Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 327
Filter
1.
Int J Mol Sci ; 24(6)2023 Mar 14.
Article in English | MEDLINE | ID: mdl-36982602

ABSTRACT

Cholesterol is a key component of all mammalian cell membranes. Disruptions in cholesterol metabolism have been observed in the context of various diseases, including neurodegenerative disorders such as Alzheimer's disease (AD). The genetic and pharmacological blockade of acyl-CoA:cholesterol acyltransferase 1/sterol O-acyltransferase 1 (ACAT1/SOAT1), a cholesterol storage enzyme found on the endoplasmic reticulum (ER) and enriched at the mitochondria-associated ER membrane (MAM), has been shown to reduce amyloid pathology and rescue cognitive deficits in mouse models of AD. Additionally, blocking ACAT1/SOAT1 activity stimulates autophagy and lysosomal biogenesis; however, the exact molecular connection between the ACAT1/SOAT1 blockade and these observed benefits remain unknown. Here, using biochemical fractionation techniques, we observe cholesterol accumulation at the MAM which leads to ACAT1/SOAT1 enrichment in this domain. MAM proteomics data suggests that ACAT1/SOAT1 inhibition strengthens the ER-mitochondria connection. Confocal and electron microscopy confirms that ACAT1/SOAT1 inhibition increases the number of ER-mitochondria contact sites and strengthens this connection by shortening the distance between these two organelles. This work demonstrates how directly manipulating local cholesterol levels at the MAM can alter inter-organellar contact sites and suggests that cholesterol buildup at the MAM is the impetus behind the therapeutic benefits of ACAT1/SOAT1 inhibition.


Subject(s)
Alzheimer Disease , Cholesterol , Animals , Mice , Alzheimer Disease/metabolism , Cholesterol/metabolism , Endoplasmic Reticulum/metabolism , Mammals/metabolism , Mitochondria/metabolism , Sterols/metabolism , Acetyl-CoA C-Acyltransferase/metabolism , Sterol O-Acyltransferase/metabolism
2.
J Pediatr Endocrinol Metab ; 35(9): 1194-1200, 2022 Sep 27.
Article in English | MEDLINE | ID: mdl-35850931

ABSTRACT

OBJECTIVES: Mitochondrial acetoacetyl-CoA thiolase (beta-ketothiolase, T2) is necessary for the catabolism of ketone bodies andisoleucine. T2 deficiency is an autosomal recessive metabolic disorder caused by variant in the ACAT1 gene. In this report, we describe two novel ACAT1 variant identified in a Chinese family. CASE PRESENTATION: The 9-month-old male proband was admitted to the pediatric intensive care unit for altered consciousness. At the time of admission, the patient had acidosis, drowsiness, and respiratory failure. Both urine organic acid analyses and LC-MS/MS suggested T2 deficiency. Novel compound heterozygous variant (c.871G>C and c.1016_1017del) in the ACAT1 gene were detected in the proband by WES and verified through direct sequencing. Family analysis demonstrated that the first variant was transmitted from his father and the second variant was from his mother, indicating autosomal recessive inheritance. This report is the first to describe the association of these variant with T2 deficiency based on genetic testing. Although these variant were identified in the patient's elder sister and elder brother, they continue to be asymptomatic. CONCLUSIONS: We identified two novel ACAT1 variants associated with T2 deficiency. The identification expands the spectrum of known variant linked to the disorder.


Subject(s)
Acetyl-CoA C-Acetyltransferase , Tandem Mass Spectrometry , Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Acetyl-CoA C-Acyltransferase/deficiency , Acetyl-CoA C-Acyltransferase/genetics , Acetyl-CoA C-Acyltransferase/metabolism , Aged , Amino Acid Metabolism, Inborn Errors , Child , Chromatography, Liquid , Humans , Infant , Male
3.
Appl Microbiol Biotechnol ; 106(8): 3021-3032, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35451630

ABSTRACT

Poly[(R)-3-hydroxybutyrate-co-(R)-3-hydroxyhexanoate] (PHBHHx) is a type of biopolyester of the polyhydroxyalkanoate group (PHA). Due to a wide range of properties resulting from the alteration of the (R)-3-hydroxyhexanoate (3HHx) composition, PHBHHx is getting a lot of attention as a substitute to conventional plastic materials for various applications. Cupriavidus necator H16 is the most promising PHA producer and has been genetically engineered to produce PHBHHx efficiently for many years. Nevertheless, the role of individual genes involved in PHBHHx biosynthesis is not well elaborated. C. necator H16 possesses six potential physiologically active ß-ketothiolase genes identified by transcriptome analysis, i.e., phaA, bktB, bktC (h16_A0170), h16_A0462, h16_A1528, and h16_B0759. In this study, we focused on the functionality of these genes in vivo in relation to 3HHx monomer supply. Gene deletion experiments identified BktB and H16_A1528 as important ß-ketothiolases for C6 metabolism in ß-oxidation. Furthermore, in the bktB/h16_A1528 double-deletion strain, the proportion of 3HHx composition of PHBHHx produced from sugar was very low, whereas that from plant oil was significantly higher. In fact, the proportion reached 36.2 mol% with overexpression of (R)-specifc enoyl-CoA hydratase (PhaJ) and PHA synthase. Furthermore, we demonstrated high-density production (196 g/L) of PHBHHx with high 3HHx (32.5 mol%) by fed-batch fermentation with palm kernel oil. The PHBHHx was amorphous according to the differential scanning calorimetry analysis. KEY POINTS: • Role of six ß-ketothiolases in PHBHHx biosynthesis was investigated in vivo. • Double-deletion of bktB/h16_A1528 results in high 3HHx composition with plant oil. • Amorphous PHBHHx with 32.5 mol% 3HHx was produced in high density by jar fermenter.


Subject(s)
Cupriavidus necator , Polyhydroxyalkanoates , Acetyl-CoA C-Acyltransferase/genetics , Acetyl-CoA C-Acyltransferase/metabolism , Cupriavidus necator/genetics , Cupriavidus necator/metabolism , Hydroxybutyrates/metabolism , Plant Oils/metabolism , Polyhydroxyalkanoates/metabolism
4.
Angew Chem Int Ed Engl ; 61(1): e202111054, 2022 01 03.
Article in English | MEDLINE | ID: mdl-34664348

ABSTRACT

Many existing in vitro biosystems harness power from the chemical energy contained in substrates and co-substrates, and light or electric energy provided from abiotic parts, leading to a compromise in atom economy, incompatibility between biological and abiotic parts, and most importantly, incapability to spatiotemporally co-regenerate ATP and NADPH. In this study, we developed a light-powered in vitro biosystem for poly(3-hydroxybutyrate) (PHB) synthesis using natural thylakoid membranes (TMs) to regenerate ATP and NADPH for a five-enzyme cascade. Through effective coupling of cofactor regeneration and mass conversion, 20 mM PHB was yielded from 50 mM sodium acetate with a molar conversion efficiency of carbon of 80.0 % and a light-energy conversion efficiency of 3.04 %, which are much higher than the efficiencies of similar in vitro PHB synthesis biosystems. This suggests the promise of installing TMs as a green engine to drive more enzyme cascades.


Subject(s)
Acetyl Coenzyme A/metabolism , Acetyl-CoA C-Acyltransferase/metabolism , Acyltransferases/metabolism , Alcohol Oxidoreductases/metabolism , Hydroxybutyrates/metabolism , Phosphotransferases/metabolism , Polyesters/metabolism , Acetyl Coenzyme A/chemistry , Acetyl-CoA C-Acyltransferase/chemistry , Acyltransferases/chemistry , Alcohol Oxidoreductases/chemistry , Hydroxybutyrates/chemistry , Light , Phosphotransferases/chemistry , Polyesters/chemistry
5.
Nat Chem ; 13(12): 1178-1185, 2021 12.
Article in English | MEDLINE | ID: mdl-34811478

ABSTRACT

Living systems provide a promising approach to chemical synthesis, having been optimized by evolution to convert renewable carbon sources, such as glucose, into an enormous range of small molecules. However, a large number of synthetic structures can still be difficult to obtain solely from cells, such as unsubstituted hydrocarbons. In this work, we demonstrate the use of a dual cellular-heterogeneous catalytic strategy to produce olefins from glucose using a selective hydrolase to generate an activated intermediate that is readily deoxygenated. Using a new family of iterative thiolase enzymes, we genetically engineered a microbial strain that produces 4.3 ± 0.4 g l-1 of fatty acid from glucose with 86% captured as 3-hydroxyoctanoic and 3-hydroxydecanoic acids. This 3-hydroxy substituent serves as a leaving group that enables heterogeneous tandem decarboxylation-dehydration routes to olefinic products on Lewis acidic catalysts without the additional redox input required for enzymatic or chemical deoxygenation of simple fatty acids.


Subject(s)
Alkenes/chemical synthesis , Fatty Acids/chemistry , Glucose/metabolism , Acetyl-CoA C-Acyltransferase/chemistry , Acetyl-CoA C-Acyltransferase/metabolism , Bacteria/enzymology , Bacteria/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Catalysis , Decarboxylation , Enoyl-CoA Hydratase/chemistry , Enoyl-CoA Hydratase/metabolism , Fatty Acid Desaturases/chemistry , Fatty Acid Desaturases/metabolism , Fatty Acids/biosynthesis , Lewis Acids/chemistry , Oxidation-Reduction , Palmitoyl-CoA Hydrolase/chemistry , Palmitoyl-CoA Hydrolase/metabolism
6.
Biochem Biophys Res Commun ; 576: 15-21, 2021 10 22.
Article in English | MEDLINE | ID: mdl-34474245

ABSTRACT

Thyroid hormones (THs) play a critical role in the metabolic phenotype of the heart; and most of the effects involve transcriptional regulation via thyroid hormone receptors (TRs). TRs ability to form combinatorial complexes with an array of partners accounts for TRs physiological flexibility in modulating gene expression. To identify proteins that associate with TRß1 in the heart we performed a pull-down assay on cardiac tissue using GST-TRß1 as bait and identified the bound proteins by LC MS/MS. ACAA2, a mitochondrial thiolase enzyme, was identified as a novel interacting protein. We confirmed ACAA2 localized to the nucleus and using a luciferase reporter assay showed ACAA2 acted as a TH-dependent coactivator for TRß1. ACAA2 showed an ability to bind to TR recognition sequences but did not alter TRß1 DNA binding ability. Thus, ACAA2 as a novel TRß1 associating protein opens a new paradigm to understanding how TH/TRs may be manipulated by energetic pathway molecules.


Subject(s)
Acetyl-CoA C-Acyltransferase/metabolism , Myocardium/metabolism , Thyroid Hormone Receptors beta/metabolism , Thyroid Hormones/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Ligands , Male , Mice , Mice, Inbred C57BL , Models, Animal , Protein Interaction Domains and Motifs , Tandem Mass Spectrometry/methods , Transcription, Genetic
7.
PLoS Comput Biol ; 17(8): e1009259, 2021 08.
Article in English | MEDLINE | ID: mdl-34383741

ABSTRACT

In this study we demonstrated through analytic considerations and numerical studies that the mitochondrial fatty-acid ß-oxidation can exhibit bistable-hysteresis behavior. In an experimentally validated computational model we identified a specific region in the parameter space in which two distinct stable and one unstable steady state could be attained with different fluxes. The two stable states were referred to as low-flux (disease) and high-flux (healthy) state. By a modular kinetic approach we traced the origin and causes of the bistability back to the distributive kinetics and the conservation of CoA, in particular in the last rounds of the ß-oxidation. We then extended the model to investigate various interventions that may confer health benefits by activating the pathway, including (i) activation of the last enzyme MCKAT via its endogenous regulator p46-SHC protein, (ii) addition of a thioesterase (an acyl-CoA hydrolysing enzyme) as a safety valve, and (iii) concomitant activation of a number of upstream and downstream enzymes by short-chain fatty-acids (SCFA), metabolites that are produced from nutritional fibers in the gut. A high concentration of SCFAs, thioesterase activity, and inhibition of the p46Shc protein led to a disappearance of the bistability, leaving only the high-flux state. A better understanding of the switch behavior of the mitochondrial fatty-acid oxidation process between a low- and a high-flux state may lead to dietary and pharmacological intervention in the treatment or prevention of obesity and or non-alcoholic fatty-liver disease.


Subject(s)
Fatty Acids/metabolism , Models, Biological , Acetyl-CoA C-Acyltransferase/antagonists & inhibitors , Acetyl-CoA C-Acyltransferase/metabolism , Animals , Computational Biology , Computer Simulation , Enzyme Stability , Fatty Acids/chemistry , Humans , Kinetics , Metabolic Networks and Pathways , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/etiology , Obesity/metabolism
8.
Nat Commun ; 11(1): 6266, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33293512

ABSTRACT

Grain weight (GW) is one of the component traits of wheat yield. Existing reports have shown that multiple phytohormones are involved in the regulation of GW in different crops. However, the potential role of jasmonic acid (JA) remains unclear. Here, we report that triticale grain weight 1 (tgw1) mutant, with marked reductions in both GW and JA content, is caused by a premature stop mutation in keto-acyl thiolase 2B (KAT-2B) involved in ß-oxidation during JA synthesis. KAT-2B overexpression increases GW in wild type and boosts yield. Additionally, KAT-2B compliments the grain defect in tgw1 and rescues the lethal phenotype of the Arabidopsis kat2 mutant in a sucrose-free medium. Despite the suppression of JA synthesis in tgw1 mutant, ABA synthesis is upregulated, which is accompanied by enhanced expression of SAG3 and reduction of chlorophyll content in leaves. Together, these results demonstrate a role of the JA synthetic gene KAT-2B in controlling GW and its potential application value for wheat improvement.


Subject(s)
Acetyl-CoA C-Acyltransferase/metabolism , Cyclopentanes/metabolism , Edible Grain/physiology , Oxylipins/metabolism , Plant Proteins/metabolism , Triticum/physiology , Abscisic Acid/metabolism , Acetyl-CoA C-Acyltransferase/genetics , Acetyl-CoA C-Acyltransferase/isolation & purification , Chlorophyll/metabolism , Cloning, Molecular , Codon, Nonsense , Plant Leaves/metabolism , Plant Proteins/genetics , Plant Proteins/isolation & purification , Plants, Genetically Modified , Quantitative Trait Loci , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
9.
Cell Rep ; 33(8): 108421, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33238129

ABSTRACT

Emerging evidence indicates that non-mutational drug tolerance mechanisms underlie the survival of residual cancer "persister" cells. Here, we find that BRAF(V600E) mutant melanoma persister cells tolerant to BRAF/MEK inhibitors switch their metabolism from glycolysis to oxidative respiration supported by peroxisomal fatty acid ß-oxidation (FAO) that is transcriptionally regulated by peroxisome proliferator-activated receptor alpha (PPARα). Knockdown of the key peroxisomal FAO enzyme, acyl-CoA oxidase 1 (ACOX1), as well as treatment with the peroxisomal FAO inhibitor thioridazine, specifically suppresses the oxidative respiration of persister cells and significantly decreases their emergence. Consistently, a combination treatment of BRAF/MEK inhibitors with thioridazine in human-melanoma-bearing mice results in a durable anti-tumor response. In BRAF(V600E) melanoma samples from patients treated with BRAF/MEK inhibitors, higher baseline expression of FAO-related genes and PPARα correlates with patients' outcomes. These results pave the way for a metabolic strategy to overcome drug resistance.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/metabolism , Acetyl-CoA C-Acyltransferase/metabolism , Acyl-CoA Oxidase/metabolism , Carbon-Carbon Double Bond Isomerases/metabolism , Enoyl-CoA Hydratase/metabolism , Melanoma/genetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Racemases and Epimerases/metabolism , Animals , Humans , Melanoma/pathology , Mice , Protein Kinase Inhibitors/pharmacology
10.
Mol Genet Metab ; 131(1-2): 90-97, 2020.
Article in English | MEDLINE | ID: mdl-32928639

ABSTRACT

BACKGROUND: The plasma acylcarnitine profile is frequently used as a biochemical assessment for follow-up in diagnosed patients with fatty acid oxidation disorders (FAODs). Disease specific acylcarnitine species are elevated during metabolic decompensation but there is clinical and biochemical heterogeneity among patients and limited data on the utility of an acylcarnitine profile for routine clinical monitoring. METHODS: We evaluated plasma acylcarnitine profiles from 30 diagnosed patients with long-chain FAODs (carnitine palmitoyltransferase-2 (CPT2), very long-chain acyl-CoA dehydrogenase (VLCAD), and long-chain 3-hydroxy acyl-CoA dehydrogenase or mitochondrial trifunctional protein (LCHAD/TFP) deficiencies) collected after an overnight fast, after feeding a controlled low-fat diet, and before and after moderate exercise. Our purpose was to describe the variability in this biomarker and how various physiologic states effect the acylcarnitine concentrations in circulation. RESULTS: Disease specific acylcarnitine species were higher after an overnight fast and decreased by approximately 60% two hours after a controlled breakfast meal. Moderate-intensity exercise increased the acylcarnitine species but it varied by diagnosis. When analyzed for a genotype/phenotype correlation, the presence of the common LCHADD mutation (c.1528G > C) was associated with higher levels of 3-hydroxyacylcarnitines than in patients with other mutations. CONCLUSIONS: We found that feeding consistently suppressed and that moderate intensity exercise increased disease specific acylcarnitine species, but the response to exercise was highly variable across subjects and diagnoses. The clinical utility of routine plasma acylcarnitine analysis for outpatient treatment monitoring remains questionable; however, if acylcarnitine profiles are measured in the clinical setting, standardized procedures are required for sample collection to be of value.


Subject(s)
Cardiomyopathies/blood , Carnitine O-Palmitoyltransferase/deficiency , Carnitine/analogs & derivatives , Congenital Bone Marrow Failure Syndromes/blood , Lipid Metabolism, Inborn Errors/blood , Metabolism, Inborn Errors/blood , Mitochondrial Diseases/blood , Mitochondrial Myopathies/blood , Mitochondrial Trifunctional Protein/deficiency , Muscular Diseases/blood , Nervous System Diseases/blood , Rhabdomyolysis/blood , 3-Hydroxyacyl CoA Dehydrogenases/genetics , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Acetyl-CoA C-Acyltransferase/genetics , Acetyl-CoA C-Acyltransferase/metabolism , Acyl-CoA Dehydrogenase, Long-Chain/blood , Carbon-Carbon Double Bond Isomerases/genetics , Carbon-Carbon Double Bond Isomerases/metabolism , Cardiomyopathies/diet therapy , Cardiomyopathies/pathology , Cardiomyopathies/therapy , Carnitine/blood , Carnitine/genetics , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/blood , Congenital Bone Marrow Failure Syndromes/diet therapy , Congenital Bone Marrow Failure Syndromes/pathology , Congenital Bone Marrow Failure Syndromes/therapy , Enoyl-CoA Hydratase/genetics , Enoyl-CoA Hydratase/metabolism , Exercise Therapy , Fasting , Female , Humans , Lipid Metabolism, Inborn Errors/diet therapy , Lipid Metabolism, Inborn Errors/pathology , Lipid Metabolism, Inborn Errors/therapy , Long-Chain-3-Hydroxyacyl-CoA Dehydrogenase/blood , Male , Metabolism, Inborn Errors/diet therapy , Metabolism, Inborn Errors/pathology , Metabolism, Inborn Errors/therapy , Mitochondrial Diseases/diet therapy , Mitochondrial Diseases/pathology , Mitochondrial Diseases/therapy , Mitochondrial Myopathies/diet therapy , Mitochondrial Myopathies/pathology , Mitochondrial Myopathies/therapy , Mitochondrial Trifunctional Protein/blood , Muscular Diseases/diet therapy , Muscular Diseases/pathology , Muscular Diseases/therapy , Nervous System Diseases/diet therapy , Nervous System Diseases/pathology , Nervous System Diseases/therapy , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Rhabdomyolysis/diet therapy , Rhabdomyolysis/pathology , Rhabdomyolysis/therapy
SELECTION OF CITATIONS
SEARCH DETAIL