leveraging simple one-pot reactions, our ivSEBs achieved a near-theoretical molar yield of 125.5%, the highest PHB titer (208.3 mM, approximately 17.9 g/L) and the fastest PHB production rate (9.4 mM/h, approximately 0.8 g/L/h) among all the reported ivSEBs to date, and demonstrated easy scalability. This study unveils the promising potential of ivBT for the industrial-scale production of PHB and other acetyl-CoA-derived chemicals from starch.


Sujet(s)
Hydroxy-butyrates , , Polyosides , Amidon , Acétyl coenzyme A/métabolisme , Amidon/métabolisme , Hydroxy-butyrates/métabolisme , Polyesters/métabolisme , NADP/métabolisme , Biotransformation
20.
Proc Natl Acad Sci U S A ; 121(13): e2318969121, 2024 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-38513105

RÉSUMÉ

Autotrophic theories for the origin of metabolism posit that the first cells satisfied their carbon needs from CO2 and were chemolithoautotrophs that obtained their energy and electrons from H2. The acetyl-CoA pathway of CO2 fixation is central to that view because of its antiquity: Among known CO2 fixing pathways it is the only one that is i) exergonic, ii) occurs in both bacteria and archaea, and iii) can be functionally replaced in full by single transition metal catalysts in vitro. In order to operate in cells at a pH close to 7, however, the acetyl-CoA pathway requires complex multi-enzyme systems capable of flavin-based electron bifurcation that reduce low potential ferredoxin-the physiological donor of electrons in the acetyl-CoA pathway-with electrons from H2. How can the acetyl-CoA pathway be primordial if it requires flavin-based electron bifurcation? Here, we show that native iron (Fe0), but not Ni0, Co0, Mo0, NiFe, Ni2Fe, Ni3Fe, or Fe3O4, promotes the H2-dependent reduction of aqueous Clostridium pasteurianum ferredoxin at pH 8.5 or higher within a few hours at 40 °C, providing the physiological function of flavin-based electron bifurcation, but without the help of enzymes or organic redox cofactors. H2-dependent ferredoxin reduction by iron ties primordial ferredoxin reduction and early metabolic evolution to a chemical process in the Earth's crust promoted by solid-state iron, a metal that is still deposited in serpentinizing hydrothermal vents today.


Sujet(s)
Ferrédoxines , Fer , Ferrédoxines/métabolisme , Fer/métabolisme , Hydrogène/métabolisme , Électrons , Acétyl coenzyme A/métabolisme , Dioxyde de carbone/métabolisme , Oxydoréduction , Flavines/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...