Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters








Publication year range
1.
Life Sci ; 258: 118240, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32781072

ABSTRACT

As a dicarboxylic acid with the structural formula HOOCCH (OH) COOH, tartronic acid is considered as an inhibitor of the transformation of carbohydrates into fat under fat-deficient diet conditions. However, the effect of tartronic acid on lipogenesis under high-fat diet conditions has yet to be established. In this work, we investigated the regulatory role of tartronic acid in lipogenesis in 3T3-L1 adipocytes and C57BL/6J mice. The results confirmed that tartronic acid promoted weight gain (without affecting food intake) and induced adipocyte hypertrophy in epididymal white adipose tissue and lipid accumulation in the livers of high-fat diet-induced obese mice. In vitro, tartronic acid promoted 3T3-L1 adipocyte differentiation by increasing the protein expression of FABP-4, PPARγ and SREBP-1. Moreover, the contents of both acetyl-CoA and malonyl-CoA were significantly upregulated by treatment with tartronic acid, while the protein expression of CPT-1ß were inhibited. In summary, we proved that tartronic acid promotes lipogenesis by serving as substrates for fatty acid synthesis and inhibiting CPT-1ß, providing a new perspective for the study of tartronic acid.


Subject(s)
Acetyl Coenzyme A/biosynthesis , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Lipogenesis/drug effects , Malonyl Coenzyme A/biosynthesis , Tartronates/pharmacology , Up-Regulation/drug effects , 3T3-L1 Cells , Animals , Carnitine O-Palmitoyltransferase/metabolism , Diet, High-Fat/adverse effects , Lipogenesis/physiology , Male , Mice , Mice, Inbred C57BL , Up-Regulation/physiology
2.
J Biol Chem ; 295(30): 10168-10179, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32493774

ABSTRACT

Feeding of rapeseed (canola) oil with a high erucic acid concentration is known to cause hepatic steatosis in animals. Mitochondrial fatty acid oxidation plays a central role in liver lipid homeostasis, so it is possible that hepatic metabolism of erucic acid might decrease mitochondrial fatty acid oxidation. However, the precise mechanistic relationship between erucic acid levels and mitochondrial fatty acid oxidation is unclear. Using male Sprague-Dawley rats, along with biochemical and molecular biology approaches, we report here that peroxisomal ß-oxidation of erucic acid stimulates malonyl-CoA formation in the liver and thereby suppresses mitochondrial fatty acid oxidation. Excessive hepatic uptake and peroxisomal ß-oxidation of erucic acid resulted in appreciable peroxisomal release of free acetate, which was then used in the synthesis of cytosolic acetyl-CoA. Peroxisomal metabolism of erucic acid also remarkably increased the cytosolic NADH/NAD+ ratio, suppressed sirtuin 1 (SIRT1) activity, and thereby activated acetyl-CoA carboxylase, which stimulated malonyl-CoA biosynthesis from acetyl-CoA. Chronic feeding of a diet including high-erucic-acid rapeseed oil diminished mitochondrial fatty acid oxidation and caused hepatic steatosis and insulin resistance in the rats. Of note, administration of a specific peroxisomal ß-oxidation inhibitor attenuated these effects. Our findings establish a cross-talk between peroxisomal and mitochondrial fatty acid oxidation. They suggest that peroxisomal oxidation of long-chain fatty acids suppresses mitochondrial fatty acid oxidation by stimulating malonyl-CoA formation, which might play a role in fatty acid-induced hepatic steatosis and related metabolic disorders.


Subject(s)
Erucic Acids/metabolism , Fatty Liver/metabolism , Liver/metabolism , Malonyl Coenzyme A/biosynthesis , Mitochondria, Liver/metabolism , Peroxisomes/metabolism , Animals , Fatty Liver/pathology , Insulin Resistance , Liver/pathology , Male , Mitochondria, Liver/pathology , Oxidation-Reduction , Peroxisomes/pathology , Rats , Rats, Sprague-Dawley
3.
Sci China Life Sci ; 63(11): 1734-1743, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32347474

ABSTRACT

Phloretin is an important skin-lightening and depigmenting agent from the peel of apples. Although de novo production of phloretin has been realized in microbes using the natural pathway from plants, the efficiency of phloretin production is still not enough for industrial application. Here, we established an artificial pathway in the yeast to produce phloretin via assembling two genes of p-coumaroyl-CoA ligase (4CL) and chalcone synthase (CHS). CHS is a key enzyme which conventionally condenses a CoA-tethered starter with three molecules of malonyl-CoA to form the backbone of flavonoids. However, there was 33% of by-product generated via CHS by condensing two molecules of malonyl-CoA during the fermentation process. Hence, we introduced a more efficient CHS and improved the supply of malonyl-CoA through two pathways; the by-product ratio was decreased from 33% to 17% and the production of phloretin was improved from 48 to 83.2 mg L-1. Finally, a fed-batch fermentation process was optimized and the production of phloretin reached 619.5 mg L-1, which was 14-fold higher than that of the previous studies. Our work established a platform for the biosynthesis of phloretin from the low-cost raw material 3-(4-hydroxyphenyl) propanoic acid and also illustrated the potential for industrial scale bio-manufacturing of phloretin.


Subject(s)
Acyltransferases/genetics , Phloretin/metabolism , Saccharomyces cerevisiae/genetics , Acyltransferases/metabolism , Bioreactors , Biosynthetic Pathways , Fermentation , Malonyl Coenzyme A/biosynthesis , Metabolic Engineering , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/metabolism
4.
N Biotechnol ; 52: 19-24, 2019 Sep 25.
Article in English | MEDLINE | ID: mdl-30995533

ABSTRACT

Lovastatin is widely prescribed to reduce elevated levels of cholesterol and prevent heart-related diseases. Cultivation of Aspergillus terreus (ATCC 20542) with carbohydrates or low-value feedstocks such as glycerol produces lovastatin as a secondary metabolite and (+)-geodin as a by-product. An A. terreus mutant strain was developed (gedCΔ) with a disrupted (+)-geodin biosynthesis pathway. The gedCΔ mutant was created by inserting the antibiotic marker hygromycin B (hyg) within the gedC gene that encodes emodin anthrone polyketide synthase (PKS), a primary gene responsible for initiating (+)-geodin biosynthesis. The effects of emodin anthrone PKS gene disruption on (+)-geodin and lovastatin biosynthesis and the production of the precursors acetyl-CoA and malonyl-CoA were investigated with cultures based on glycerol alone and in combination with lactose. The gedCΔ strain showed improved lovastatin production, particularly when cultivated on the glycerol-lactose mixture, increasing lovastatin production by 80% (113 mg/L) while simultaneously inhibiting (+)-geodin biosynthesis compared to the wild-type strain. This study thus shows that suppression of the (+)-geodin pathway increases lovastatin yield and demonstrates a practical approach of manipulating carbon flux by modulating enzyme activity.


Subject(s)
Aspergillus/metabolism , Benzofurans/metabolism , Biosynthetic Pathways , Lovastatin/biosynthesis , Acetyl Coenzyme A/biosynthesis , Glycerol/metabolism , Lactose/metabolism , Malonyl Coenzyme A/biosynthesis
5.
N Biotechnol ; 44: 64-71, 2018 Sep 25.
Article in English | MEDLINE | ID: mdl-29727712

ABSTRACT

The present work describes the application of homologous recombination techniques in a wild-type Aspergillus terreus (ATCC 20542) strain to increase the flow of precursors towards the lovastatin biosynthesis pathway. A new strain was generated to overexpress acetyl-CoA carboxylase (ACCase) by replacing the native ACCase promoter with a strong constitutive PadhA promoter from Aspergillus nidulans. Glycerol and a mixture of lactose and glycerol were used independently as the carbon feedstock to determine the degree of response by the A. terreus strains towards the production of acetyl-CoA, and malonyl-CoA. The new strain increased the levels of malonyl-CoA and acetyl-CoA by 240% and 14%, respectively, compared to the wild-type strain. As a result, lovastatin production was increased by 40% and (+)-geodin was decreased by 31% using the new strain. This study shows for the first time that the metabolism of Aspergillus terreus can be manipulated to attain higher levels of precursors and valuable secondary metabolites.


Subject(s)
Acetyl Coenzyme A/biosynthesis , Acetyl-CoA Carboxylase , Aspergillus , Fungal Proteins , Lovastatin/biosynthesis , Malonyl Coenzyme A/biosynthesis , Acetyl-CoA Carboxylase/biosynthesis , Acetyl-CoA Carboxylase/genetics , Aspergillus/enzymology , Aspergillus/genetics , Fungal Proteins/biosynthesis , Fungal Proteins/genetics
7.
J Biol Chem ; 290(45): 26994-27011, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26378232

ABSTRACT

Understanding the biosynthetic mechanism of the atypical polyketide extender unit is important for the development of bioactive natural products. Reveromycin (RM) derivatives produced by Streptomyces sp. SN-593 possess several aliphatic extender units. Here, we studied the molecular basis of 2-alkylmalonyl-CoA formation by analyzing the revR and revS genes, which form a transcriptional unit with the revT gene, a crotonyl-CoA carboxylase/reductase homolog. We mainly focused on the uncharacterized adenylate-forming enzyme (RevS). revS gene disruption resulted in the reduction of all RM derivatives, whereas reintroduction of the gene restored the yield of RMs. Although RevS was classified in the fatty acyl-AMP ligase clade based on phylogenetic analysis, biochemical characterization revealed that the enzyme catalyzed the middle chain fatty acyl-CoA ligase (FACL) but not the fatty acyl-AMP ligase activity, suggesting the molecular evolution for acyl-CoA biosynthesis. Moreover, we examined the in vitro conversion of fatty acid into 2-alkylmalonyl-CoA using purified RevS and RevT. The coupling reaction showed efficient conversion of hexenoic acid into butylmalonyl-CoA. RevS efficiently catalyzed C8-C10 middle chain FACL activity; therefore, we speculated that the acyl-CoA precursor was truncated via ß-oxidation and converted into (E)-2-enoyl-CoA, a RevT substrate. To determine whether the ß-oxidation process is involved between the RevS and RevT reaction, we performed the feeding experiment using [1,2,3,4-(13)C]octanoic acid. (13)C NMR analysis clearly demonstrated incorporation of the [3,4-(13)C]octanoic acid moiety into the structure of RM-A. Our results provide insight into the role of uncharacterized RevS homologs that may catalyze middle chain FACL to produce a unique polyketide extender unit.


Subject(s)
Bacterial Proteins/metabolism , Coenzyme A Ligases/metabolism , Malonyl Coenzyme A/analogs & derivatives , Streptomyces/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Coenzyme A Ligases/genetics , Genes, Bacterial , Malonyl Coenzyme A/biosynthesis , Malonyl Coenzyme A/chemistry , Molecular Sequence Data , Molecular Structure , Phylogeny , Polyketides/chemistry , Polyketides/metabolism , Pyrans/chemistry , Pyrans/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Spiro Compounds/chemistry , Spiro Compounds/metabolism , Streptomyces/genetics , Substrate Specificity
8.
PLoS One ; 7(4): e34443, 2012.
Article in English | MEDLINE | ID: mdl-22509304

ABSTRACT

BACKGROUND: Numerous vitamin-D analogs exhibited poor response rates, high systemic toxicities and hypercalcemia in human trials to treat cancer. We identified the first non-hypercalcemic anti-cancer vitamin D analog MT19c by altering the A-ring of ergocalciferol. This study describes the therapeutic efficacy and mechanism of action of MT19c in both in vitro and in vivo models. METHODOLOGY/PRINCIPAL FINDING: Antitumor efficacy of MT19c was evaluated in ovarian cancer cell (SKOV-3) xenografts in nude mice and a syngenic rat ovarian cancer model. Serum calcium levels of MT19c or calcitriol treated animals were measured. In-silico molecular docking simulation and a cell based VDR reporter assay revealed MT19c-VDR interaction. Genomewide mRNA analysis of MT19c treated tumors identified drug targets which were verified by immunoblotting and microscopy. Quantification of cellular malonyl CoA was carried out by HPLC-MS. A binding study with PPAR-Y receptor was performed. MT19c reduced ovarian cancer growth in xenograft and syngeneic animal models without causing hypercalcemia or acute toxicity. MT19c is a weak vitamin-D receptor (VDR) antagonist that disrupted the interaction between VDR and coactivator SRC2-3. Genome-wide mRNA analysis and western blot and microscopy of MT19c treated xenograft tumors showed inhibition of fatty acid synthase (FASN) activity. MT19c reduced cellular levels of malonyl CoA in SKOV-3 cells and inhibited EGFR/phosphoinositol-3kinase (PI-3K) activity independently of PPAR-gamma protein. SIGNIFICANCE: Antitumor effects of non-hypercalcemic agent MT19c provide a new approach to the design of vitamin-D based anticancer molecules and a rationale for developing MT19c as a therapeutic agent for malignant ovarian tumors by targeting oncogenic de novo lipogenesis.


Subject(s)
Ergocalciferols/chemistry , Fatty Acids/biosynthesis , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Xenograft Model Antitumor Assays , Amino Acid Sequence , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Calcium/blood , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Citric Acid/metabolism , Down-Regulation/drug effects , ErbB Receptors/metabolism , Ergocalciferols/adverse effects , Ergocalciferols/metabolism , Ergocalciferols/pharmacology , Fatty Acid Synthases/metabolism , Female , Homeostasis/drug effects , Humans , Hypercalcemia/chemically induced , L-Lactate Dehydrogenase/metabolism , Malonyl Coenzyme A/biosynthesis , Membrane Potential, Mitochondrial/drug effects , Mice , Molecular Dynamics Simulation , Molecular Sequence Data , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Rats , Receptors, Calcitriol/antagonists & inhibitors , Receptors, Calcitriol/chemistry , Receptors, Calcitriol/metabolism , Safety , Signal Transduction/drug effects
9.
J Bacteriol ; 194(1): 72-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22037404

ABSTRACT

Acetyl coenzyme A (acteyl-CoA) carboxylase (ACC) is the first committed enzyme of the fatty acid synthesis pathway. Escherichia coli ACC is composed of four different proteins. The first enzymatic activity of the ACC complex, biotin carboxylase (BC), catalyzes the carboxylation of the protein-bound biotin moiety of another subunit with bicarbonate in an ATP-dependent reaction. Although BC is found as a dimer in cell extracts and the carboxylase activities of the two subunits of the dimer are interdependent, mutant BC proteins deficient in dimerization are reported to retain appreciable activity in vitro (Y. Shen, C. Y. Chou, G. G. Chang, and L. Tong, Mol. Cell 22:807-818, 2006). However, in vivo BC must interact with the other proteins of the complex, and thus studies of the isolated BC may not reflect the intracellular function of the enzyme. We have tested the abilities of three BC mutant proteins deficient in dimerization to support growth and report that the two BC proteins most deficient in dimerization fail to support growth unless expressed at high levels. In contrast, the wild-type protein supports growth at low expression levels. We conclude that BC must be dimeric to fulfill its physiological function.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Carbon-Nitrogen Ligases/metabolism , Escherichia coli/enzymology , Acetyl-CoA Carboxylase/genetics , Alleles , Carbon-Nitrogen Ligases/chemistry , Carbon-Nitrogen Ligases/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic , Genetic Complementation Test , Malonyl Coenzyme A/biosynthesis , Malonyl Coenzyme A/chemistry , Molecular Structure , Plasmids , Protein Multimerization , Protein Subunits
10.
FEBS Lett ; 585(20): 3337-41, 2011 Oct 20.
Article in English | MEDLINE | ID: mdl-21959040

ABSTRACT

Very long-chain fatty acids (VLCFAs) have a variety of physiological functions and are related to numerous disorders. The key step of VLCFA elongation is catalyzed by members of the elongase family, ELOVLs. Mammals have seven ELOVLs (ELOVL1-7), yet none of them has been purified and analyzed. In the presented study we purified ELOVL7 and measured its activity by reconstituting it into proteoliposomes. Purified ELOVL7 exhibited high activity toward acyl-CoAs with C18 carbon chain length. The calculated K(m) values toward C18:3(n-3)-CoA and malonyl-CoA were both in the µM range. We also found that progression of the VLCFA cycle enhances ELOVL7 activity.


Subject(s)
Acetyltransferases/chemistry , Fatty Acids/chemistry , Malonyl Coenzyme A/chemistry , Acetyltransferases/genetics , Acetyltransferases/isolation & purification , Acetyltransferases/metabolism , Catalysis , Fatty Acid Elongases , Fatty Acids/biosynthesis , Fatty Acids/genetics , HEK293 Cells , Humans , Malonyl Coenzyme A/biosynthesis , Malonyl Coenzyme A/metabolism
11.
Metab Eng ; 13(5): 578-87, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21763447

ABSTRACT

Malonyl-coenzyme A is an important precursor metabolite for the biosynthesis of polyketides, flavonoids and biofuels. However, malonyl-CoA naturally synthesized in microorganisms is consumed for the production of fatty acids and phospholipids leaving only a small amount available for the production of other metabolic targets in recombinant biosynthesis. Here we present an integrated computational and experimental approach aimed at improving the intracellular availability of malonyl-CoA in Escherichia coli. We used a customized version of the recently developed OptForce methodology to predict a minimal set of genetic interventions that guarantee a prespecified yield of malonyl-CoA in E. coli strain BL21 Star™. In order to validate the model predictions, we have successfully constructed an E. coli recombinant strain that exhibits a 4-fold increase in the levels of intracellular malonyl-CoA compared to the wild type strain. Furthermore, we demonstrate the potential of this E. coli strain for the production of plant-specific secondary metabolites naringenin (474mg/L) with the highest yield ever achieved in a lab-scale fermentation process. Combined effect of the genetic interventions was found to be synergistic based on a developed analysis method that correlates genetic modification to cell phenotype, specifically the identified knockout targets (ΔfumC and ΔsucC) and overexpression targets (ACC, PGK, GAPD and PDH) can cooperatively force carbon flux towards malonyl-CoA. The presented strategy can also be readily expanded for the production of other malonyl-CoA-derived compounds like polyketides and biofuels.


Subject(s)
Escherichia coli , Genome, Bacterial , Malonyl Coenzyme A , Models, Biological , Organisms, Genetically Modified , Carbon/metabolism , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Flavanones/biosynthesis , Flavanones/genetics , Malonyl Coenzyme A/biosynthesis , Malonyl Coenzyme A/genetics , Organisms, Genetically Modified/genetics , Organisms, Genetically Modified/growth & development , Organisms, Genetically Modified/metabolism
12.
J Am Chem Soc ; 133(4): 976-85, 2011 Feb 02.
Article in English | MEDLINE | ID: mdl-21175203

ABSTRACT

The allyl moiety of the immunosuppressive agent FK506 is structurally unique among polyketides and critical for its potent biological activity. Here, we detail the biosynthetic pathway to allylmalonyl-coenzyme A (CoA), from which the FK506 allyl group is derived, based on a comprehensive chemical, biochemical, and genetic interrogation of three FK506 gene clusters. A discrete polyketide synthase (PKS) with noncanonical domain architecture presumably in coordination with the fatty acid synthase pathway of the host catalyzes a multistep enzymatic reaction to allylmalonyl-CoA via trans-2-pentenyl-acyl carrier protein. Characterization of this discrete pathway facilitated the engineered biosynthesis of novel allyl group-modified FK506 analogues, 36-fluoro-FK520 and 36-methyl-FK506, the latter of which exhibits improved neurite outgrowth activity. This unique feature of FK506 biosynthesis, in which a dedicated PKS provides an atypical extender unit for the main modular PKS, illuminates a new strategy for the combinatorial biosynthesis of designer macrolide scaffolds as well as FK506 analogues.


Subject(s)
Malonyl Coenzyme A/biosynthesis , Malonyl Coenzyme A/chemistry , Polyketide Synthases/metabolism , Sequence Deletion , Tacrolimus/analogs & derivatives , Tacrolimus/metabolism , Malonyl Coenzyme A/metabolism , Multigene Family , Streptomyces/enzymology , Streptomyces/genetics , Streptomyces/metabolism
13.
Biotechnol Lett ; 32(2): 277-82, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19838628

ABSTRACT

Using metabolic engineering, we developed Streptomyces venezuelae YJ028 as an efficient heterologous host to increase the malonyl-CoA pool to be directed towards enhanced production of various polyketides. To probe the applicability of newly developed hosts in the heterologous production of polyketides, we expressed type III polyketide synthase, 1,3,6,8-tetrahydroxynaphthalene synthase, in these hosts. Flaviolin production was doubled by expression of acetyl-CoA carboxylase (ACCase) and 4-fold by combined expression of ACCase, metK1-sp and afsR-sp. Thus, the newly developed Streptomyces venezuelae YJ028 hosts produce heterologous polyketides more efficiently than the parent strain.


Subject(s)
Genetic Enhancement/methods , Macrolides/metabolism , Malonyl Coenzyme A/biosynthesis , Metabolome/physiology , Protein Engineering/methods , Streptomyces/physiology , Malonyl Coenzyme A/genetics , Recombinant Proteins/metabolism , Signal Transduction/physiology
14.
Appl Environ Microbiol ; 75(18): 5831-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19633125

ABSTRACT

Identification of genetic targets able to bring about changes to the metabolite profiles of microorganisms continues to be a challenging task. We have independently developed a cipher of evolutionary design (CiED) to identify genetic perturbations, such as gene deletions and other network modifications, that result in optimal phenotypes for the production of end products, such as recombinant natural products. Coupled to an evolutionary search, our method demonstrates the utility of a purely stoichiometric network to predict improved Escherichia coli genotypes that more effectively channel carbon flux toward malonyl coenzyme A (CoA) and other cofactors in an effort to generate recombinant strains with enhanced flavonoid production capacity. The engineered E. coli strains were constructed first by the targeted deletion of native genes predicted by CiED and then second by incorporating selected overexpressions, including those of genes required for the coexpression of the plant-derived flavanones, acetate assimilation, acetyl-CoA carboxylase, and the biosynthesis of coenzyme A. As a result, the specific flavanone production from our optimally engineered strains was increased by over 660% for naringenin (15 to 100 mg/liter/optical density unit [OD]) and by over 420% for eriodictyol (13 to 55 mg/liter/OD).


Subject(s)
Escherichia coli/genetics , Escherichia coli/metabolism , Flavanones/biosynthesis , Genetic Engineering , Malonyl Coenzyme A/biosynthesis , Metabolic Networks and Pathways/genetics , Escherichia coli Proteins/genetics , Gene Deletion , Gene Expression , Models, Biological
15.
Chembiochem ; 10(10): 1714-23, 2009 Jul 06.
Article in English | MEDLINE | ID: mdl-19507202

ABSTRACT

In order to study intermediates in polyketide biosynthesis two nonhydrolyzable malonyl coenzyme A analogues were synthesised by a chemoenzymatic route. In these analogues the sulfur atom of CoA was replaced either by a methylene group (carbadethia analogue) or by an oxygen atom (oxadethia analogue). These malonyl-CoA analogues were found to compete with the natural extender unit malonyl-CoA and to trap intermediates from stilbene synthase, a type III polyketide synthase (PKS). From the reaction of stilbene synthase with its natural phenylpropanoid substrates, diketide, triketide and tetraketide species were successfully off-loaded and characterised by LC-MS. Moreover, the reactivity of the nonhydrolyzable analogues offers insights into the flexibility of substrate alignment in the PKS active site for efficient malonyl decarboxylation and condensation.


Subject(s)
Acyltransferases/metabolism , Adenine Nucleotides/chemistry , Antioxidants/chemistry , Macrolides/chemistry , Malonyl Coenzyme A/chemistry , Adenine Nucleotides/biosynthesis , Adenine Nucleotides/pharmacology , Biocatalysis , Chromatography, Liquid , Malonyl Coenzyme A/biosynthesis , Malonyl Coenzyme A/pharmacology , Mass Spectrometry
16.
Hepatology ; 49(4): 1166-75, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19177596

ABSTRACT

UNLABELLED: Hepatic steatosis is a hallmark of nonalcoholic fatty liver disease (NAFLD) and a key component of obesity-associated metabolic dysfunctions featuring dyslipidemia, insulin resistance, and loss of glycemic control. It has yet to be completely understood how much dysregulated de novo lipogenesis contributes to the pathogenic development of hepatic steatosis and insulin resistance. ATP-citrate lyase (ACL) is a lipogenic enzyme that catalyzes the critical reaction linking cellular glucose catabolism and lipogenesis, converting cytosolic citrate to acetyl-coenzyme A (CoA). Acetyl-CoA is further converted to malonyl-CoA, the essential precursor for fatty acid biosynthesis. We investigated whether dysregulation of hepatic ACL is metabolically connected to hepatic steatosis, insulin resistance, and hyperglycemia. We found that in leptin receptor-deficient db/db mice, the expression of ACL was selectively elevated in the liver but not in the white adipose tissue. Liver-specific ACL abrogation via adenovirus-mediated RNA interference prominently reduced the hepatic contents of both acetyl-CoA and malonyl-CoA, markedly inhibited hepatic de novo lipogenesis, and protected against hepatic steatosis in db/db mice. Surprisingly, liver-specific ACL abrogation markedly inhibited the expression of peroxisome proliferator-activated receptor-gamma and the entire lipogenic program in the liver. Moreover, hepatic ACL deficiency resulted in significantly down-regulated expression of gluconeogenic genes in the liver as well as enhanced insulin sensitivity in the muscle, leading to substantially improved systemic glucose metabolism. CONCLUSION: These findings establish a crucial role of hepatic ACL in lipid and glucose metabolism; therefore, hepatic ACL may serve as a potential target to treat NAFLD and type 2 diabetes.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Fatty Liver/metabolism , Hyperglycemia/metabolism , Lipid Metabolism , Receptors, Leptin/deficiency , Acetyl Coenzyme A/biosynthesis , Animals , Fatty Acids, Nonesterified/blood , Fatty Liver/prevention & control , Glucose/metabolism , Homeostasis , Insulin Resistance , Lipogenesis , Liver/enzymology , Male , Malonyl Coenzyme A/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA Interference
17.
J Mol Cell Cardiol ; 41(5): 868-75, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17020764

ABSTRACT

We previously showed that, in the perfused rat heart, the capacity of n-fatty acids to generate mitochondrial acetyl-CoA decreases as their chain length increases. In the present study, we investigated whether the oxidation of a long-chain fatty acid, oleate, is inhibited by short-chain fatty acids, acetate or propionate (which do and do not generate mitochondrial acetyl-CoA, respectively). We perfused rat hearts with buffer containing 4 mM glucose, 0.2 mM pyruvate, 1 mM lactate, and various concentrations of either (i) [U-(13)C]acetate, (ii) [U-(13)C]acetate plus [1-(13)C]oleate, or (iii) unlabeled propionate plus [1-(13)C]oleate. Using mass isotopomer analysis, we determined the contributions of the labeled substrates to the acetyl moiety of citrate (a probe of mitochondrial acetyl-CoA) and to malonyl-CoA. We found that acetate, even at low concentration, markedly inhibits the oxidation of [1-(13)C]oleate in the heart, without change in malonyl-CoA concentration. We also found that propionate, at a concentration higher than 1 mM, decreases (i) the contribution of [1-(13)C]oleate to mitochondrial acetyl-CoA and (ii) malonyl-CoA concentration. The inhibition by acetate or propionate of acetyl-CoA production from oleate probably results from a competition for mitochondrial CoA between the CoA-utilizing enzymes.


Subject(s)
Acetyl Coenzyme A/biosynthesis , Malonyl Coenzyme A/biosynthesis , Mitochondria, Heart/metabolism , Myocardium/metabolism , Oleic Acid/metabolism , Acetates/chemistry , Animals , Fatty Acids , In Vitro Techniques , Male , Models, Cardiovascular , Oxidation-Reduction , Perfusion , Propionates/chemistry , Rats , Rats, Sprague-Dawley
18.
Appl Microbiol Biotechnol ; 66(1): 85-91, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15179529

ABSTRACT

Development of host microorganisms for heterologous expression of polyketide synthases (PKS) that possess the intrinsic capacity to overproduce polyketides with a broad spectrum of precursors supports the current demand for new tools to create novel chemical structures by combinatorial engineering of modular and other classes of PKS. Streptomyces fradiae is an ideal host for development of generic polyketide-overproducing strains because it contains three of the most common precursors--malonyl-CoA, methylmalonyl-CoA and ethylmalonyl-CoA--used by modular PKS, and is a host that is amenable to genetic manipulation. We have expanded the utility of an overproducing S. fradiae strain for engineered biosynthesis of polyketides by engineering a biosynthetic pathway for methoxymalonyl-ACP, a fourth precursor used by many 16-membered macrolide PKS. This was achieved by introducing a set of five genes, fkbG-K from Streptomyces hygroscopicus, putatively encoding the methoxymalonyl-ACP biosynthetic pathway, into the S. fradiae chromosome. Heterologous expression of the midecamycin PKS genes in this strain resulted in 1 g/l production of a midecamycin analog. These results confirm the ability to engineer unusual precursor pathways to support high levels of polyketide production, and validate the use of S. fradiae for overproduction of 16-membered macrolides derived from heterologous PKS that require a broad range of precursors.


Subject(s)
Acyl Carrier Protein/metabolism , Macrolides/metabolism , Malonyl Coenzyme A/analogs & derivatives , Streptomyces/metabolism , Fermentation , Gene Expression Regulation, Bacterial , Genes, Bacterial , Genetic Engineering , Leucomycins/biosynthesis , Malonyl Coenzyme A/biosynthesis , Malonyl Coenzyme A/metabolism , Streptomyces/genetics
19.
J Biol Chem ; 279(33): 34298-301, 2004 Aug 13.
Article in English | MEDLINE | ID: mdl-15181001

ABSTRACT

The goal of this study was to test the relationship between malonyl-CoA concentration and its turnover measured in isolated rat hearts perfused with NaH(13)CO(3). This turnover is a direct measurement of the flux of acetyl-CoA carboxylation in the intact heart. It also reflects the rate of malonyl-CoA decarboxylation, i.e. the only known fate of malonyl-CoA in the heart. Conditions were selected to result in stable malonyl-CoA concentrations ranging from 1.5 to 5 nmol.g wet weight-(1). The malonyl-CoA concentration was directly correlated with the turnover of malonyl-CoA, ranging from 0.7 to 4.2 nmol.min(-) (1).g wet weight(-1) (slope = 0.98, r(2) = 0.94). The V(max) activities of acetyl-CoA carboxylase and of malonyl-CoA decarboxylase exceeded the rate of malonyl-CoA turnover by 2 orders of magnitude and did not correlate with either concentration or turnover of malonyl-CoA. However, conditions of perfusion that increased acetyl-CoA supply resulted in higher turnover and concentration, demonstrating that malonyl-CoA turnover is regulated by the supply of acetyl-CoA. The only condition where the activity of malonyl-CoA decarboxylase regulated malonyl-CoA kinetics was when the enzyme was pharmacologically inhibited, resulting in increased malonyl-CoA concentration and decreased turnover. Our data show that, in the absence of enzyme inhibitors, the rate of acetyl-CoA carboxylation is the main determinant of the malonyl-CoA concentration in the heart.


Subject(s)
Malonyl Coenzyme A/biosynthesis , Myocardium/metabolism , Acetyl Coenzyme A/chemistry , Acetyl-CoA Carboxylase/metabolism , Animals , Biochemical Phenomena , Biochemistry , Carboxy-Lyases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Heart/physiology , Kinetics , Perfusion , Rats , Rats, Sprague-Dawley , Swine , Time Factors
20.
J Biochem Mol Biol ; 35(5): 443-51, 2002 Sep 30.
Article in English | MEDLINE | ID: mdl-12359084

ABSTRACT

Malonate is a three-carbon dicarboxylic acid. It is well known as a competitive inhibitor of succinate dehydrogenase. It occurs naturally in biological systems, such as legumes and developing rat brains, which indicates that it may play an important role in symbiotic nitrogen metabolism and brain development. Recently, enzymes that are related to malonate metabolism were discovered and characterized. The genes that encode the enzymes were isolated, and the regulation of their expression was also studied. The mutant bacteria, in which the malonate-metabolizing gene was deleted, lost its primary function, symbiosis, between Rhizobium leguminosarium bv trifolii and clover. This suggests that malonate metabolism is essential in symbiotic nitrogen metabolism, at least in clover nodules. In addition to these, the genes matB and matC have been successfully used for generation of the industrial strain of Streptomyces for the production of antibiotics.


Subject(s)
Bacterial Proteins , Malonates/metabolism , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Base Sequence , Carboxy-Lyases/chemistry , Carboxy-Lyases/metabolism , DNA , Fabaceae/metabolism , Fabaceae/microbiology , Industrial Microbiology , Malonates/chemistry , Malonyl Coenzyme A/biosynthesis , Molecular Sequence Data , Protein Structure, Tertiary , Rhizobium/genetics , Rhizobium/metabolism , Symbiosis
SELECTION OF CITATIONS
SEARCH DETAIL