Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 807
Filter
1.
J Lipid Res ; 64(8): 100407, 2023 08.
Article in English | MEDLINE | ID: mdl-37356666

ABSTRACT

Acetoacetyl-CoA synthetase (AACS) is the key enzyme in the anabolic utilization of ketone bodies (KBs) for denovo lipid synthesis, a process that bypasses citrate and ATP citrate lyase. This review shows that AACS is a highly regulated, cytosolic, and lipogenic enzyme and that many tissues can readily use KBs for denovo lipid synthesis. AACS has a low micromolar Km for acetoacetate, and supply of acetoacetate should not limit its activity in the fed state. In many tissues, AACS appears to be regulated in conjunction with the need for cholesterol, but in adipose tissue, it seems tied to fatty acid synthesis. KBs are readily utilized as substrates for lipid synthesis in lipogenic tissues, including liver, adipose tissue, lactating mammary gland, skin, intestinal mucosa, adrenals, and developing brain. In numerous studied cases, KBs served several-fold better than glucose as substrates for lipid synthesis, and when present, KBs suppressed the utilization of glucose for lipid synthesis. Here, it is hypothesized that a physiological role for the utilization of KBs for lipid synthesis is a metabolic process of lipid interconversion. Fatty acids are converted to KBs in liver, and then, the KBs are utilized to synthesize cholesterol and other long-chain fatty acids in liver and nonhepatic tissues. The conversion of fatty acids to cholesterol via the KBs may be a particularly important example of lipid interconversion. Utilizing KBs for lipid synthesis is glucose sparing and probably is important with low carbohydrate diets. Metabolic situations and tissues where this pathway may be important are discussed.


Subject(s)
Acetoacetates , Lactation , Female , Humans , Acetoacetates/metabolism , Ketone Bodies/metabolism , Fatty Acids , Liver/metabolism , Cholesterol , Glucose
2.
J Leukoc Biol ; 113(6): 577-587, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36999365

ABSTRACT

Neutrophils express many surface receptors that sense environmental changes. One such sensor is FFAR2 (free fatty acid receptor 2), a receptor that detects gut microbiota-derived short-chain fatty acids. As such, FFAR2 has been regarded as a molecular link between metabolism and inflammation. Our recent studies on FFAR2, using its endogenous agonist propionate in combination with allosteric modulators, have identified several novel aspects of FFAR2 regulation. A recent study has also identified the ketone body acetoacetate as an endogenous ligand for mouse FFAR2. Whether human FFAR2 also recognizes acetoacetate and how this recognition modulates human neutrophil functions has not been investigated. In this study, we found that acetoacetate can induce a decrease of cAMP and translocation of ß-arrestin in cells overexpressing FFAR2. In addition, we show that similar to propionate, FFAR2-specific allosteric modulators enhance acetoacetate-induced transient rise in cytosolic calcium, production of reactive oxygen species, and cell migration in human neutrophils. In summary, we demonstrate that human neutrophils recognize the ketone body acetoacetate through FFAR2. Thus, our data further highlight the key role of FFAR2 in inflammation and metabolism.


Subject(s)
Propionates , Receptors, G-Protein-Coupled , Humans , Mice , Animals , Receptors, G-Protein-Coupled/metabolism , Propionates/pharmacology , Neutrophils/metabolism , Acetoacetates/pharmacology , Acetoacetates/metabolism , Ketone Bodies/metabolism , Inflammation/chemically induced , Inflammation/metabolism
3.
J Environ Manage ; 331: 117300, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-36657207

ABSTRACT

Waste activated sludge has been frequently used as mixed substrate to produce polyhydroxyalkanoate (PHA). However, insufficient research on microbial metabolism has led to difficulties in regulating PHA accumulation in mixed microbial cultures (MMCs). To explore the variation of functional genes during domestication and the effect of different pH conditions on metabolic pathways during PHA accumulation, MMCs were domesticated by adding acetate and propionate with aerobic dynamic feeding strategy for 60 days. As the domestication progressed, the microbial community diversity declined and PHA-producing bacteria, Brevundimonas, Dechloromonas and Hyphomonas, were enriched. Through bacterial function prediction by PICRUSt the gene rpoE involved in starvation resistance of bacteria was enriched after the domestication. The pH value of 8.5 was the best condition for PHA accumulation in MMCs, under which a maximum PHA content reached 23.50% and hydroxybutyric (HB)/hydroxyvaleric (HV) reached 2.22. Untargeted metabolomics analysis exhibited that pH conditions of 7 and 8.5 could promote the up-regulation of significant differential metabolites, while higher alkaline conditions caused the inhibition of metabolic activity. Functional annotation showed that pH condition of 8.5 significantly affected Pyrimidine metabolism, resulting in an increase in PHA production. Regarding the pathways of PHA biosynthesis, acetoacetate was found to be significant in the metabolism of hydroxybutyric, and the alkaline condition could restrain the conversion from hydroxybutyric (HB) to the acetoacetate to protect PHB accumulation in MMCs compared with neutral condition. Taken together, the present results can advance the fundamental understanding of metabolic function in PHA accumulation under different pH conditions.


Subject(s)
Polyhydroxyalkanoates , Polyhydroxyalkanoates/chemistry , Polyhydroxyalkanoates/metabolism , Sewage/chemistry , Acetoacetates/metabolism , Metabolomics , Bacteria/genetics , Hydrogen-Ion Concentration , Bioreactors/microbiology
4.
J Biotechnol ; 359: 29-34, 2022 Nov 20.
Article in English | MEDLINE | ID: mdl-36150604

ABSTRACT

Isopropanol has a good potential as a new fuel substitution. In the model biosynthesis pathway of isopropanol synthesis, acetoacetyl-CoA is converted to acetoacetate by acetoacetyl-CoA transferases, which requires an acetate molecule as a substrate. Herein, a novel isopropanol synthesis pathway based on mammalian ketone metabolic pathway was developed. In this pathway, acetoacetyl-CoA is condensed with acetyl-CoA to generate 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) by HMG-CoA synthase, and then catalyzed by HMG-CoA lyase to generate acetoacetate. This process is acetate-independent. Under the same experimental system using glycerol as carbon source, the E. coli strain MG::ISOP1 containing the novel pathway produced 11.7 times more isopropanol than the strain MG::ISOP0 containing the model pathway. The pta-ackA knockout mutant strain MG∆pta-ackA::ISOP1, which reduced the conversion of acetyl-CoA to acetate, further increased the production from 76 mg/L to 360 mg/L. In another strategy, knocking out atoDA to block the acetoacetate degradation pathway in strain MG∆atoDA::ISOP1 increased the production to 680 mg/L. By knocking out both of pta-ackA and atoDA, strain MGΔpta-ackAΔatoDA::ISOP1 produced 964 mg/L of isopropanol, which was 12.7 times that of MG::ISOP1. This study indicated that the novel pathway is competent for isopropanol synthesis, and provides a new perspective for biosynthesis of isopropanol.


Subject(s)
2-Propanol , Escherichia coli , Escherichia coli/genetics , Escherichia coli/metabolism , 2-Propanol/metabolism , Acetoacetates/metabolism , Acetyl Coenzyme A/metabolism , Coenzyme A-Transferases/metabolism , Hydroxymethylglutaryl-CoA Synthase/genetics , Hydroxymethylglutaryl-CoA Synthase/metabolism , Glycerol/metabolism , Acetates/metabolism , Carbon/metabolism
5.
J Biol Chem ; 298(5): 101884, 2022 05.
Article in English | MEDLINE | ID: mdl-35367206

ABSTRACT

2-Ketopropyl-coenzyme M oxidoreductase/carboxylase (2-KPCC) is a member of the flavin and cysteine disulfide containing oxidoreductase family (DSOR) that catalyzes the unique reaction between atmospheric CO2 and a ketone/enolate nucleophile to generate acetoacetate. However, the mechanism of this reaction is not well understood. Here, we present evidence that 2-KPCC, in contrast to the well-characterized DSOR enzyme glutathione reductase, undergoes conformational changes during catalysis. Using a suite of biophysical techniques including limited proteolysis, differential scanning fluorimetry, and native mass spectrometry in the presence of substrates and inhibitors, we observed conformational differences between different ligand-bound 2-KPCC species within the catalytic cycle. Analysis of site-specific amino acid variants indicated that 2-KPCC-defining residues, Phe501-His506, within the active site are important for transducing these ligand induced conformational changes. We propose that these conformational changes promote substrate discrimination between H+ and CO2 to favor the metabolically preferred carboxylation product, acetoacetate.


Subject(s)
Carboxy-Lyases , Mesna , Acetoacetates/metabolism , Carbon Dioxide/metabolism , Carboxy-Lyases/metabolism , Catalysis , Ligands , Mesna/metabolism , Oxidoreductases/metabolism , Xanthobacter/metabolism
6.
ACS Sens ; 6(11): 3967-3977, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34761912

ABSTRACT

Cellular redox is intricately linked to energy production and normal cell function. Although the redox states of mitochondria and cytosol are connected by shuttle mechanisms, the redox state of mitochondria may differ from redox in the cytosol in response to stress. However, detecting these differences in functioning tissues is difficult. Here, we employed 13C magnetic resonance spectroscopy (MRS) and co-polarized [1-13C]pyruvate and [1,3-13C2]acetoacetate ([1,3-13C2]AcAc) to monitor production of hyperpolarized (HP) lactate and ß-hydroxybutyrate as indicators of cytosolic and mitochondrial redox, respectively. Isolated rat hearts were examined under normoxic conditions, during low-flow ischemia, and after pretreatment with either aminooxyacetate (AOA) or rotenone. All interventions were associated with an increase in [Pi]/[ATP] measured by 31P NMR. In well-oxygenated untreated hearts, rapid conversion of HP [1-13C]pyruvate to [1-13C]lactate and [1,3-13C2]AcAc to [1,3-13C2]ß-hydroxybutyrate ([1,3-13C2]ß-HB) was readily detected. A significant increase in HP [1,3-13C2]ß-HB but not [1-13C]lactate was observed in rotenone-treated and ischemic hearts, consistent with an increase in mitochondrial NADH but not cytosolic NADH. AOA treatments did not alter the productions of HP [1-13C]lactate or [1,3-13C2]ß-HB. This study demonstrates that biomarkers of mitochondrial and cytosolic redox may be detected simultaneously in functioning tissues using co-polarized [1-13C]pyruvate and [1,3-13C2]AcAc and 13C MRS and that changes in mitochondrial redox may precede changes in cytosolic redox.


Subject(s)
Acetoacetates , Pyruvic Acid , Acetoacetates/metabolism , Animals , Cytosol/metabolism , Lactic Acid , Mitochondria/metabolism , Oxidation-Reduction , Pyruvic Acid/metabolism , Rats
7.
Biochem Biophys Res Commun ; 585: 61-67, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34794035

ABSTRACT

Leucine, isoleucine and valine, known as branched chain amino acids (BCAAs), have been reported to be degraded by different cancer cells, and their biodegradation pathways have been suggested as anticancer targets. However, the mechanisms by which the degradation of BCAAs could support the growth of cancer cells remains unclear. In this work, 13C experiments have been carried out in order to elucidate the metabolic role of BCAA degradation in two breast cancer cell lines (MCF-7 and BCC). The results revealed that up to 36% of the energy production via respiration by MCF-7 cells was supported by the degradation of BCAAs. Also, 67% of the mevalonate (the precursor of cholesterol) synthesized by the cells was coming from the degradation of leucine. The results were lower for BCC cells (14 and 30%, respectively). The non-tumorigenic epythelial cell line MCF-10A was used as a control, showing that 10% of the mitochondrial acetyl-CoA comes from the degradation of BCAAs and no mevalonate production. Metabolic flux analysis around the mevalonate node, also revealed that significant amounts of acetoacetate are being produced from BCAA derived carbon, which could be at the source of lipid synthesis. From these results we can conclude that the degradation of BCAAs is an important energy and carbon source for the proliferation of some cancer cells and its therapeutic targeting could be an interesting option.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Breast Neoplasms/metabolism , Energy Metabolism , Metabolic Flux Analysis/methods , Mevalonic Acid/metabolism , Acetoacetates/metabolism , Algorithms , Breast Neoplasms/pathology , Carbon/metabolism , Cell Line , Cell Line, Tumor , Citric Acid Cycle , Female , Humans , Leucine/metabolism , MCF-7 Cells , Metabolic Networks and Pathways , Mitochondria/metabolism , Models, Biological
8.
Biotechnol Bioeng ; 118(11): 4278-4289, 2021 11.
Article in English | MEDLINE | ID: mdl-34289076

ABSTRACT

Whole-cell biosensors hold potential in a variety of industrial, medical, and environmental applications. These biosensors can be constructed through the repurposing of bacterial sensing mechanisms, including the common two-component system (TCS). Here we report on the construction of a range of novel biosensors that are sensitive to acetoacetate, a molecule that plays a number of roles in human health and biology. These biosensors are based on the AtoSC TCS. An ordinary differential equation model to describe the action of the AtoSC TCS was developed and sensitivity analysis of this model used to help inform biosensor design. The final collection of biosensors constructed displayed a range of switching behaviours at physiologically relevant acetoacetate concentrations and can operate in several Escherichia coli host strains. It is envisaged that these biosensor strains will offer an alternative to currently available commercial strip tests and, in future, may be adopted for more complex in vivo or industrial monitoring applications.


Subject(s)
Acetoacetates/metabolism , Biosensing Techniques , Escherichia coli Proteins , Escherichia coli , Gene Expression Regulation, Bacterial , Acetoacetates/analysis , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Operon
9.
Int J Mol Sci ; 22(9)2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33946157

ABSTRACT

The metabolic ratios lactate/pyruvate and ß-hydroxybutyrate/acetoacetate are considered valuable tools to evaluate the in vivo redox cellular state by estimating the free NAD+/NADH in cytoplasm and mitochondria, respectively. The aim of the current study was to validate a gas-chromatography mass spectrometry method for simultaneous determination of the four metabolites in plasma and liver tissue. The procedure included an o-phenylenediamine microwave-assisted derivatization, followed by liquid-liquid extraction with ethyl acetate and silylation with bis(trimethylsilyl)trifluoroacetamide:trimethylchlorosilane 99:1. The calibration curves presented acceptable linearity, with a limit of quantification of 0.001 mM for pyruvate, ß-hydroxybutyrate and acetoacetate and of 0.01 mM for lactate. The intra-day and inter-day accuracy and precision were within the European Medicines Agency's Guideline specifications. No significant differences were observed in the slope coefficient of three-point standard metabolite-spiked curves in plasma or liver and water, and acceptable recoveries were obtained in the metabolite-spiked samples. Applicability of the method was tested in precision-cut liver rat slices and also in HepG2 cells incubated under different experimental conditions challenging the redox state. In conclusion, the validated method presented good sensitivity, specificity and reproducibility in the quantification of lactate/pyruvate and ß-hydroxybutyrate/acetate metabolites and may be useful in the evaluation of in vivo redox states.


Subject(s)
3-Hydroxybutyric Acid/metabolism , Acetoacetates/metabolism , Gas Chromatography-Mass Spectrometry , Lactates/metabolism , Pyruvates/metabolism , 3-Hydroxybutyric Acid/analysis , 3-Hydroxybutyric Acid/blood , Acetoacetates/analysis , Acetoacetates/blood , Animals , Female , Gas Chromatography-Mass Spectrometry/methods , Hep G2 Cells , Humans , Lactates/analysis , Lactates/blood , Limit of Detection , Liver/chemistry , Liver/metabolism , Oxidation-Reduction , Pyruvates/analysis , Pyruvates/blood , Rats, Wistar
10.
Int J Mol Sci ; 22(6)2021 Mar 13.
Article in English | MEDLINE | ID: mdl-33805788

ABSTRACT

Persistent chronic liver diseases increase the scar formation and extracellular matrix accumulation that further progress to liver fibrosis and cirrhosis. Nevertheless, there is no antifibrotic therapy to date. The ketogenic diet is composed of high fat, moderate to low-protein, and very low carbohydrate content. It is mainly used in epilepsy and Alzheimer's disease. However, the effects of the ketogenic diet on liver fibrosis remains unknown. Through ketogenic diet consumption, ß-hydroxybutyrate (bHB) and acetoacetate (AcAc) are two ketone bodies that are mainly produced in the liver. It is reported that bHB and AcAc treatment decreases cancer cell proliferation and promotes apoptosis. However, the influence of bHB and AcAc in hepatic stellate cell (HSC) activation and liver fibrosis are still unclear. Therefore, this study aimed to investigate the effect of the ketogenic diet and ketone bodies in affecting liver fibrosis progression. Our study revealed that feeding a high-fat ketogenic diet increased cholesterol accumulation in the liver, which further enhanced the carbon tetrachloride (CCl4)- and thioacetamide (TAA)-induced liver fibrosis. In addition, more severe liver inflammation and the loss of hepatic antioxidant and detoxification ability were also found in ketogenic diet-fed fibrotic mouse groups. However, the treatment with ketone bodies (bHB and AcAc) did not suppress transforming growth factor-ß (TGF-ß)-induced HSC activation, platelet-derived growth factor (PDGF)-BB-triggered proliferation, and the severity of CCl4-induced liver fibrosis in mice. In conclusion, our study demonstrated that feeding a high-fat ketogenic diet may trigger severe steatohepatitis and thereby promote liver fibrosis progression. Since a different ketogenic diet composition may exert different metabolic effects, more evidence is necessary to clarify the effects of a ketogenic diet on disease treatment.


Subject(s)
3-Hydroxybutyric Acid/pharmacology , Acetoacetates/pharmacology , Cholesterol/biosynthesis , Diet, Ketogenic/adverse effects , Liver Cirrhosis/metabolism , Liver/drug effects , 3-Hydroxybutyric Acid/biosynthesis , Acetoacetates/metabolism , Actins/genetics , Actins/metabolism , Animals , Becaplermin/pharmacology , Carbon Tetrachloride/administration & dosage , Catalase/genetics , Catalase/metabolism , Cell Proliferation/drug effects , Cholesterol/blood , Collagen Type I/genetics , Collagen Type I/metabolism , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Desmin/genetics , Desmin/metabolism , Disease Progression , Gene Expression Regulation/drug effects , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Primary Cell Culture , Severity of Illness Index , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Thioacetamide/administration & dosage , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/pharmacology
11.
Sci Rep ; 10(1): 13714, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792583

ABSTRACT

Fumarylacetoacetate hydrolase (FAH) catalyzes the final step in Tyr degradation pathway essential to animals but not well understood in plants. Previously, we found that mutation of SSCD1 encoding Arabidopsis FAH causes cell death under short day, which uncovered an important role of Tyr degradation pathway in plants. Since phytohormones salicylic acid (SA) and jasmonate (JA) are involved in programmed cell death, in this study, we investigated whether sscd1 cell death is related to SA and JA, and found that (1) it is accompanied by up-regulation of JA- and SA-inducible genes as well as accumulation of JA but not SA; (2) it is repressed by breakdown of JA signaling but not SA signaling; (3) the up-regulation of reactive oxygen species marker genes in sscd1 is repressed by breakdown of JA signaling; (4) treatment of wild-type Arabidopsis with succinylacetone, an abnormal metabolite caused by loss of FAH, induces expression of JA-inducible genes whereas treatment with JA induces expression of some Tyr degradation genes with dependence of JA signaling. These results demonstrated that cell death resulted from loss of FAH in Arabidopsis is related to JA but not SA, and suggested that JA signaling positively regulates sscd1 cell death by up-regulating Tyr degradation.


Subject(s)
Arabidopsis Proteins/antagonists & inhibitors , Arabidopsis/growth & development , Cell Death , Cyclopentanes/pharmacology , Gene Expression Regulation, Plant/drug effects , Hydrolases/antagonists & inhibitors , Oxylipins/pharmacology , Salicylic Acid/pharmacology , Acetoacetates/metabolism , Anti-Infective Agents/pharmacology , Arabidopsis/drug effects , Arabidopsis/enzymology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Hydrolases/genetics , Hydrolases/metabolism , Plant Growth Regulators/pharmacology , Reactive Oxygen Species , Signal Transduction
12.
Biochem J ; 477(11): 2027-2038, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32497192

ABSTRACT

Alkenes and ketones are two classes of ubiquitous, toxic organic compounds in natural environments produced in several biological and anthropogenic processes. In spite of their toxicity, these compounds are utilized as primary carbon and energy sources or are generated as intermediate metabolites in the metabolism of other compounds by many diverse bacteria. The aerobic metabolism of some of the smallest and most volatile of these compounds (propylene, acetone, isopropanol) involves novel carboxylation reactions resulting in a common product acetoacetate. Propylene is metabolized in a four-step pathway involving five enzymes where the penultimate step is a carboxylation reaction catalyzed by a unique disulfide oxidoreductase that couples reductive cleavage of a thioether linkage with carboxylation to produce acetoacetate. The carboxylation of isopropanol begins with conversion to acetone via an alcohol dehydrogenase. Acetone is converted to acetoacetate in a single step by an acetone carboxylase which couples the hydrolysis of MgATP to the activation of both acetone and bicarbonate, generating highly reactive intermediates that are condensed into acetoacetate at a Mn2+ containing the active site. Acetoacetate is then utilized in central metabolism where it is readily converted to acetyl-coenzyme A and subsequently converted into biomass or utilized in energy metabolism via the tricarboxylic acid cycle. This review summarizes recent structural and biochemical findings that have contributed significant insights into the mechanism of these two unique carboxylating enzymes.


Subject(s)
Acetone/metabolism , Alkenes/metabolism , Bacteria/metabolism , 2-Propanol/metabolism , Acetoacetates/metabolism , Acetyl Coenzyme A/metabolism , Bicarbonates/metabolism , Catalysis , Citric Acid Cycle/physiology
13.
Nutrients ; 12(3)2020 Mar 17.
Article in English | MEDLINE | ID: mdl-32192146

ABSTRACT

Ketone bodies (KBs), comprising ß-hydroxybutyrate, acetoacetate and acetone, are a set of fuel molecules serving as an alternative energy source to glucose. KBs are mainly produced by the liver from fatty acids during periods of fasting, and prolonged or intense physical activity. In diabetes, mainly type-1, ketoacidosis is the pathological response to glucose malabsorption. Endogenous production of ketone bodies is promoted by consumption of a ketogenic diet (KD), a diet virtually devoid of carbohydrates. Despite its recently widespread use, the systemic impact of KD is only partially understood, and ranges from physiologically beneficial outcomes in particular circumstances to potentially harmful effects. Here, we firstly review ketone body metabolism and molecular signaling, to then link the understanding of ketone bodies' biochemistry to controversies regarding their putative or proven medical benefits. We overview the physiological consequences of ketone bodies' consumption, focusing on (i) KB-induced histone post-translational modifications, particularly ß-hydroxybutyrylation and acetylation, which appears to be the core epigenetic mechanisms of activity of ß-hydroxybutyrate to modulate inflammation; (ii) inflammatory responses to a KD; (iii) proven benefits of the KD in the context of neuronal disease and cancer; and (iv) consequences of the KD's application on cardiovascular health and on physical performance.


Subject(s)
Diabetes Mellitus, Type 1 , Diet, Ketogenic , Epigenesis, Genetic , Neoplasms , Nervous System Diseases , 3-Hydroxybutyric Acid/metabolism , Acetoacetates/metabolism , Animals , Diabetes Mellitus, Type 1/diet therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Epigenomics , Humans , Ketone Bodies/genetics , Ketone Bodies/metabolism , Ketosis/diet therapy , Ketosis/genetics , Ketosis/metabolism , Ketosis/pathology , Metabolomics , Neoplasms/diet therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nervous System Diseases/diet therapy , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Nervous System Diseases/pathology
14.
Biosci Rep ; 40(3)2020 03 27.
Article in English | MEDLINE | ID: mdl-32068790

ABSTRACT

FAH domain containing protein 1 (FAHD1) is a mammalian mitochondrial protein, displaying bifunctionality as acylpyruvate hydrolase (ApH) and oxaloacetate decarboxylase (ODx) activity. We report the crystal structure of mouse FAHD1 and structural mapping of the active site of mouse FAHD1. Despite high structural similarity with human FAHD1, a rabbit monoclonal antibody (RabMab) could be produced that is able to recognize mouse FAHD1, but not the human form, whereas a polyclonal antibody recognized both proteins. Epitope mapping in combination with our deposited crystal structures revealed that the epitope overlaps with a reported SIRT3 deacetylation site in mouse FAHD1.


Subject(s)
Hydrolases/genetics , Acetoacetates/metabolism , Animals , Carboxy-Lyases/metabolism , Catalytic Domain , Crystallography, X-Ray , Epitope Mapping/methods , Humans , Hydrolases/chemistry , Hydrolases/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Structure-Activity Relationship
15.
Biochim Biophys Acta Mol Basis Dis ; 1866(6): 165739, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32084511

ABSTRACT

The ketone bodies, d-ß-hydroxybutyrate and acetoacetate, are soluble 4-carbon compounds derived principally from fatty acids, that can be metabolised by many oxidative tissues, including heart, in carbohydrate-depleted conditions as glucose-sparing energy substrates. They also have important signalling functions, acting through G-protein coupled receptors and histone deacetylases to regulate metabolism and gene expression including that associated with anti-oxidant activity. Their concentration, and hence availability, increases in diabetes mellitus and heart failure. Whilst known to be substrates for ATP production, especially in starvation, their role(s) in the heart, and in heart disease, is uncertain. Recent evidence, reviewed here, indicates that increased ketone body metabolism is a feature of heart failure, and is accompanied by other changes in substrate selection. Whether the change in myocardial ketone body metabolism is adaptive or maladaptive is unknown, but it offers the possibility of using exogenous ketones to treat the failing heart.


Subject(s)
Heart Failure/metabolism , Ketone Bodies/metabolism , Ketones/metabolism , Myocardium/metabolism , Acetoacetates/metabolism , Fatty Acids/metabolism , Glucose/metabolism , Heart Failure/pathology , Humans , Myocardium/pathology
16.
NMR Biomed ; 32(6): e4091, 2019 06.
Article in English | MEDLINE | ID: mdl-30968985

ABSTRACT

Mitochondrial dysfunction is considered to be an important component of many metabolic diseases yet there is no reliable imaging biomarker for monitoring mitochondrial damage in vivo. A large prior literature on inter-conversion of ß-hydroxybutyrate and acetoacetate indicates that the process is mitochondrial and that the ratio reflects a specifically mitochondrial redox state. Therefore, the conversion of [1,3-13 C]acetoacetate to [1,3-13 C]ß-hydroxybutyrate is expected to be sensitive to the abnormal redox state present in dysfunctional mitochondria. In this study, we examined the conversion of hyperpolarized (HP) 13 C-acetoacetate (AcAc) to 13 C-ß-hydroxybutyrate (ß-HB) as a potential imaging biomarker for mitochondrial redox and dysfunction in perfused rat hearts. Conversion of HP-AcAc to ß-HB was investigated using 13 C magnetic resonance spectroscopy in Langendorff-perfused rat hearts in four groups: control, global ischemic reperfusion, low-flow ischemic, and rotenone (mitochondrial complex-I inhibitor)-treated hearts. We observed that more ß-HB was produced from AcAc in ischemic hearts and the hearts exposed to complex I inhibitor rotenone compared with controls, consistent with the accumulation of excess mitochondrial NADH. The increase in ß-HB, as detected by 13 C MRS, was validated by a direct measure of tissue ß-HB by 1 H nuclear magnetic resonance in tissue extracts. The redox ratio, NAD+ /NADH, measured by enzyme assays of homogenized tissue, also paralleled production of ß-HB from AcAc. Transmission electron microscopy of tissues provided direct evidence for abnormal mitochondrial structure in each ischemic tissue model. The results suggest that conversion of HP-AcAc to HP-ß-HB detected by 13 C-MRS may serve as a useful diagnostic marker of mitochondrial redox and dysfunction in heart tissue in vivo.


Subject(s)
3-Hydroxybutyric Acid/metabolism , Acetoacetates/metabolism , Carbon Isotopes/metabolism , Heart/physiopathology , Magnetic Resonance Spectroscopy , Mitochondria/metabolism , Animals , Carbon-13 Magnetic Resonance Spectroscopy , Freezing , Hemodynamics , Male , Mitochondria/ultrastructure , Myocardium/metabolism , Myocardium/ultrastructure , NAD/metabolism , Oxidation-Reduction , Perfusion , Proton Magnetic Resonance Spectroscopy , Rats, Sprague-Dawley
17.
Sci Rep ; 9(1): 3402, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30833594

ABSTRACT

Dysregulation in NAD+/NADH levels is associated with increased cell division and elevated levels of reactive oxygen species in rapidly proliferating cancer cells. Conversion of the ketone body acetoacetate (AcAc) to ß-hydroxybutyrate (ß-HB) by the mitochondrial enzyme ß-hydroxybutyrate dehydrogenase (BDH) depends upon NADH availability. The ß-HB-to-AcAc ratio is therefore expected to reflect mitochondrial redox. Previous studies reported the potential of hyperpolarized 13C-AcAc to monitor mitochondrial redox in cells, perfused organs and in vivo. However, the ability of hyperpolarized 13C-AcAc to cross the blood brain barrier (BBB) and its potential to monitor brain metabolism remained unknown. Our goal was to assess the value of hyperpolarized [1,3-13C2]AcAc in healthy and tumor-bearing mice in vivo. Following hyperpolarized [1,3-13C2]AcAc injection, production of [1,3-13C2]ß-HB was detected in normal and tumor-bearing mice. Significantly higher levels of [1-13C]AcAc and lower [1-13C]ß-HB-to-[1-13C]AcAc ratios were observed in tumor-bearing mice. These results were consistent with decreased BDH activity in tumors and associated with increased total cellular NAD+/NADH. Our study confirmed that AcAc crosses the BBB and can be used for monitoring metabolism in the brain. It highlights the potential of AcAc for future clinical translation and its potential utility for monitoring metabolic changes associated with glioma, and other neurological disorders.


Subject(s)
Acetoacetates/metabolism , Brain/metabolism , Glioma/metabolism , Acetoacetates/chemistry , Animals , Female , Magnetic Resonance Spectroscopy , Mice , Mitochondria/metabolism , Oxidation-Reduction , Spectrophotometry
18.
Cell Metab ; 29(2): 383-398.e7, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30449686

ABSTRACT

Metabolic plasticity has been linked to polarized macrophage function, but mechanisms connecting specific fuels to tissue macrophage function remain unresolved. Here we apply a stable isotope tracing, mass spectrometry-based untargeted metabolomics approach to reveal the metabolome penetrated by hepatocyte-derived glucose and ketone bodies. In both classically and alternatively polarized macrophages, [13C]acetoacetate (AcAc) labeled ∼200 chemical features, but its reduced form D-[13C]ß-hydroxybutyrate (D-ßOHB) labeled almost none. [13C]glucose labeled ∼500 features, and while unlabeled AcAc competed with only ∼15% of them, the vast majority required the mitochondrial enzyme succinyl-coenzyme A-oxoacid transferase (SCOT). AcAc carbon labeled metabolites within the cytoplasmic glycosaminoglycan pathway, which regulates tissue fibrogenesis. Accordingly, livers of mice lacking SCOT in macrophages were predisposed to accelerated fibrogenesis. Exogenous AcAc, but not D-ßOHB, ameliorated diet-induced hepatic fibrosis. These data support a hepatocyte-macrophage ketone shuttle that segregates AcAc from D-ßOHB, coordinating the fibrogenic response to hepatic injury via mitochondrial metabolism in tissue macrophages.


Subject(s)
3-Hydroxybutyric Acid/metabolism , Acetoacetates/metabolism , Hepatocytes/metabolism , Liver Cirrhosis, Experimental/metabolism , Macrophages/metabolism , Mitochondria/metabolism , Animals , Hepatocytes/pathology , Macrophages/cytology , Mice , Mice, Inbred C57BL
19.
Biochemistry ; 57(49): 6757-6761, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30472832

ABSTRACT

The temperature dependence of psychrophilic and mesophilic ( R)-3-hydroxybutyrate dehydrogenase steady-state rates yields nonlinear and linear Eyring plots, respectively. Solvent viscosity effects and multiple- and single-turnover pre-steady-state kinetics demonstrate that while product release is rate-limiting at high temperatures for the psychrophilic enzyme, either interconversion between enzyme-substrate and enzyme-product complexes or a step prior to it limits the rate at low temperatures. Unexpectedly, a similar change in the rate-limiting step is observed with the mesophilic enzyme, where a step prior to chemistry becomes rate-limiting at low temperatures. This observation may have implications for past and future interpretations of temperature-rate profiles.


Subject(s)
Hydroxybutyrate Dehydrogenase/chemistry , Hydroxybutyrate Dehydrogenase/metabolism , Acetoacetates/metabolism , Acinetobacter baumannii/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Biocatalysis , Kinetics , Linear Models , Models, Biological , Psychrobacter/enzymology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Solvents , Substrate Specificity , Temperature , Valerates/metabolism , Viscosity
20.
J Oleo Sci ; 67(10): 1235-1246, 2018.
Article in English | MEDLINE | ID: mdl-30305556

ABSTRACT

Terpene synthase (TPS) genes were isolated and functionally characterized from three traditional edible plants, Acanthopanax sciadophylloides ("Koshiabura") and Acanthopanax sieboldianus ("Himeukogi"), belonging to the family Araliaceae, and Curcuma zedoaria (zedoary, "Gajutsu"), belonging to the family Zingiberaceae. These plants emit characteristic fragrances and are used for traditional foods and folk medicines. From their fragrant tissues, i.e., sprouts of Araliaceae plants and developing rhizomes of zedoary, total RNAs were extracted and reverse transcribed. The resultant cDNAs were used for degenerate PCR followed by rapid amplification of cDNA ends. From the contig sequences obtained, full-length Tps genes were amplified by PCR with newly synthesized primer sets. The isolated full-length genes were introduced into engineered Escherichia coli cells, which can utilize acetoacetate to synthesize farnesyl diphosphate, the substrate for TPSs, through the mevalonate pathway. TPS products synthesized in the transformed E. coli cells were analysed by gas chromatography-mass spectrometry, nuclear magnetic resonance, and optical rotation. Consequently, the isolated Tps genes were found to encode ß-caryophyllene synthase, germacrene D synthase, linalool/(3S)-(+)-nerolidol synthase, ß-eudesmol synthase, and germacrene B synthase. These results lead us to expect that some of the effective ingredients in folk medicines are volatile terpenes and that intake of traditional foods including these edible plants would have some positive effects on our health.


Subject(s)
Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/isolation & purification , Curcuma/enzymology , Curcuma/genetics , Eleutherococcus/enzymology , Eleutherococcus/genetics , Plants, Edible/enzymology , Plants, Edible/genetics , Acetoacetates/metabolism , Curcuma/chemistry , DNA, Complementary , Eleutherococcus/chemistry , Escherichia coli/metabolism , Mevalonic Acid/metabolism , Plants, Edible/chemistry , Polyisoprenyl Phosphates/metabolism , Polymerase Chain Reaction , RNA, Plant/isolation & purification , Sesquiterpenes/metabolism , Terpenes , Volatile Organic Compounds
SELECTION OF CITATIONS
SEARCH DETAIL
...