Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Metab Eng ; 79: 192-202, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37611820

RESUMEN

(2S)-Naringenin is a key precursor for biosynthesis of various high-value flavonoids and possesses a variety of nutritional and pharmaceutical properties on human health. Systematic optimization approaches have been employed to improve (2S)-naringenin production in different microbial hosts. However, very few studies have focused on the spatiotemporal distribution of (2S)-naringenin and the related pathway intermediate p-coumaric acid, which is an important factor for efficient production. Here, we first optimized the (2S)-naringenin biosynthetic pathway by alleviating the bottleneck downstream of p-coumaric acid and increasing malonyl-CoA supply, which improved (2S)-naringenin production but significant accumulation of p-coumaric acid still existed extracellularly. We thus established a dual dynamic control system through combining a malonyl-CoA biosensor regulator and an RNAi strategy, to autonomously control the synthesis of p-coumaric acid with the supply of malonyl-CoA. Furthermore, screening potential transporters led to identification of Pdr12 for improved (2S)-naringenin production and reduced accumulation of p-coumaric acid. Finally, a titer of 2.05 g/L (2S)-naringenin with negligible accumulation of p-coumaric acid was achieved in a fed batch fermentation. Our work highlights the importance of systematic control of pathway intermediates for efficient microbial production of plant natural products.


Asunto(s)
Flavanonas , Saccharomyces cerevisiae , Humanos , Ácidos Cumáricos , Malonil Coenzima A/genética
2.
Metab Eng ; 75: 143-152, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36549411

RESUMEN

Flavonoids are a group of secondary metabolites from plants that have received attention as high value-added pharmacological substances. Recently, a robust and efficient bioprocess using recombinant microbes has emerged as a promising approach to supply flavonoids. In the flavonoid biosynthetic pathway, the rate of chalcone synthesis, the first committed step, is a major bottleneck. However, chalcone synthase (CHS) engineering was difficult because of high-level conservation and the absence of effective screening tools, which are limited to overexpression or homolog-based combinatorial strategies. Furthermore, it is necessary to precisely regulate the metabolic flux for the optimum availability of malonyl-CoA, a substrate of chalcone synthesis. In this study, we engineered CHS and optimized malonyl-CoA availability to establish a platform strain for naringenin production, a key molecular scaffold for various flavonoids. First, we engineered CHS through synthetic riboswitch-based high-throughput screening of rationally designed mutant libraries. Consequently, the catalytic efficiency (kcat/Km) of the optimized CHS enzyme was 62% higher than that of the wild-type enzyme. In addition to CHS engineering, we designed genetic circuits using transcriptional repressors to fine-tune the malonyl-CoA availability. The best mutant with synergistic effects of the engineered CHS and the optimized genetic circuit produced 98.71 mg/L naringenin (12.57 mg naringenin/g glycerol), which is the highest naringenin concentration and yield from glycerol in similar culture conditions reported to date, a 2.5-fold increase compared to the parental strain. Overall, this study provides an effective strategy for efficient production of flavonoids.


Asunto(s)
Chalconas , Flavanonas , Riboswitch , Flavonoides/genética , Glicerol , Flavanonas/genética , Malonil Coenzima A/genética , Malonil Coenzima A/metabolismo , Ingeniería Metabólica
3.
Metab Eng ; 73: 1-10, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35643281

RESUMEN

Malonate is a platform chemical that has been utilized to synthesize many valuable chemical compounds. Here, Saccharomyces cerevisiae was metabolically engineered to produce malonate through the malonyl-CoA pathway. To construct the key step of converting malonyl-CoA to malonate, a native mitochondrial 3-hydroxyisobutyryl-CoA hydrolase gene EHD3 was mutated to target the cytoplasm and obtain malonyl-CoA hydrolase activity. The malonyl-CoA hydrolase activity of Ehd3 was achieved by mutating the malonyl-CoA binding site F121 to I121 and the active site E124 to seven amino acids (S/T/H/K/R/N/Q). We identified that the strain with E124S mutation had the highest malonate titer with 13.6 mg/L. Genomic integration of the mutant EHD3 and ACC1** to delta sequence sites was further explored to increase their reliable expression. Accordingly, a screening method with the work flow of fluorescence detection, shake-tube fermentation, and shake-flask fermentation was constructed to screen high copy delta sequences efficiently. The malonate titer was improved to 73.55 mg/L after screening the ∼1500 integrative strains, which was increased 4.4-folds than that of the episomal strain. We further engineered the strain by regulating the expression of key enzyme in the malonyl-CoA pathway to improve the precursor supply and inhibiting its competing pathways, and the final engineered strain LMA-16 produced 187.25 mg/L in the flask, 14-fold compared with the initial episomal expression strain. Finally, the combined efforts increased the malonate titer to 1.62 g/L in fed-batch fermentation.


Asunto(s)
Hidrolasas , Malonatos , Malonil Coenzima A , Ingeniería Metabólica , Saccharomyces cerevisiae , Fermentación , Hidrolasas/genética , Hidrolasas/metabolismo , Malonatos/metabolismo , Malonil Coenzima A/genética , Malonil Coenzima A/metabolismo , Ingeniería Metabólica/métodos , Mutación , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
4.
J Exp Bot ; 72(4): 1349-1369, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33130852

RESUMEN

Malonyl-CoA:flavonoid acyltransferases (MaTs) modify isoflavones, but only a few have been characterized for activity and assigned to specific physiological processes. Legume roots exude isoflavone malonates into the rhizosphere, where they are hydrolyzed into isoflavone aglycones. Soybean GmMaT2 was highly expressed in seeds, root hairs, and nodules. GmMaT2 and GmMaT4 recombinant enzymes used isoflavone 7-O-glucosides as acceptors and malonyl-CoA as an acyl donor to generate isoflavone glucoside malonates. GmMaT2 had higher activity towards isoflavone glucosides than GmMaT4. Overexpression in hairy roots of GmMaT2 and GmMaT4 produced more malonyldaidzin, malonylgenistin, and malonylglycitin, and resulted in more nodules than control. However, only GmMaT2 knockdown (KD) hairy roots showed reduced levels of malonyldaidzin, malonylgenistin, and malonylglycitin, and, likewise, reduced nodule numbers. These were consistent with the up-regulation of only GmMaT2 by rhizobial infection, and higher expression levels of early nodulation genes in GmMaT2- and GmMaT4-overexpressing roots, but lower only in GmMaT2-KD roots compared with control roots. Higher malonyl isoflavonoid levels in transgenic hairy roots were associated with higher levels of isoflavones in root exudates and more nodules, and vice versa. We suggest that GmMaT2 participates in soybean nodulation by catalyzing isoflavone malonylation and affecting malonyl isoflavone secretion for activation of Nod factor and nodulation.


Asunto(s)
Aciltransferasas/fisiología , Glycine max , Isoflavonas , Malonil Coenzima A/fisiología , Nodulación de la Raíz de la Planta , Aciltransferasas/genética , Malonil Coenzima A/genética , Glycine max/enzimología , Glycine max/genética
5.
Int J Mol Sci ; 22(23)2021 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-34884438

RESUMEN

Malonic aciduria is an extremely rare inborn error of metabolism due to malonyl-CoA decarboxylase deficiency. This enzyme is encoded by the MLYCD (Malonyl-CoA Decarboxylase) gene, and the disease has an autosomal recessive inheritance. Malonic aciduria is characterized by systemic clinical involvement, including neurologic and digestive symptoms, metabolic acidosis, hypoglycemia, failure to thrive, seizures, developmental delay, and cardiomyopathy. We describe here two index cases belonging to the same family that, despite an identical genotype, present very different clinical pictures. The first case is a boy with neonatal metabolic symptoms, abnormal brain MRI, and dilated cardiomyopathy. The second case, the cousin of the first patient in a consanguineous family, showed later symptoms, mainly with developmental delay. Both patients showed high levels of malonylcarnitine on acylcarnitine profiles and malonic acid on urinary organic acid chromatographies. The same homozygous pathogenic variant was identified, c.346C > T; p. (Gln116*). We also provide a comprehensive literature review of reported cases. A review of the literature yielded 52 cases described since 1984. The most common signs were developmental delay and cardiomyopathy. Increased levels of malonic acid and malonylcarnitine were constant. Presentations ranged from neonatal death to patients surviving past adolescence. These two cases and reported patients in the literature highlight the inter- and intrafamilial variability of malonic aciduria.


Asunto(s)
Carboxiliasas/deficiencia , Errores Innatos del Metabolismo/genética , Mutación Puntual , Carboxiliasas/genética , Carnitina/análogos & derivados , Carnitina/análisis , Preescolar , Consanguinidad , Homocigoto , Humanos , Masculino , Malonatos/orina , Malonil Coenzima A/genética , Ácido Metilmalónico , Linaje
6.
Metab Eng ; 61: 79-88, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32445959

RESUMEN

Metabolic addiction, an organism that is metabolically addicted with a compound to maintain its growth fitness, is an underexplored area in metabolic engineering. Microbes with heavily engineered pathways or genetic circuits tend to experience metabolic burden leading to degenerated or abortive production phenotype during long-term cultivation or scale-up. A promising solution to combat metabolic instability is to tie up the end-product with an intermediary metabolite that is essential to the growth of the producing host. Here we present a simple strategy to improve both metabolic stability and pathway yield by coupling chemical addiction with negative autoregulatory genetic circuits. Naringenin and lipids compete for the same precursor malonyl-CoA with inversed pathway yield in oleaginous yeast. Negative autoregulation of the lipogenic pathways, enabled by CRISPRi and fatty acid-inducible promoters, repartitions malonyl-CoA to favor flavonoid synthesis and increased naringenin production by 74.8%. With flavonoid-sensing transcriptional activator FdeR and yeast hybrid promoters to control leucine synthesis and cell grwoth fitness, this amino acid feedforward metabolic circuit confers a flavonoid addiction phenotype that selectively enrich the naringenin-producing pupulation in the leucine auxotrophic yeast. The engineered yeast persisted 90.9% of naringenin titer up to 324 generations. Cells without flavonoid addiction regained growth fitness but lost 94.5% of the naringenin titer after cell passage beyond 300 generations. Metabolic addiction and negative autoregulation may be generalized as basic tools to eliminate metabolic heterogeneity, improve strain stability and pathway yield in long-term and large-scale bioproduction.


Asunto(s)
Homeostasis , Ingeniería Metabólica , Yarrowia , Malonil Coenzima A/genética , Malonil Coenzima A/metabolismo , Yarrowia/genética , Yarrowia/metabolismo
7.
Biotechnol Bioeng ; 116(6): 1380-1391, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30684355

RESUMEN

In recent years microorganisms have been engineered towards synthesizing interesting plant polyphenols such as flavonoids and stilbenes from glucose. Currently, the low endogenous supply of malonyl-CoA, indispensable for plant polyphenol synthesis, impedes high product titers. Usually, limited malonyl-CoA availability during plant polyphenol production is avoided by supplementing fatty acid synthesis-inhibiting antibiotics such as cerulenin, which are known to increase the intracellular malonyl-CoA pool as a side effect. Motivated by the goal of microbial polyphenol synthesis being independent of such expensive additives, we used rational metabolic engineering approaches to modulate regulation of fatty acid synthesis and flux into the tricarboxylic acid cycle (TCA cycle) in Corynebacterium glutamicum strains capable of flavonoid and stilbene synthesis. Initial experiments showed that sole overexpression of genes coding for the native malonyl-CoA-forming acetyl-CoA carboxylase is not sufficient for increasing polyphenol production in C. glutamicum. Hence, the intracellular acetyl-CoA availability was also increased by reducing the flux into the TCA cycle through reduction of citrate synthase activity. In defined cultivation medium, the constructed C. glutamicum strains accumulated 24 mg·L -1 (0.088 mM) naringenin or 112 mg·L -1 (0.49 mM) resveratrol from glucose without supplementation of phenylpropanoid precursor molecules or any inhibitors of fatty acid synthesis.


Asunto(s)
Corynebacterium glutamicum , Malonil Coenzima A , Ingeniería Metabólica/métodos , Fitoquímicos , Polifenoles , Reactores Biológicos , Citrato (si)-Sintasa/metabolismo , Ciclo del Ácido Cítrico/genética , Corynebacterium glutamicum/genética , Corynebacterium glutamicum/metabolismo , Flavanonas , Malonil Coenzima A/análisis , Malonil Coenzima A/genética , Malonil Coenzima A/metabolismo , Fitoquímicos/análisis , Fitoquímicos/metabolismo , Polifenoles/análisis , Polifenoles/metabolismo , Resveratrol
8.
Int J Mol Sci ; 20(6)2019 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-30897730

RESUMEN

Fibroblast growth factor 21 (FGF-21) is known to be a biomarker for mitochondrial disorders. An upregulation of FGF-21 in serum and muscle of carnitine palmitoyltransferase I (CPT I) and carnitine palmitoyltransferase II (CPT II) knock-out mice has been reported. In human CPT II deficiency, enzyme activity and protein content are normal, but the enzyme is abnormally regulated by malonyl-CoA and is abnormally thermolabile. Citrate synthase (CS) activity is increased in patients with CPT II deficiency. This may indicate a compensatory response to an impaired function of CPT II. In this study, FGF-21 serum levels in patients with CPT II deficiency during attack free intervals and in healthy controls were measured by enzyme linked immunosorbent assay (ELISA). The data showed no significant difference between FGF-21 concentration in the serum of patients with CPT II deficiency and that in the healthy controls. The results of the present work support the hypothesis that in muscle CPT II deficiency, in contrast to the mouse knockout model, mitochondrial fatty acid utilization is not persistently reduced. Thus, FGF-21 does not seem to be a useful biomarker in the diagnosis of CPT II deficiency.


Asunto(s)
Carnitina O-Palmitoiltransferasa/sangre , Carnitina O-Palmitoiltransferasa/deficiencia , Factores de Crecimiento de Fibroblastos/sangre , Errores Innatos del Metabolismo/sangre , Enfermedades Mitocondriales/sangre , Adulto , Animales , Biomarcadores/sangre , Carnitina O-Palmitoiltransferasa/genética , Citrato (si)-Sintasa/genética , Citrato (si)-Sintasa/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Malonil Coenzima A/genética , Malonil Coenzima A/metabolismo , Errores Innatos del Metabolismo/genética , Ratones , Ratones Noqueados , Persona de Mediana Edad , Enfermedades Mitocondriales/genética
9.
Plant Cell Physiol ; 59(7): 1398-1414, 2018 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-29660070

RESUMEN

Drynaria roosii (Nakaike) is a traditional Chinese medicinal fern, known as 'GuSuiBu'. The effective components, naringin and neoeriocitrin, share a highly similar chemical structure and medicinal function. Our HPLC-tandem mass spectrometry (MS/MS) results showed that the accumulation of naringin/neoeriocitrin depended on specific tissues or ages. However, little was known about the expression patterns of naringin/neoeriocitrin-related genes involved in their regulatory pathways. Due to a lack of basic genetic information, we applied a combination of single molecule real-time (SMRT) sequencing and second-generation sequencing (SGS) to generate the complete and full-length transcriptome of D. roosii. According to the SGS data, the differentially expressed gene (DEG)-based heat map analysis revealed that naringin/neoeriocitrin-related gene expression exhibited obvious tissue- and time-specific transcriptomic differences. Using the systems biology method of modular organization analysis, we clustered 16,472 DEGs into 17 gene modules and studied the relationships between modules and tissue/time point samples, as well as modules and naringin/neoeriocitrin contents. We found that naringin/neoeriocitrin-related DEGs distributed in nine distinct modules, and DEGs in these modules showed significantly different patterns of transcript abundance to be linked to specific tissues or ages. Moreover, weighted gene co-expression network analysis (WGCNA) results further identified that PAL, 4CL and C4H, and C3H and HCT acted as the major hub genes involved in naringin and neoeriocitrin synthesis, respectively, and exhibited high co-expression with MYB- and basic helix-leucine-helix (bHLH)-regulated genes. In this work, modular organization and co-expression networks elucidated the tissue and time specificity of the gene expression pattern, as well as hub genes associated with naringin/neoeriocitrin synthesis in D. roosii. Simultaneously, the comprehensive transcriptome data set provided important genetic information for further research on D. roosii.


Asunto(s)
Disacáridos/genética , Flavanonas/genética , Regulación de la Expresión Génica de las Plantas , Polypodiaceae/genética , Cromatografía Líquida de Alta Presión , Disacáridos/metabolismo , Flavanonas/metabolismo , Redes Reguladoras de Genes , Malonil Coenzima A/genética , Malonil Coenzima A/metabolismo , Fenilalanina/genética , Fenilalanina/metabolismo , Proteínas de Plantas/genética , Polypodiaceae/metabolismo , Análisis de Secuencia de ARN/métodos , Espectrometría de Masas en Tándem , Factores de Transcripción/genética , Transcriptoma
10.
Biochem J ; 474(22): 3783-3797, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-28986507

RESUMEN

Mitochondrial fatty acid synthesis (mtFAS) is a highly conserved pathway essential for mitochondrial biogenesis. The mtFAS process is required for mitochondrial respiratory chain assembly and function, synthesis of the lipoic acid cofactor indispensable for the function of several mitochondrial enzyme complexes and essential for embryonic development in mice. Mutations in human mtFAS have been reported to lead to neurodegenerative disease. The source of malonyl-CoA for mtFAS in mammals has remained unclear. We report the identification of a conserved vertebrate mitochondrial isoform of ACC1 expressed from an ACACA transcript splicing variant. A specific knockdown (KD) of the corresponding transcript in mouse cells, or CRISPR/Cas9-mediated inactivation of the putative mitochondrial targeting sequence in human cells, leads to decreased lipoylation and mitochondrial fragmentation. Simultaneous KD of ACSF3, encoding a mitochondrial malonyl-CoA synthetase previously implicated in the mtFAS process, resulted in almost complete ablation of protein lipoylation, indicating that these enzymes have a redundant function in mtFAS. The discovery of a mitochondrial isoform of ACC1 required for lipoic acid synthesis has intriguing consequences for our understanding of mitochondrial disorders, metabolic regulation of mitochondrial biogenesis and cancer.


Asunto(s)
Acetil-CoA Carboxilasa/metabolismo , Coenzima A Ligasas/metabolismo , Malonil Coenzima A/metabolismo , Mitocondrias/patología , Acetil-CoA Carboxilasa/genética , Secuencia de Aminoácidos , Animales , Línea Celular , Coenzima A Ligasas/genética , Secuencia Conservada , Regulación Enzimológica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Isoenzimas , Malonil Coenzima A/genética , Ratones , Mitocondrias/enzimología , ARN Interferente Pequeño , Ácido Tióctico
11.
J Biol Chem ; 291(48): 25154-25166, 2016 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-27733682

RESUMEN

AMP-activated protein kinase (AMPK) is an energy sensor and master regulator of metabolism. AMPK functions as a fuel gauge monitoring systemic and cellular energy status. Activation of AMPK occurs when the intracellular AMP/ATP ratio increases and leads to a metabolic switch from anabolism to catabolism. AMPK phosphorylates and inhibits acetyl-CoA carboxylase (ACC), which catalyzes carboxylation of acetyl-CoA to malonyl-CoA, the first and rate-limiting reaction in de novo synthesis of fatty acids. AMPK thus regulates homeostasis of acetyl-CoA, a key metabolite at the crossroads of metabolism, signaling, chromatin structure, and transcription. Nucleocytosolic concentration of acetyl-CoA affects histone acetylation and links metabolism and chromatin structure. Here we show that activation of AMPK with the widely used antidiabetic drug metformin or with the AMP mimetic 5-aminoimidazole-4-carboxamide ribonucleotide increases the inhibitory phosphorylation of ACC and decreases the conversion of acetyl-CoA to malonyl-CoA, leading to increased protein acetylation and altered gene expression in prostate and ovarian cancer cells. Direct inhibition of ACC with allosteric inhibitor 5-(tetradecyloxy)-2-furoic acid also increases acetylation of histones and non-histone proteins. Because AMPK activation requires liver kinase B1, metformin does not induce protein acetylation in liver kinase B1-deficient cells. Together, our data indicate that AMPK regulates the availability of nucleocytosolic acetyl-CoA for protein acetylation and that AMPK activators, such as metformin, have the capacity to increase protein acetylation and alter patterns of gene expression, further expanding the plethora of metformin's physiological effects.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Metformina/farmacología , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias de la Próstata/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/genética , Acetilcoenzima A/genética , Acetilcoenzima A/metabolismo , Acetilación/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Células HeLa , Humanos , Masculino , Malonil Coenzima A/genética , Malonil Coenzima A/metabolismo , Proteínas de Neoplasias/genética , Neoplasias Ováricas/genética , Neoplasias de la Próstata/genética , Procesamiento Proteico-Postraduccional/genética
12.
Metab Eng ; 44: 253-264, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29097310

RESUMEN

Malonyl-CoA is the basic building block for synthesizing a range of important compounds including fatty acids, phenylpropanoids, flavonoids and non-ribosomal polyketides. Centering around malonyl-CoA, we summarized here the various metabolic engineering strategies employed recently to regulate and control malonyl-CoA metabolism and improve cellular productivity. Effective metabolic engineering of microorganisms requires the introduction of heterologous pathways and dynamically rerouting metabolic flux towards products of interest. Transcriptional factor-based biosensors translate an internal cellular signal to a transcriptional output and drive the expression of the designed genetic/biomolecular circuits to compensate the activity loss of the engineered biosystem. Recent development of genetically-encoded malonyl-CoA sensor has stood out as a classical example to dynamically reprogram cell metabolism for various biotechnological applications. Here, we reviewed the design principles of constructing a transcriptional factor-based malonyl-CoA sensor with superior detection limit, high sensitivity and broad dynamic range. We discussed various synthetic biology strategies to remove pathway bottleneck and how genetically-encoded metabolite sensor could be deployed to improve pathway efficiency. Particularly, we emphasized that integration of malonyl-CoA sensing capability with biocatalytic function would be critical to engineer efficient microbial cell factory. Biosensors have also advanced beyond its classical function of a sensor actuator for in situ monitoring of intracellular metabolite concentration. Applications of malonyl-CoA biosensors as a sensor-invertor for negative feedback regulation of metabolic flux, a metabolic switch for oscillatory balancing of malonyl-CoA sink pathway and source pathway and a screening tool for engineering more efficient biocatalyst are also presented in this review. We envision the genetically-encoded malonyl-CoA sensor will be an indispensable tool to optimize cell metabolism and cost-competitively manufacture malonyl-CoA-derived compounds.


Asunto(s)
Técnicas Biosensibles/métodos , Malonil Coenzima A/análisis , Ingeniería Metabólica/métodos , Microorganismos Modificados Genéticamente , Malonil Coenzima A/genética , Malonil Coenzima A/metabolismo , Microorganismos Modificados Genéticamente/genética , Microorganismos Modificados Genéticamente/metabolismo
13.
Mol Cell Proteomics ; 14(11): 3056-71, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26320211

RESUMEN

The protein substrates of sirtuin 5-regulated lysine malonylation (Kmal) remain unknown, hindering its functional analysis. In this study, we carried out proteomic screening, which identified 4042 Kmal sites on 1426 proteins in mouse liver and 4943 Kmal sites on 1822 proteins in human fibroblasts. Increased malonyl-CoA levels in malonyl-CoA decarboxylase (MCD)-deficient cells induces Kmal levels in substrate proteins. We identified 461 Kmal sites showing more than a 2-fold increase in response to MCD deficiency as well as 1452 Kmal sites detected only in MCD-/- fibroblast but not MCD+/+ cells, suggesting a pathogenic role of Kmal in MCD deficiency. Cells with increased lysine malonylation displayed impaired mitochondrial function and fatty acid oxidation, suggesting that lysine malonylation plays a role in pathophysiology of malonic aciduria. Our study establishes an association between Kmal and a genetic disease and offers a rich resource for elucidating the contribution of the Kmal pathway and malonyl-CoA to cellular physiology and human diseases.


Asunto(s)
Carboxiliasas/deficiencia , Hígado/metabolismo , Lisina/metabolismo , Malonatos/metabolismo , Errores Innatos del Metabolismo/metabolismo , Mitocondrias/metabolismo , Animales , Carboxiliasas/genética , Carboxiliasas/metabolismo , Línea Celular , Ácidos Grasos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Hígado/patología , Masculino , Malonil Coenzima A/genética , Malonil Coenzima A/metabolismo , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/patología , Ácido Metilmalónico/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/patología , Modelos Moleculares , Oxidación-Reducción , Sirtuinas/deficiencia , Sirtuinas/genética
14.
Am J Med Genet A ; 170A(5): 1347-51, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26858006

RESUMEN

Malonyl-CoA decarboxylase deficiency is an extremely rare autosomal recessive inborn error of fatty acid metabolism. It usually follows a severe disease course and presents poor prognosis without treatment. Here, we report an affected female juvenile with a mild clinical and biochemical phenotype who mainly featured poor schooling without cardiomyopathy and metabolic acidosis. She was suspected of malonyl-CoA decarboxylase deficiency due to a 57-kb deletion in 16q23.3 encompassing the MLCYD gene revealed by chromosome microarray. Malonyl-CoA decarboxylase deficiency was then confirmed by acylcarnitine analysis and organic acid analysis. Real-time PCR analysis of the patient revealed the first three exon deletion of the MLYCD gene, which was maternally inherited. DNA sequencing of the MLYCD gene of the patient identified a novel heterozygous mutation (c.911G>A, p.G304E) in exon 4 that was paternally inherited. The patient urine malonic acid dissolved and had a better school record in 6 month after initiation of fat-limited diet. At 1 year post treatment, the blood malonylcarnitine level decreased remarkably. Our result expands the phenotype of malonyl-CoA decarboxylase deficiency and suggests attentions should be paid to the mild form of disorders, for example, malonyl-CoA decarboxylase deficiency, which usually present a severe disease course.


Asunto(s)
Acidosis/genética , Carboxiliasas/deficiencia , Errores Innatos del Metabolismo/genética , Acidosis/fisiopatología , Adolescente , Secuencia de Bases , Carboxiliasas/genética , Cardiomiopatías/genética , Cardiomiopatías/fisiopatología , Niño , Cromosomas/genética , Exones , Femenino , Humanos , Malonatos/metabolismo , Malonil Coenzima A/genética , Errores Innatos del Metabolismo/fisiopatología , Ácido Metilmalónico , Análisis por Micromatrices , Eliminación de Secuencia
15.
PLoS Pathog ; 9(5): e1003333, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23696731

RESUMEN

Human cytomegalovirus hijacks host cell metabolism, increasing the flux of carbon from glucose to malonyl-CoA, the committed precursor to fatty acid synthesis and elongation. Inhibition of acetyl-CoA carboxylase blocks the production of progeny virus. To probe further the role of fatty acid metabolism during infection, we performed an siRNA screen to identify host cell metabolic enzymes needed for the production of infectious cytomegalovirus progeny. The screen predicted that multiple long chain acyl-CoA synthetases and fatty acid elongases are needed during infection, and the levels of RNAs encoding several of these enzymes were upregulated by the virus. Roles for acyl-CoA synthetases and elongases during infection were confirmed by using small molecule antagonists. Consistent with a role for these enzymes, mass spectrometry-based fatty acid analysis with ¹³C-labeling revealed that malonyl-CoA is consumed by elongases to produce very long chain fatty acids, generating an approximately 8-fold increase in C26-C34 fatty acid tails in infected cells. The virion envelope was yet further enriched in C26-C34 saturated fatty acids, and elongase inhibitors caused the production of virions with lower levels of these fatty acids and markedly reduced infectivity. These results reveal a dependence of cytomegalovirus on very long chain fatty acid metabolism.


Asunto(s)
Coenzima A Ligasas/metabolismo , Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/metabolismo , Ácidos Grasos/biosíntesis , Malonil Coenzima A/metabolismo , Virión/metabolismo , Línea Celular , Coenzima A Ligasas/genética , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Ácidos Grasos/genética , Humanos , Malonil Coenzima A/genética , ARN Interferente Pequeño , Virión/genética
16.
Metab Eng ; 29: 217-226, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25863265

RESUMEN

Malonyl-CoA is the building block for fatty acid biosynthesis and also a precursor to various pharmaceutically and industrially valuable molecules, such as polyketides and biopolymers. However, intracellular malonyl-CoA is usually maintained at low levels, which poses great challenges to efficient microbial production of malonyl-CoA derived molecules. Inactivation of the malonyl-CoA consumption pathway to increase its intracellular availability is not applicable, since it is usually lethal to microorganisms. In this work, we employ synthetic antisense RNAs (asRNAs) to conditionally down-regulate fatty acid biosynthesis and achieve malonyl-CoA enrichment in Escherichia coli. The optimized asRNA constructs with a loop-stem structure exhibit high interference efficiency up to 80%, leading to a 4.5-fold increase in intracellular malonyl-CoA concentration when fabD gene expression is inhibited. Strikingly, this strategy allows the improved production of natural products 4-hydroxycoumarin, resveratrol, and naringenin by 2.53-, 1.70-, and 1.53-fold in E. coli, respectively. In addition, down-regulation of other fab genes including fabH, fabB, and fabF also leads to remarkable increases in 4-hydroxycoumarin production. This study demonstrates a novel strategy to enhance intracellular malonyl-CoA and indicates the effectiveness of asRNA as a powerful tool for use in metabolic engineering.


Asunto(s)
S-Maloniltransferasa de la Proteína Transportadora de Grupos Acilo/biosíntesis , Proteínas de Escherichia coli/biosíntesis , Regulación Bacteriana de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Malonil Coenzima A , ARN sin Sentido , Acido Graso Sintasa Tipo II/biosíntesis , Malonil Coenzima A/genética , Malonil Coenzima A/metabolismo , ARN sin Sentido/biosíntesis , ARN sin Sentido/genética
17.
Am J Med Genet A ; 167A(10): 2300-5, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25982642

RESUMEN

Methyl malonic academia (MMA) is characterized by abnormal accumulation of methyl malonic acid in body fluids. Patients usually have a variety of clinical symptoms including recurrent vomiting, metabolic acidosis, developmental delay, seizure, or death. However, a few cases where the patients have no symptom are also reported. Here, we conducted clinical, biochemical, and molecular analysis of eight Chinese patients identified through newborn screening between 2003 and 2013. All the patients had significantly higher blood propionylcarnitine (C3) concentrations, ratio of propionylcarnitine/acetylcarnitine (C3/C2); and their urine methyl malonic acid and methylcitric acid (MCA) excretions were remarkably higher than normal at diagnosis and during follow-ups. In addition, five different known mutations were identified in seven of the eight patients in either MUT or MMACHC. All these mutations were expected to produce defective proteins that would result in decreased or even total loss of methyl malonyl-CoA mutase activity. However, normal outcomes were found in all patients in physical growth, intellectual performance and cerebral MRI analysis at diagnosis (range, 14-53 days) and during follow-ups (range, 1.8-10 years). Our study is the first report of Chinese MMA patients with increased secretion of methyl malonic acid and molecular defects in MUT or MMACHC yet remain asymptomatic.


Asunto(s)
Acidosis/genética , Carboxiliasas/deficiencia , Proteínas Portadoras/genética , Malonatos/sangre , Errores Innatos del Metabolismo/genética , Metilmalonil-CoA Mutasa/genética , Acetilcarnitina/sangre , Acidosis/sangre , Acidosis/diagnóstico , Acidosis/etnología , Pueblo Asiatico , Enfermedades Asintomáticas , Carboxiliasas/sangre , Carboxiliasas/genética , Carnitina/análogos & derivados , Carnitina/sangre , Niño , Citratos/orina , Femenino , Expresión Génica , Humanos , Lactante , Recién Nacido , Masculino , Malonatos/orina , Malonil Coenzima A/sangre , Malonil Coenzima A/genética , Errores Innatos del Metabolismo/sangre , Errores Innatos del Metabolismo/diagnóstico , Errores Innatos del Metabolismo/etnología , Ácido Metilmalónico/sangre , Mutación , Tamizaje Neonatal , Oxidorreductasas
18.
Metab Eng ; 22: 104-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24502850

RESUMEN

3-Hydroxypropionic acid (3-HP) is an attractive platform chemical, which can be used to produce a variety of commodity chemicals, such as acrylic acid and acrylamide. For enabling a sustainable alternative to petrochemicals as the feedstock for these commercially important chemicals, fermentative production of 3-HP is widely investigated and is centered on bacterial systems in most cases. However, bacteria present certain drawbacks for large-scale organic acid production. In this study, we have evaluated the production of 3-HP in the budding yeast Saccharomyces cerevisiae through a route from malonyl-CoA, because this allows performing the fermentation at low pH thus making the overall process cheaper. We have further engineered the host strain by increasing availability of the precursor malonyl-CoA and by coupling the production with increased NADPH supply we were able to substantially improve 3-HP production by five-fold, up to a final titer of 463 mg l⁻¹. Our work thus led to a demonstration of 3-HP production in yeast via the malonyl-CoA pathway, and this opens for the use of yeast as a cell factory for production of bio-based 3-HP and derived acrylates in the future.


Asunto(s)
Ácido Láctico/análogos & derivados , Malonil Coenzima A/metabolismo , NADP/metabolismo , Saccharomyces cerevisiae/metabolismo , Concentración de Iones de Hidrógeno , Ácido Láctico/biosíntesis , Malonil Coenzima A/genética , NADP/genética , Saccharomyces cerevisiae/genética
20.
Metab Eng ; 20: 187-97, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24148183

RESUMEN

Mithramycin (MTM) is a polyketide antitumor compound produced by Streptomyces argillaceus constituted by a tricyclic aglycone with two aliphatic side chains, a trisaccharide and a disaccharide chain. The biosynthesis of the polyketide aglycone is initiated by the condensation of ten malonyl-CoA units to render a carbon chain that is modified to a tetracyclic intermediate and sequentially glycosylated by five deoxysugars originated from glucose-1-phosphate. Further oxidation and reduction render the final compound. We aimed to increase the precursor supply of malonyl-CoA and/or glucose-1-phosphate in S. argillaceus to enhance MTM production. We have shown that by overexpressing either the S. coelicolor phosphoglucomutase gene pgm or the acetyl-CoA carboxylase ovmGIH genes from the oviedomycin biosynthesis gene cluster in S. argillaceus, we were able to increase the intracellular pool of glucose-1-phosphate and malonyl-CoA, respectively. Moreover, we have cloned the S. argillaceus ADP-glucose pyrophosphorylase gene glgCa and the acyl-CoA:diacylglycerol acyltransferase gene aftAa, and we showed that by inactivating them, an increase of the intracellular concentration of glucose-1-phosphate/glucose-6-phosphate and malonyl-CoA/acetyl-CoA was observed, respectively. Each individual modification resulted in an enhancement of MTM production but the highest production level was obtained by combining all strategies together. In addition, some of these strategies were successfully applied to increase production of four MTM derivatives with improved pharmacological properties: demycarosyl-mithramycin, demycarosyl-3D-ß-D-digitoxosyl-mithramycin, mithramycin SK and mithramycin SDK.


Asunto(s)
Antibióticos Antineoplásicos/biosíntesis , Glucofosfatos , Malonil Coenzima A , Ingeniería Metabólica , Plicamicina/biosíntesis , Streptomyces , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Glucofosfatos/genética , Glucofosfatos/metabolismo , Malonil Coenzima A/genética , Malonil Coenzima A/metabolismo , Streptomyces/genética , Streptomyces/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA