Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Biol Chem ; 300(7): 107412, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38796064

ABSTRACT

The heart alters the rate and relative oxidation of fatty acids and glucose based on availability and energetic demand. Insulin plays a crucial role in this process diminishing fatty acid and increasing glucose oxidation when glucose availability increases. Loss of insulin sensitivity and metabolic flexibility can result in cardiovascular disease. It is therefore important to identify mechanisms by which insulin regulates substrate utilization in the heart. Mitochondrial pyruvate dehydrogenase (PDH) is the key regulatory site for the oxidation of glucose for ATP production. Nevertheless, the impact of insulin on PDH activity has not been fully delineated, particularly in the heart. We sought in vivo evidence that insulin stimulates cardiac PDH and that this process is driven by the inhibition of fatty acid oxidation. Mice injected with insulin exhibited dephosphorylation and activation of cardiac PDH. This was accompanied by an increase in the content of malonyl-CoA, an inhibitor of carnitine palmitoyltransferase 1 (CPT1), and, thus, mitochondrial import of fatty acids. Administration of the CPT1 inhibitor oxfenicine was sufficient to activate PDH. Malonyl-CoA is produced by acetyl-CoA carboxylase (ACC). Pharmacologic inhibition or knockout of cardiac ACC diminished insulin-dependent production of malonyl-CoA and activation of PDH. Finally, circulating insulin and cardiac glucose utilization exhibit daily rhythms reflective of nutritional status. We demonstrate that time-of-day-dependent changes in PDH activity are mediated, in part, by ACC-dependent production of malonyl-CoA. Thus, by inhibiting fatty acid oxidation, insulin reciprocally activates PDH. These studies identify potential molecular targets to promote cardiac glucose oxidation and treat heart disease.


Subject(s)
Fatty Acids , Insulin , Myocardium , Oxidation-Reduction , Pyruvate Dehydrogenase Complex , Animals , Insulin/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Mice , Myocardium/metabolism , Myocardium/enzymology , Fatty Acids/metabolism , Acetyl-CoA Carboxylase/metabolism , Acetyl-CoA Carboxylase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Carnitine O-Palmitoyltransferase/genetics , Malonyl Coenzyme A/metabolism , Male , Mice, Knockout , Glucose/metabolism , Mice, Inbred C57BL
2.
Biol Pharm Bull ; 47(1): 23-27, 2024.
Article in English | MEDLINE | ID: mdl-38171776

ABSTRACT

Mammalian type 2 carnitine parmitoyltransferase (EC 2.3.1.21), abbreviated as CPT2, is an enzyme involved in the translocation of fatty acid into the mitochondrial matrix space, and catalyzes the reaction acylcarnitine + CoA = acyl-CoA + carnitine. When rat CPT2 was expressed in Escherichia coli, its behavior was dependent on the presence or absence of i) its mitochondrial localization sequence and ii) a short amino acid sequence thought to anchor it to the mitochondrial inner membrane: CPT2 containing both sequences behaved as a hydrophobic protein, while recombinant CPT2 lacking both regions behaved as a water soluble protein; if only one region was present, the resultant proteins were observed in both fractions. Because relatively few protein species could be obtained from bacterial lysates as insoluble pellets under the experimental conditions used, selective enrichment of recombinant CPT2 protein containing both hydrophobic sequences was easily achieved. Furthermore, when CPT2 enriched in insoluble fraction was resuspended in an appropriate medium, it showed catalytic activity typical of CPT2: it was completely suppressed by the CPT2 inhibitor, ST1326, but not by the CPT1 inhibitor, malonyl-CoA. Therefore, we conclude that the bacterial expression system is an effective tool for characterization studies of mammalian CPT2.


Subject(s)
Carnitine O-Palmitoyltransferase , Mitochondria , Rats , Animals , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/chemistry , Mitochondria/metabolism , Malonyl Coenzyme A/metabolism , Malonyl Coenzyme A/pharmacology , Fatty Acids/metabolism , Recombinant Proteins/genetics , Carnitine/metabolism , Mammals/metabolism
3.
J Agric Food Chem ; 72(1): 94-107, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38126236

ABSTRACT

Microbial cell factories have shown great potential for industrial production with the benefit of being environmentally friendly and sustainable. Yarrowia lipolytica is a promising and superior non-model host for biomanufacturing due to its cumulated advantages compared to model microorganisms, such as high fluxes of metabolic precursors (acetyl-CoA and malonyl-CoA) and its naturally hydrophobic microenvironment. However, although diverse compounds have been synthesized in Y. lipolytica cell factories, most of the relevant studies have not reached the level of industrialization and commercialization due to a number of remaining challenges, including unbalanced metabolic flux, conflict between cell growth and product synthesis, and cytotoxic effects. Here, various metabolic engineering strategies for solving the challenges are summarized, which is developing fast and extremely conducive to rational design and reconstruction of robust Y. lipolytica cell factories for advanced biomanufacturing. Finally, future engineering efforts for enhancing the production efficiency of this platform strain are highlighted.


Subject(s)
Yarrowia , Yarrowia/metabolism , Metabolic Engineering , Acetyl Coenzyme A/metabolism , Malonyl Coenzyme A/metabolism , Industry
4.
J Agric Food Chem ; 72(1): 566-576, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38154088

ABSTRACT

Curcumin is a natural phenylpropanoid compound with various biological activities and is widely used in food and pharmaceuticals. A de novo curcumin biosynthetic pathway was constructed in Escherichia coli BL21(DE3). Optimization of the curcumin biosynthesis module achieved a curcumin titer of 26.8 ± 0.6 mg/L. Regulating the metabolic fluxes of the ß-oxidation pathway and fatty acid elongation cycle and blocking the endogenous malonyl-CoA consumption pathway increased the titer to 113.6 ± 7.1 mg/L. Knockout of endogenous curcumin reductase (curA) and intermediate product detoxification by heterologous expression of the solvent-resistant pump (srpB) increased the titer to 137.5 ± 3.0 mg/L. A 5 L pilot-scale fermentation, using a three-stage pH alternation strategy, increased the titer to 696.2 ± 20.9 mg/L, 178.5-fold higher than the highest curcumin titer from de novo biosynthesis previously reported, thereby laying the foundation for efficient biosynthesis of curcumin and its derivatives.


Subject(s)
Curcumin , Escherichia coli Proteins , Escherichia coli/genetics , Escherichia coli/metabolism , Curcumin/metabolism , Malonyl Coenzyme A/metabolism , Escherichia coli Proteins/metabolism , Biosynthetic Pathways , Metabolic Engineering
5.
Cell Metab ; 36(5): 1088-1104.e12, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38447582

ABSTRACT

Acetyl-CoA carboxylase (ACC) promotes prandial liver metabolism by producing malonyl-CoA, a substrate for de novo lipogenesis and an inhibitor of CPT-1-mediated fat oxidation. We report that inhibition of ACC also produces unexpected secondary effects on metabolism. Liver-specific double ACC1/2 knockout (LDKO) or pharmacologic inhibition of ACC increased anaplerosis, tricarboxylic acid (TCA) cycle intermediates, and gluconeogenesis by activating hepatic CPT-1 and pyruvate carboxylase flux in the fed state. Fasting should have marginalized the role of ACC, but LDKO mice maintained elevated TCA cycle intermediates and preserved glycemia during fasting. These effects were accompanied by a compensatory induction of proteolysis and increased amino acid supply for gluconeogenesis, which was offset by increased protein synthesis during feeding. Such adaptations may be related to Nrf2 activity, which was induced by ACC inhibition and correlated with fasting amino acids. The findings reveal unexpected roles for malonyl-CoA synthesis in liver and provide insight into the broader effects of pharmacologic ACC inhibition.


Subject(s)
Acetyl-CoA Carboxylase , Amino Acids , Gluconeogenesis , Liver , Malonyl Coenzyme A , Mice, Knockout , Oxidation-Reduction , Animals , Malonyl Coenzyme A/metabolism , Liver/metabolism , Acetyl-CoA Carboxylase/metabolism , Mice , Amino Acids/metabolism , Male , Pyruvate Carboxylase/metabolism , Citric Acid Cycle , Pyruvic Acid/metabolism , Mice, Inbred C57BL , Fasting/metabolism , Carnitine O-Palmitoyltransferase/metabolism
6.
Biochim Biophys Acta Proteins Proteom ; 1872(5): 141033, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39019246

ABSTRACT

Malonyl-CoA reductase utilizes two equivalents of NADPH to catalyze the reduction of malonyl-CoA to 3-hydroxypropionic acid (3HP). This reaction is part of the carbon fixation pathway in the phototrophic bacterium Chloroflexus aurantiacus. The enzyme is composed of two domains. The C-terminal domain catalyzes the reduction of malonyl-CoA to malonic semialdehyde, while the N-terminal domain catalyzes the reduction of the aldehyde to 3HP. The two domains can be produced independently and retain their enzymatic activity. This report focuses on the kinetic characterization of the C-terminal domain. Initial velocity patterns and inhibition studies showed the kinetic mechanism is ordered with NADPH binding first followed by malonyl-CoA. Malonic semialdehyde is released first, while CoA and NADP+ are released randomly. Analogs of malonyl-CoA showed that the thioester carbon is reduced, while the carboxyl group is needed for proper positioning. The enzyme transfers the pro-S hydrogen of NADPH to malonyl-CoA and pH rate profiles revealed that a residue with a pKa value of about 8.8 must be protonated for activity. Kinetic isotope effects indicated that NADPH is not sticky (that is, NADPH dissociates from the enzyme faster than the rate of product formation) and product release is partially rate-limiting. Moreover, the mechanism is stepwise with the pH dependent step occurring before or after hydride transfer. The findings from this study will aid in the development of an eco-friendly biosynthesis of 3HP which is an industrial chemical used in the production of plastics and adhesives.


Subject(s)
Chloroflexus , Malonyl Coenzyme A , NADP , Kinetics , NADP/metabolism , NADP/chemistry , Malonyl Coenzyme A/metabolism , Chloroflexus/metabolism , Chloroflexus/enzymology , Protein Domains , Alcohol Oxidoreductases/metabolism , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Hydrogen-Ion Concentration , Oxidoreductases , Lactic Acid/analogs & derivatives
7.
Biomolecules ; 14(6)2024 Jun 16.
Article in English | MEDLINE | ID: mdl-38927115

ABSTRACT

Resveratrol, a phenylpropanoid compound, exhibits diverse pharmacological properties, making it a valuable candidate for health and disease management. However, the demand for resveratrol exceeds the capacity of plant extraction methods, necessitating alternative production strategies. Microbial synthesis offers several advantages over plant-based approaches and presents a promising alternative. Yarrowia lipolytica stands out among microbial hosts due to its safe nature, abundant acetyl-CoA and malonyl-CoA availability, and robust pentose phosphate pathway. This study aimed to engineer Y. lipolytica for resveratrol production. The resveratrol biosynthetic pathway was integrated into Y. lipolytica by adding genes encoding tyrosine ammonia lyase from Rhodotorula glutinis, 4-coumarate CoA ligase from Nicotiana tabacum, and stilbene synthase from Vitis vinifera. This resulted in the production of 14.3 mg/L resveratrol. A combination of endogenous and exogenous malonyl-CoA biosynthetic modules was introduced to enhance malonyl-CoA availability. This included genes encoding acetyl-CoA carboxylase 2 from Arabidopsis thaliana, malonyl-CoA synthase, and a malonate transporter protein from Bradyrhizobium diazoefficiens. These strategies increased resveratrol production to 51.8 mg/L. The further optimization of fermentation conditions and the utilization of sucrose as an effective carbon source in YP media enhanced the resveratrol concentration to 141 mg/L in flask fermentation. By combining these strategies, we achieved a titer of 400 mg/L resveratrol in a controlled fed-batch bioreactor. These findings demonstrate the efficacy of Y. lipolytica as a platform for the de novo production of resveratrol and highlight the importance of metabolic engineering, enhancing malonyl-CoA availability, and media optimization for improved resveratrol production.


Subject(s)
Metabolic Engineering , Resveratrol , Sucrose , Yarrowia , Resveratrol/metabolism , Yarrowia/genetics , Yarrowia/metabolism , Metabolic Engineering/methods , Sucrose/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Vitis/microbiology , Vitis/genetics , Vitis/metabolism , Coenzyme A Ligases/metabolism , Coenzyme A Ligases/genetics , Malonyl Coenzyme A/metabolism , Nicotiana/genetics , Nicotiana/metabolism , Nicotiana/microbiology , Rhodotorula/genetics , Rhodotorula/metabolism , Fermentation , Arabidopsis/genetics , Arabidopsis/metabolism , Ammonia-Lyases , Bacterial Proteins
8.
mBio ; 15(5): e0341423, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38572988

ABSTRACT

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.


Subject(s)
Acetyl-CoA Carboxylase , Carbon-Nitrogen Ligases , Chloroflexus , Acetyl-CoA Carboxylase/metabolism , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/chemistry , Carbon-Nitrogen Ligases/metabolism , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/chemistry , Chloroflexus/genetics , Chloroflexus/metabolism , Chloroflexus/enzymology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/enzymology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Biotin/metabolism , Biotin/biosynthesis , Malonyl Coenzyme A/metabolism , Acetyl Coenzyme A/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , Fatty Acid Synthase, Type II
9.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167299, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38878833

ABSTRACT

STING (stimulator of interferon genes) is a critical immunoregulatory protein in sepsis and is regulated by various mechanisms, especially palmitoylation. FASN (fatty acid synthase) is the rate-limiting enzyme to generate cellular palmitic acid (PA) via acetyl-CoA and malonyl-CoA and participates in protein palmitoylation. However, the mechanisms underlying the interaction between STING and FASN have not been completely understood. In this study, STING-knockout mice were used to confirm the pivotal role of STING in sepsis-induced liver injury. Metabolomics confirmed the dyslipidemia in septic mice and patients. The compounds library was screened, revealing that FASN inhibitors exerted a significant inhibitory effect on the STING pathway. Mechanically, the regulatory effect of FASN on the STING pathway was dependent on palmitoylation. Further experiments indicated that the upstream of FASN, malonyl-CoA inhibited STING pathway possibly due to C91 (palmitoylated residue) of STING. Overall, this study reveals a novel paradigm of STING regulation and provides a new perspective on immunity and metabolism.


Subject(s)
Fatty Acid Synthase, Type I , Lipoylation , Macrophages , Malonyl Coenzyme A , Membrane Proteins , Sepsis , Animals , Humans , Male , Mice , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Malonyl Coenzyme A/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Palmitic Acid/pharmacology , Sepsis/metabolism , Sepsis/complications , Sepsis/drug therapy , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL