Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 477
1.
J Biol Chem ; 300(3): 105728, 2024 Mar.
Article En | MEDLINE | ID: mdl-38325740

Serine palmitoyltransferase (SPT) catalyzes the pyridoxal-5'-phosphate (PLP)-dependent decarboxylative condensation of l-serine and palmitoyl-CoA to form 3-ketodihydrosphingosine (KDS). Although SPT was shown to synthesize corresponding products from amino acids other than l-serine, it is still arguable whether SPT catalyzes the reaction with d-serine, which is a question of biological importance. Using high substrate and enzyme concentrations, KDS was detected after the incubation of SPT from Sphingobacterium multivorum with d-serine and palmitoyl-CoA. Furthermore, the KDS comprised equal amounts of 2S and 2R isomers. 1H-NMR study showed a slow hydrogen-deuterium exchange at Cα of serine mediated by SPT. We further confirmed that SPT catalyzed the racemization of serine. The rate of the KDS formation from d-serine was comparable to those for the α-hydrogen exchange and the racemization reaction. The structure of the d-serine-soaked crystal (1.65 Å resolution) showed a distinct electron density of the PLP-l-serine aldimine, interpreted as the racemized product trapped in the active site. The structure of the α-methyl-d-serine-soaked crystal (1.70 Å resolution) showed the PLP-α-methyl-d-serine aldimine, mimicking the d-serine-SPT complex prior to racemization. Based on these enzymological and structural analyses, the synthesis of KDS from d-serine was explained as the result of the slow racemization to l-serine, followed by the reaction with palmitoyl-CoA, and SPT would not catalyze the direct condensation between d-serine and palmitoyl-CoA. It was also shown that the S. multivorum SPT catalyzed the racemization of the product KDS, which would explain the presence of (2R)-KDS in the reaction products.


Serine C-Palmitoyltransferase , Serine , Sphingobacterium , Catalytic Domain , Crystallization , Deuterium Exchange Measurement , Electrons , Hydrogen/metabolism , Palmitoyl Coenzyme A/metabolism , Serine/analogs & derivatives , Serine/metabolism , Serine C-Palmitoyltransferase/chemistry , Serine C-Palmitoyltransferase/metabolism , Sphingobacterium/enzymology , Sphingobacterium/metabolism , Sphingosine/analogs & derivatives , Sphingosine/biosynthesis , Sphingosine/metabolism , Stereoisomerism , Substrate Specificity
2.
J Biol Chem ; 299(5): 104684, 2023 05.
Article En | MEDLINE | ID: mdl-37030501

Serine palmitoyltransferase (SPT) is a key enzyme of sphingolipid biosynthesis, which catalyzes the pyridoxal-5'-phosphate-dependent decarboxylative condensation reaction of l-serine (l-Ser) and palmitoyl-CoA (PalCoA) to form 3-ketodihydrosphingosine called long chain base (LCB). SPT is also able to metabolize l-alanine (l-Ala) and glycine (Gly), albeit with much lower efficiency. Human SPT is a membrane-bound large protein complex containing SPTLC1/SPTLC2 heterodimer as the core subunits, and it is known that mutations of the SPTLC1/SPTLC2 genes increase the formation of deoxy-type of LCBs derived from l-Ala and Gly to cause some neurodegenerative diseases. In order to study the substrate recognition of SPT, we examined the reactivity of Sphingobacterium multivorum SPT on various amino acids in the presence of PalCoA. The S. multivorum SPT could convert not only l-Ala and Gly but also l-homoserine, in addition to l-Ser, into the corresponding LCBs. Furthermore, we obtained high-quality crystals of the ligand-free form and the binary complexes with a series of amino acids, including a nonproductive amino acid, l-threonine, and determined the structures at 1.40 to 1.55 Å resolutions. The S. multivorum SPT accommodated various amino acid substrates through subtle rearrangements of the active-site amino acid residues and water molecules. It was also suggested that non-active-site residues mutated in the human SPT genes might indirectly influence the substrate specificity by affecting the hydrogen-bonding networks involving the bound substrate, water molecules, and amino acid residues in the active site of this enzyme. Collectively, our results highlight SPT structural features affecting substrate specificity for this stage of sphingolipid biosynthesis.


Serine C-Palmitoyltransferase , Sphingobacterium , Humans , Palmitoyl Coenzyme A/chemistry , Palmitoyl Coenzyme A/metabolism , Serine/chemistry , Serine C-Palmitoyltransferase/genetics , Serine C-Palmitoyltransferase/metabolism , Sphingobacterium/enzymology , Sphingolipids/metabolism , Substrate Specificity
3.
Mol Cell ; 81(24): 5025-5038.e10, 2021 12 16.
Article En | MEDLINE | ID: mdl-34890564

The Sonic Hedgehog (SHH) morphogen pathway is fundamental for embryonic development and stem cell maintenance and is implicated in various cancers. A key step in signaling is transfer of a palmitate group to the SHH N terminus, catalyzed by the multi-pass transmembrane enzyme Hedgehog acyltransferase (HHAT). We present the high-resolution cryo-EM structure of HHAT bound to substrate analog palmityl-coenzyme A and a SHH-mimetic megabody, revealing a heme group bound to HHAT that is essential for HHAT function. A structure of HHAT bound to potent small-molecule inhibitor IMP-1575 revealed conformational changes in the active site that occlude substrate binding. Our multidisciplinary analysis provides a detailed view of the mechanism by which HHAT adapts the membrane environment to transfer an acyl chain across the endoplasmic reticulum membrane. This structure of a membrane-bound O-acyltransferase (MBOAT) superfamily member provides a blueprint for other protein-substrate MBOATs and a template for future drug discovery.


Acyltransferases/antagonists & inhibitors , Acyltransferases/metabolism , Enzyme Inhibitors/pharmacology , Hedgehog Proteins/metabolism , Membrane Proteins/metabolism , Acylation , Acyltransferases/genetics , Acyltransferases/ultrastructure , Allosteric Regulation , Animals , COS Cells , Catalytic Domain , Chlorocebus aethiops , Cryoelectron Microscopy , HEK293 Cells , Heme/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/ultrastructure , Molecular Dynamics Simulation , Palmitoyl Coenzyme A/metabolism , Protein Conformation , Signal Transduction , Structure-Activity Relationship
4.
Science ; 372(6547): 1215-1219, 2021 06 11.
Article En | MEDLINE | ID: mdl-34112694

Hedgehog proteins govern crucial developmental steps in animals and drive certain human cancers. Before they can function as signaling molecules, Hedgehog precursor proteins must undergo amino-terminal palmitoylation by Hedgehog acyltransferase (HHAT). We present cryo-electron microscopy structures of human HHAT in complex with its palmitoyl-coenzyme A substrate and of a product complex with a palmitoylated Hedgehog peptide at resolutions of 2.7 and 3.2 angstroms, respectively. The structures reveal how HHAT overcomes the challenges of bringing together substrates that have different physiochemical properties from opposite sides of the endoplasmic reticulum membrane within a membrane-embedded active site for catalysis. These principles are relevant to related enzymes that catalyze the acylation of Wnt and of the appetite-stimulating hormone ghrelin. The structural and mechanistic insights may advance the development of inhibitors for cancer.


Acyltransferases/chemistry , Acyltransferases/metabolism , Endoplasmic Reticulum/enzymology , Hedgehog Proteins/chemistry , Palmitoyl Coenzyme A/chemistry , Acylation , Biocatalysis , Catalytic Domain , Cryoelectron Microscopy , Hedgehog Proteins/metabolism , Humans , Intracellular Membranes/enzymology , Lipoylation , Models, Molecular , Molecular Dynamics Simulation , Palmitoyl Coenzyme A/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , Protein Structure, Secondary
5.
Biochem J ; 478(13): 2539-2553, 2021 07 16.
Article En | MEDLINE | ID: mdl-34129667

Reductions in mitochondrial function have been proposed to cause insulin resistance, however the possibility that impairments in insulin signaling negatively affects mitochondrial bioenergetics has received little attention. Therefore, we tested the hypothesis that insulin could rapidly improve mitochondrial ADP sensitivity, a key process linked to oxidative phosphorylation and redox balance, and if this phenomenon would be lost following high-fat diet (HFD)-induced insulin resistance. Insulin acutely (60 min post I.P.) increased submaximal (100-1000 µM ADP) mitochondrial respiration ∼2-fold without altering maximal (>1000 µM ADP) respiration, suggesting insulin rapidly improves mitochondrial bioenergetics. The consumption of HFD impaired submaximal ADP-supported respiration ∼50%, however, despite the induction of insulin resistance, the ability of acute insulin to stimulate ADP sensitivity and increase submaximal respiration persisted. While these data suggest that insulin mitigates HFD-induced impairments in mitochondrial bioenergetics, the presence of a high intracellular lipid environment reflective of an HFD (i.e. presence of palmitoyl-CoA) completely prevented the beneficial effects of insulin. Altogether, these data show that while insulin rapidly stimulates mitochondrial bioenergetics through an improvement in ADP sensitivity, this phenomenon is possibly lost following HFD due to the presence of intracellular lipids.


Adenosine Diphosphate/pharmacology , Energy Metabolism/drug effects , Insulin/pharmacology , Mitochondria, Muscle/drug effects , Muscle, Skeletal/drug effects , Adenosine Diphosphate/metabolism , Animals , Body Weight/drug effects , Diet, High-Fat , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Injections, Intraperitoneal , Insulin/administration & dosage , Insulin/metabolism , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Palmitoyl Coenzyme A/metabolism , Palmitoyl Coenzyme A/pharmacology
6.
Angew Chem Int Ed Engl ; 60(24): 13542-13547, 2021 06 07.
Article En | MEDLINE | ID: mdl-33768725

The mammalian membrane-bound O-acyltransferase (MBOAT) superfamily is involved in biological processes including growth, development and appetite sensing. MBOATs are attractive drug targets in cancer and obesity; however, information on the binding site and molecular mechanisms underlying small-molecule inhibition is elusive. This study reports rational development of a photochemical probe to interrogate a novel small-molecule inhibitor binding site in the human MBOAT Hedgehog acyltransferase (HHAT). Structure-activity relationship investigation identified single enantiomer IMP-1575, the most potent HHAT inhibitor reported to-date, and guided design of photocrosslinking probes that maintained HHAT-inhibitory potency. Photocrosslinking and proteomic sequencing of HHAT delivered identification of the first small-molecule binding site in a mammalian MBOAT. Topology and homology data suggested a potential mechanism for HHAT inhibition which was confirmed by kinetic analysis. Our results provide an optimal HHAT tool inhibitor IMP-1575 (Ki =38 nM) and a strategy for mapping small molecule interaction sites in MBOATs.


Acetyltransferases/antagonists & inhibitors , Affinity Labels/chemistry , Small Molecule Libraries/chemistry , Acetyltransferases/metabolism , Binding Sites , Humans , Kinetics , Light , Palmitoyl Coenzyme A/antagonists & inhibitors , Palmitoyl Coenzyme A/metabolism , Pyridines/chemistry , Pyridines/metabolism , Small Molecule Libraries/metabolism , Structure-Activity Relationship
7.
Nat Metab ; 2(9): 873-881, 2020 09.
Article En | MEDLINE | ID: mdl-32719536

Long-chain fatty acids (LCFAs) play important roles in cellular energy metabolism, acting as both an important energy source and signalling molecules1. LCFA-CoA esters promote their own oxidation by acting as allosteric inhibitors of acetyl-CoA carboxylase, which reduces the production of malonyl-CoA and relieves inhibition of carnitine palmitoyl-transferase 1, thereby promoting LCFA-CoA transport into the mitochondria for ß-oxidation2-6. Here we report a new level of regulation wherein LCFA-CoA esters per se allosterically activate AMP-activated protein kinase (AMPK) ß1-containing isoforms to increase fatty acid oxidation through phosphorylation of acetyl-CoA carboxylase. Activation of AMPK by LCFA-CoA esters requires the allosteric drug and metabolite site formed between the α-subunit kinase domain and the ß-subunit. ß1 subunit mutations that inhibit AMPK activation by the small-molecule activator A769662, which binds to the allosteric drug and metabolite site, also inhibit activation by LCFA-CoAs. Thus, LCFA-CoA metabolites act as direct endogenous AMPK ß1-selective activators and promote LCFA oxidation.


AMP-Activated Protein Kinases/metabolism , Acyl Coenzyme A/physiology , Allosteric Regulation/physiology , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/genetics , Animals , Biphenyl Compounds , Catalytic Domain , Esters , Isoenzymes/chemistry , Isoenzymes/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Mutation/genetics , Oxidation-Reduction , Palmitoyl Coenzyme A/metabolism , Phosphorylation , Pyrones/pharmacology , Thiophenes/pharmacology
8.
Nucleic Acids Res ; 48(11): 5967-5985, 2020 06 19.
Article En | MEDLINE | ID: mdl-32406921

During infection of a host, Pseudomonas aeruginosa orchestrates global gene expression to adapt to the host environment and counter the immune attacks. P. aeruginosa harbours hundreds of regulatory genes that play essential roles in controlling gene expression. However, their contributions to the bacterial pathogenesis remain largely unknown. In this study, we analysed the transcriptomic profile of P. aeruginosa cells isolated from lungs of infected mice and examined the roles of upregulated regulatory genes in bacterial virulence. Mutation of a novel regulatory gene pvrA (PA2957) attenuated the bacterial virulence in an acute pneumonia model. Chromatin immunoprecipitation (ChIP)-Seq and genetic analyses revealed that PvrA directly regulates genes involved in phosphatidylcholine utilization and fatty acid catabolism. Mutation of the pvrA resulted in defective bacterial growth when phosphatidylcholine or palmitic acid was used as the sole carbon source. We further demonstrated that palmitoyl coenzyme A is a ligand for the PvrA, enhancing the binding affinity of PvrA to its target promoters. An arginine residue at position 136 was found to be essential for PvrA to bind palmitoyl coenzyme A. Overall, our results revealed a novel regulatory pathway that controls genes involved in phosphatidylcholine and fatty acid utilization and contributes to the bacterial virulence.


Bacterial Proteins/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Genes, Bacterial/genetics , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/pathogenicity , Animals , Arginine/metabolism , Base Sequence , Chromatin Immunoprecipitation , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Ligands , Mice , Models, Molecular , Mutation , Palmitic Acid/metabolism , Palmitoyl Coenzyme A/metabolism , Phosphatidylcholines/metabolism , Pneumonia, Bacterial/microbiology , Promoter Regions, Genetic , Pseudomonas aeruginosa/genetics , Transcriptome , Virulence/genetics
9.
Cell Rep ; 29(13): 4608-4619.e4, 2019 12 24.
Article En | MEDLINE | ID: mdl-31875564

Attachment of palmitate to the N terminus of Sonic hedgehog (Shh) is essential for Shh signaling. Shh palmitoylation is catalyzed on the luminal side of the endoplasmic reticulum (ER) by Hedgehog acyltransferase (Hhat), an ER-resident enzyme. Palmitoyl-coenzyme A (CoA), the palmitate donor, is produced in the cytosol and is not permeable across membrane bilayers. It is not known how palmitoyl-CoA crosses the ER membrane to access the active site of Hhat. Here, we use fluorescent and radiolabeled palmitoyl-CoA probes to demonstrate that Hhat promotes the uptake of palmitoyl-CoA across the ER membrane in microsomes and semi-intact cells. Reconstitution of purified Hhat into liposomes provided further evidence that palmitoyl-CoA uptake activity is an intrinsic property of Hhat. Palmitoyl-CoA uptake was regulated by and could be uncoupled from Hhat enzymatic activity, implying that Hhat serves a dual function as a palmitoyl acyltransferase and a conduit to supply palmitoyl-CoA to the luminal side of the ER.


Acyltransferases/metabolism , Endoplasmic Reticulum/metabolism , Hedgehog Proteins/metabolism , Microsomes/metabolism , Palmitoyl Coenzyme A/metabolism , Protein Processing, Post-Translational , Acyltransferases/genetics , Animals , Biological Transport , COS Cells , Cell Line , Chlorocebus aethiops , Endoplasmic Reticulum/ultrastructure , Fibroblasts/metabolism , Fibroblasts/ultrastructure , HEK293 Cells , Hedgehog Proteins/genetics , Humans , Liposomes/metabolism , Liposomes/ultrastructure , Lipoylation , Mice , Microsomes/ultrastructure , Signal Transduction , Staining and Labeling/methods
10.
PLoS One ; 14(9): e0222558, 2019.
Article En | MEDLINE | ID: mdl-31550253

A fatty acid analogue, 2-(tridec-12-yn-1-ylthio)acetic acid (1-triple TTA), was previously shown to have hypolipidemic effects in rats by targeting mitochondrial activity predominantly in liver. This study aimed to determine if 1-triple TTA could influence carbohydrate metabolism. Male Wistar rats were treated for three weeks with oral supplementation of 100 mg/kg body weight 1-triple TTA. Blood glucose and insulin levels, and liver carbohydrate metabolism gene expression and enzyme activities were determined. In addition, human myotubes and Huh7 liver cells were treated with 1-triple TTA, and glucose and fatty acid oxidation were determined. The level of plasma insulin was significantly reduced in 1-triple TTA-treated rats, resulting in a 32% reduction in the insulin/glucose ratio. The hepatic glucose and glycogen levels were lowered by 22% and 49%, respectively, compared to control. This was accompanied by lower hepatic gene expression of phosphenolpyruvate carboxykinase, the rate-limiting enzyme in gluconeogenesis, and Hnf4A, a regulator of gluconeogenesis. Gene expression of pyruvate kinase, catalysing the final step of glycolysis, was also reduced by 1-triple TTA. In addition, pyruvate dehydrogenase activity was reduced, accompanied by 10-15-fold increased gene expression of its regulator pyruvate dehydrogenase kinase 4 compared to control, suggesting reduced entry of pyruvate into the TCA cycle. Indeed, the NADPH-generating enzyme malic enzyme 1 (ME1) catalysing production of pyruvate from malate, was increased 13-fold at the gene expression level. Despite the decreased glycogen level, genes involved in glycogen synthesis were not affected in livers of 1-triple TTA treated rats. In contrast, the pentose phosphate pathway seemed to be increased as the hepatic gene expression of glucose-6-phosphate dehydrogenase (G6PD) was higher in 1-triple TTA treated rats compared to controls. In human Huh7 liver cells, but not in myotubes, 1-triple-TTA reduced glucose oxidation and induced fatty acid oxidation, in line with previous observations of increased hepatic mitochondrial palmitoyl-CoA oxidation in rats. Importantly, this work recognizes the liver as an important organ in glucose homeostasis. The mitochondrially targeted fatty acid analogue 1-triple TTA seemed to lower hepatic glucose and glycogen levels by inhibition of gluconeogenesis. This was also linked to a reduction in glucose oxidation accompanied by reduced PHD activity and stimulation of ME1 and G6PD, favouring a shift from glucose- to fatty acid oxidation. The reduced plasma insulin/glucose ratio indicate that 1-triple TTA may improve glucose tolerance in rats.


Acetates/pharmacology , Blood Glucose/analysis , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Insulin/blood , Liver/drug effects , Mitochondria, Liver/drug effects , Animals , Cell Line , Fructosephosphates/metabolism , Humans , Liver/metabolism , Liver Glycogen/metabolism , Male , Metabolic Networks and Pathways/drug effects , Muscle Fibers, Skeletal/metabolism , NADP/metabolism , Palmitoyl Coenzyme A/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Rats , Rats, Wistar
11.
J Appl Physiol (1985) ; 127(2): 312-319, 2019 08 01.
Article En | MEDLINE | ID: mdl-31161881

We investigated the effect of temperature increase on mitochondrial fatty acid (FA) and carbohydrate oxidation in the slow-oxidative skeletal muscles (soleus) of rats. We measured mitochondrial respiration at 35°C and 40°C with the physiological substrates pyruvate + 4 mM malate (Pyr) and palmitoyl-CoA (PCoA) + 0.5 mM malate + 2 mM carnitine in permeabilized myofibers under nonphosphorylating (V˙0) or phosphorylating (V˙max) conditions. Mitochondrial efficiency was calculated by the respiratory control ratio (RCR = V˙max/V˙0). We used guanosine triphosphate (GTP), an inhibitor of uncoupling protein (UCP), to study the mechanisms responsible for alterations of mitochondrial efficiency. We measured hydrogen peroxide (H2O2) production under nonphosphorylating and phosphorylating conditions at both temperatures and substrates. We studied citrate synthase (CS) and 3-hydroxyl acyl coenzyme A dehydrogenase (3-HAD) activities at both temperatures. Elevating the temperature from 35°C to 40°C increased PCoA-V˙0 and decreased PCoA-RCR, corresponding to the uncoupling of oxidative phosphorylation (OXPHOS). GTP blocked the heat-induced increase of PCoA-V˙0. Rising temperature moved toward a Pyr-V˙0 increase, without significance. Heat did not alter H2O2 production, resulting from either PCoA or Pyr oxidation. Heat induced an increase in 3-HAD but not in CS activities. In conclusion, heat induced OXPHOS uncoupling for PCoA oxidation, which was at least partially mediated by UCP and independent of oxidative stress. The classically described heat-induced glucose shift may actually be mostly due to a less efficient FA oxidation. These findings raise questions concerning the consequences of heat-induced alterations in mitochondrial efficiency of FA metabolism on thermoregulation.NEW & NOTEWORTHY Ex vivo exposure of skeletal myofibers to heat uncouples substrate oxidation from ADP phosphorylation, decreasing the efficiency of mitochondria to produce ATP. This heat effect alters fatty acids (FAs) more than carbohydrate oxidation. Alteration of FA oxidation involves uncoupling proteins without inducing oxidative stress. This alteration in lipid metabolism may underlie the preferential use of carbohydrates in the heat and could decrease aerobic endurance.


Fatty Acids/metabolism , Mitochondria, Muscle/metabolism , Myofibrils/metabolism , Animals , Carnitine/metabolism , Cell Respiration/physiology , Citrate (si)-Synthase/metabolism , Glucose/metabolism , Hydrogen Peroxide/metabolism , Lipid Metabolism/physiology , Malates/metabolism , Male , Mitochondrial Proteins/metabolism , Muscle, Skeletal/metabolism , Oxidation-Reduction , Oxidative Phosphorylation , Oxidative Stress/physiology , Oxygen Consumption/physiology , Palmitoyl Coenzyme A/metabolism , Pyruvic Acid/metabolism , Rats , Rats, Wistar , Temperature
12.
Methods Mol Biol ; 2009: 243-255, 2019.
Article En | MEDLINE | ID: mdl-31152409

Hedgehog and Wnt proteins are modified by covalent attachment of the fatty acids palmitate and palmitoleate, respectively. These lipid modifications are essential for Hedgehog and Wnt protein signaling activities and are catalyzed by related, but distinct fatty acyltransferases: Hedgehog acyltransferase (Hedgehog) and Porcupine (Wnt). In this chapter, we provide detailed methods to directly monitor Hedgehog and Wnt protein fatty acylation in vitro. Palmitoylation of Sonic hedgehog (Shh), a representative Hedgehog family member, is assayed using purified Hedgehog acyltransferase (Hhat) or Hhat-enriched membranes, a recombinant 19 kDa Shh protein or C-terminally biotinylated Shh 10-mer peptide, and 125I-iodopalmitoyl CoA as the donor fatty acyl CoA substrate. The radiolabeled reaction products are quantified by SDS-PAGE and phosphorimaging or by γ-counting. To assay Wnt acylation, the reaction consists of a biotinylated, double disulfide-bonded Wnt peptide containing the sequence surrounding the Wnt3a acylation site, [125I] iodo-cis-9-pentadecenoyl CoA, and Porcupine-enriched membranes. Radiolabeled, biotinylated Wnt3a peptide is captured on streptavidin coated beads and the reaction product is quantified by γ-counting.


Acyltransferases/chemistry , Hedgehog Proteins/chemistry , Membrane Proteins/chemistry , Palmitoyl Coenzyme A/chemistry , Protein Processing, Post-Translational , Wnt Proteins/chemistry , Acylation , Acyltransferases/metabolism , Hedgehog Proteins/metabolism , Humans , Iodine Radioisotopes/chemistry , Membrane Proteins/metabolism , Membranes, Artificial , Palmitoyl Coenzyme A/metabolism , Wnt Proteins/metabolism
13.
Biochem J ; 475(18): 2997-3008, 2018 09 28.
Article En | MEDLINE | ID: mdl-30111574

The mechanisms regulating oxidative phosphorylation during exercise remain poorly defined; however, key mitochondrial proteins, including carnitine palmitoyltransferase-I (CPT-I) and adenine nucleotide translocase, have redox-sensitive sites. Interestingly, muscle contraction has recently been shown to increase mitochondrial membrane potential and reactive oxygen species (ROS) production; therefore, we aimed to determine if mitochondrial-derived ROS influences bioenergetic responses to exercise. Specifically, we examined the influence of acute exercise on mitochondrial bioenergetics in WT (wild type) and transgenic mice (MCAT, mitochondrial-targeted catalase transgenic) possessing attenuated mitochondrial ROS. We found that ablating mitochondrial ROS did not alter palmitoyl-CoA (P-CoA) respiratory kinetics or influence the exercise-mediated reductions in malonyl CoA sensitivity, suggesting that mitochondrial ROS does not regulate CPT-I. In contrast, while mitochondrial protein content, maximal coupled respiration, and ADP (adenosine diphosphate) sensitivity in resting muscle were unchanged in the absence of mitochondrial ROS, exercise increased the apparent ADP Km (decreased ADP sensitivity) ∼30% only in WT mice. Moreover, while the presence of P-CoA decreased ADP sensitivity, it did not influence the basic response to exercise, as the apparent ADP Km was increased only in the presence of mitochondrial ROS. This basic pattern was also mirrored in the ability of ADP to suppress mitochondrial H2O2 emission rates, as exercise decreased the suppression of H2O2 only in WT mice. Altogether, these data demonstrate that while exercise-induced mitochondrial-derived ROS does not influence CPT-I substrate sensitivity, it inhibits ADP sensitivity independent of P-CoA. These data implicate mitochondrial redox signaling as a regulator of oxidative phosphorylation.


Adenosine Diphosphate/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Hydrogen Peroxide/metabolism , Mitochondria, Muscle/metabolism , Physical Conditioning, Animal , Adenosine Diphosphate/genetics , Animals , Carnitine O-Palmitoyltransferase/genetics , Mice , Mice, Transgenic , Mitochondria, Muscle/genetics , Palmitoyl Coenzyme A/genetics , Palmitoyl Coenzyme A/metabolism , Substrate Specificity
14.
Exp Physiol ; 103(9): 1206-1212, 2018 09.
Article En | MEDLINE | ID: mdl-30088302

NEW FINDINGS: What is the central question of this study? Do peripheral sensory neurons metabolize fat-based fuel sources, and does a ketogenic diet modify these processes? What is the main finding and its importance We show that peripheral axons from mice fed a ketogenic diet respond to fat-based fuel sources with reduced respiration and H2 O2 emission compared with mice fed a control diet. These results add to our understanding of the responses of sensory neurons to neuropathy associated with poor diet, obesity and metabolic syndrome. These findings should be incorporated into current ideas of axonal protection and might identify how dietary interventions may change mitochondrial function in settings of sensory dysfunction. ABSTRACT: Metabolic syndrome and obesity are increasing epidemics that significantly impact the peripheral nervous system and lead to negative changes in sensation and peripheral nerve function. Research to understand the consequences of diet, obesity and fuel usage in sensory neurons has commonly focused on glucose metabolism. Here, we tested whether mouse sensory neurons and nerves have the capacity to metabolize fat-based fuels (palmitoyl-CoA) and whether these effects are altered by feeding of a ketogenic (90% kcal fat) diet compared with a control diet (14% kcal fat). Male C57Bl/6 mice were placed on the diets for 10 weeks, and after the mice were killed, the dorsal root ganglion (DRG) and sciatic nerve (SN) were placed in an Oroboros oxygraph-2K to examine diet-induced alterations in metabolism (respiration) of palmitoyl-CoA and H2 O2 emission (fluorescence). In addition, RNAseq was performed on the DRG of mice fed a control or a ketogenic diet for 12 weeks, and genes associated with mitochondrial respiratory function were analysed. Our results suggest that the sciatic nerves from mice fed a ketogenic diet display reduced O2 respiration and H2 O2 emission when metabolizing palmitoyl-CoA compared with mice fed a control diet. Assessments of changes in mRNA gene expression reveal alterations in genes encoding the NADH dehydrogenase complex and complex IV, which could alter production of reactive oxygen species. These new findings highlight the ability of sensory neurons and axons to oxidize fat-based fuel sources and show that these mechanisms are adaptable to dietary changes.


Diet, Ketogenic , Mitochondria/metabolism , Peripheral Nerves/metabolism , Reactive Oxygen Species/metabolism , Animals , Blood Glucose/metabolism , Ganglia, Spinal/metabolism , Gene Expression/genetics , Hydrogen Peroxide/metabolism , Male , Mice , Mice, Inbred C57BL , Palmitoyl Coenzyme A/metabolism , Phosphorylation , Sciatic Nerve/metabolism , Sensory Receptor Cells/metabolism
15.
Mol Cell Endocrinol ; 472: 40-49, 2018 09 05.
Article En | MEDLINE | ID: mdl-29180108

Dietary fats can modulate brain function. How free fatty acids (FFAs) alter hypothalamic pro-opiomelanocortin (POMC) neurons remain undefined. The saturated FFA, palmitate, increased neuroinflammatory and ER stress markers, as well as Pomc mRNA levels, but did not affect insulin signaling, in mHypoA-POMC/GFP-2 neurons. This effect was mediated through the MAP kinases JNK and ERK. Further, the increase in Pomc was dependent on palmitoyl-coA synthesis, but not de novo ceramide synthesis, as inhibition of SPT enhanced palmitate-induced Pomc expression, while methylpalmitate had no effect. While palmitate concomitantly induces neuroinflammation and ER stress, these effects were independent of changes in Pomc expression. Palmitate thus has direct acute effects on Pomc, which appears to be important for negative feedback, but not directly related to neuroinflammation. The monounsaturated FFA oleate completely blocked the palmitate-mediated increase in neuroinflammation, ER stress, and Pomc mRNAs. This study provides insight into the complex central metabolic regulation by FFAs.


Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation/drug effects , Hypothalamus/metabolism , Neurons/pathology , Oleic Acid/pharmacology , Palmitates/toxicity , Pro-Opiomelanocortin/metabolism , Animals , Biomarkers/metabolism , Ceramides/biosynthesis , Green Fluorescent Proteins/metabolism , Hypothalamus/drug effects , I-kappa B Kinase/metabolism , Inflammation/pathology , Insulin/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice, Transgenic , Models, Biological , Neurons/drug effects , Neurons/metabolism , Palmitoyl Coenzyme A/metabolism , Pro-Opiomelanocortin/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Toll-Like Receptor 4/metabolism
16.
PLoS Comput Biol ; 13(6): e1005588, 2017 Jun.
Article En | MEDLINE | ID: mdl-28598967

Lipids are main fuels for cellular energy and mitochondria their major oxidation site. Yet unknown is to what extent the fuel role of lipids is influenced by their uncoupling effects, and how this affects mitochondrial energetics, redox balance and the emission of reactive oxygen species (ROS). Employing a combined experimental-computational approach, we comparatively analyze ß-oxidation of palmitoyl CoA (PCoA) in isolated heart mitochondria from Sham and streptozotocin (STZ)-induced type 1 diabetic (T1DM) guinea pigs (GPs). Parallel high throughput measurements of the rates of oxygen consumption (VO2) and hydrogen peroxide (H2O2) emission as a function of PCoA concentration, in the presence of L-carnitine and malate, were performed. We found that PCoA concentration < 200 nmol/mg mito protein resulted in low H2O2 emission flux, increasing thereafter in Sham and T1DM GPs under both states 4 and 3 respiration with diabetic mitochondria releasing higher amounts of ROS. Respiratory uncoupling and ROS excess occurred at PCoA > 600 nmol/mg mito prot, in both control and diabetic animals. Also, for the first time, we show that an integrated two compartment mitochondrial model of ß-oxidation of long-chain fatty acids and main energy-redox processes is able to simulate the relationship between VO2 and H2O2 emission as a function of lipid concentration. Model and experimental results indicate that PCoA oxidation and its concentration-dependent uncoupling effect, together with a partial lipid-dependent decrease in the rate of superoxide generation, modulate H2O2 emission as a function of VO2. Results indicate that keeping low levels of intracellular lipid is crucial for mitochondria and cells to maintain ROS within physiological levels compatible with signaling and reliable energy supply.


Diabetes Mellitus/metabolism , Lipid Metabolism , Mitochondria, Heart/metabolism , Models, Cardiovascular , Palmitoyl Coenzyme A/metabolism , Reactive Oxygen Species/metabolism , Animals , Cell Respiration , Cells, Cultured , Computer Simulation , Electron Transport , Guinea Pigs , Hydrogen Peroxide/metabolism , Male , Metabolism , Oxidation-Reduction , Oxygen/metabolism
17.
Biochem Pharmacol ; 138: 185-192, 2017 08 15.
Article En | MEDLINE | ID: mdl-28450226

Human carbonyl reductase 1 (CBR1), a member of the short-chain dehydrogenase/reductase (SDR) superfamily, reduces a variety of carbonyl compounds including endogenous isatin, prostaglandin E2 and 4-oxo-2-nonenal. It is also a major non-cytochrome P450 enzyme in the phase I metabolism of carbonyl-containing drugs, and is highly expressed in the intestine. In this study, we found that long-chain fatty acids and their CoA ester derivatives inhibit CBR1. Among saturated fatty acids, myristic, palmitic and stearic acids were inhibitory, and stearic acid was the most potent (IC50 9µM). Unsaturated fatty acids (oleic, elaidic, γ-linolenic and docosahexaenoic acids) and acyl-CoAs (palmitoyl-, stearoyl- and oleoyl-CoAs) were more potent inhibitors (IC50 1.0-2.5µM), and showed high inhibitory selectivity to CBR1 over its isozyme CBR3 and other SDR superfamily enzymes (DCXR and DHRS4) with CBR activity. The inhibition by these fatty acids and acyl-CoAs was competitive with respect to the substrate, showing the Ki values of 0.49-1.2µM. Site-directed mutagenesis of the substrate-binding residues of CBR1 suggested that the interactions between the fatty acyl chain and the enzyme's Met141 and Trp229 are important for the inhibitory selectivity. We also examined CBR1 inhibition by oleic acid in cellular levels: The fatty acid effectively inhibited CBR1-mediated 4-oxo-2-nonenal metabolism in colon cancer DLD1 cells and increased sensitivity to doxorubicin in the drug-resistant gastric cancer MKN45 cells that highly express CBR1. The results suggest a possible new food-drug interaction through inhibition of CBR1-mediated intestinal first-pass drug metabolism by dietary fatty acids.


Acyl Coenzyme A/metabolism , Alcohol Oxidoreductases/antagonists & inhibitors , Fatty Acids, Nonesterified/metabolism , Intestinal Mucosa/enzymology , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Binding Sites , Binding, Competitive , Cell Line, Tumor , Drug Resistance, Neoplasm , Food-Drug Interactions , Humans , Mutation , Myristic Acid/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/genetics , Oxidoreductases/metabolism , Palmitic Acid/metabolism , Palmitoyl Coenzyme A/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Stearic Acids/metabolism , Sugar Alcohol Dehydrogenases/antagonists & inhibitors , Sugar Alcohol Dehydrogenases/genetics , Sugar Alcohol Dehydrogenases/metabolism
18.
Biochem J ; 474(4): 557-569, 2017 02 15.
Article En | MEDLINE | ID: mdl-27941154

The obligatory role of carnitine palmitoyltransferase-I (CPT-I) in mediating mitochondrial lipid transport is well established, a process attenuated by malonyl-CoA (M-CoA). However, the necessity of reducing M-CoA concentrations to promote lipid oxidation has recently been challenged, suggesting external regulation on CPT-I. Since previous work in hepatocytes suggests the involvement of the intermediate filament fraction of the cytoskeleton in regulating CPT-I, we investigated in skeletal muscle if CPT-I sensitivity for M-CoA inhibition could be regulated by the intermediate filaments, and whether AMP-activated protein kinase (AMPK) could be involved in this process. Chemical disruption (3,3'-iminodipropionitrile, IDPN) of the intermediate filaments did not alter mitochondrial respiration or sensitivity for numerous substrates (palmitoyl-CoA, ADP, palmitoyl carnitine and pyruvate). In contrast, IDPN reduced CPT-I sensitivity for M-CoA inhibition in permeabilized muscle fibers, identifying M-CoA kinetics as a specific target for intermediate filament regulation. Importantly, exercise mimicked the effect of IDPN on M-CoA sensitivity, suggesting that intermediate filament disruption in vivo is physiologically important for CPT-I regulation. To ascertain a potential mechanism, since AMPK is activated during exercise, AMPK ß1ß2-KO mice were utilized in an attempt to ablate the observed exercise response. Unexpectedly, these mice displayed drastic attenuation in resting M-CoA sensitivity, such that exercise and IDPN could not further alter M-CoA sensitivity. These data suggest that AMPK is not required for the regulation of the intermediate filament interaction with CPT-I. Altogether, these data highlight that M-CoA sensitivity is important for regulating mitochondrial lipid transport. Moreover, M-CoA sensitivity appears to be regulated by intermediate filament interaction with CPT-I, a process that is important when metabolic homeostasis is challenged.


AMP-Activated Protein Kinases/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Intermediate Filaments/metabolism , Malonyl Coenzyme A/metabolism , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , AMP-Activated Protein Kinases/genetics , Adenosine Diphosphate/metabolism , Animals , Carnitine O-Palmitoyltransferase/genetics , Gene Expression Regulation , Intermediate Filaments/drug effects , Male , Mice , Mice, Knockout , Mitochondria, Muscle/genetics , Muscle, Skeletal/drug effects , Nitriles/pharmacology , Oxidation-Reduction , Oxidative Phosphorylation , Palmitoyl Coenzyme A/metabolism , Palmitoylcarnitine/metabolism , Physical Conditioning, Animal , Pyruvic Acid/metabolism , Signal Transduction , Substrate Specificity
19.
Article En | MEDLINE | ID: mdl-29312893

The structure of Vibrio cholerae FadR (VcFadR) complexed with the ligand oleoyl-CoA suggests an additional ligand-binding site. However, the fatty acid metabolism and its regulation is poorly addressed in Vibrio alginolyticus, a species closely-related to V. cholerae. Here, we show crystal structures of V. alginolyticus FadR (ValFadR) alone and its complex with the palmitoyl-CoA, a long-chain fatty acyl ligand different from the oleoyl-CoA occupied by VcFadR. Structural comparison indicates that both VcFadR and ValFadR consistently have an additional ligand-binding site (called site 2), which leads to more dramatic conformational-change of DNA-binding domain than that of the E. coli FadR (EcFadR). Isothermal titration calorimetry (ITC) analyses defines that the ligand-binding pattern of ValFadR (2:1) is distinct from that of EcFadR (1:1). Together with surface plasmon resonance (SPR), electrophoresis mobility shift assay (EMSA) demonstrates that ValFadR binds fabA, an important gene of unsaturated fatty acid (UFA) synthesis. The removal of fadR from V. cholerae attenuates fabA transcription and results in the unbalance of UFA/SFA incorporated into membrane phospholipids. Genetic complementation of the mutant version of fadR (Δ42, 136-177) lacking site 2 cannot restore the defective phenotypes of ΔfadR while the wild-type fadR gene and addition of exogenous oleate can restore them. Mice experiments reveals that VcFadR and its site 2 have roles in bacterial colonizing. Together, the results might represent an additional example that illustrates the Vibrio FadR-mediated lipid regulation and its role in pathogenesis.


Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Palmitoyl Coenzyme A/chemistry , Palmitoyl Coenzyme A/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Vibrio alginolyticus/enzymology , Animals , Binding Sites , Cholera/microbiology , Cholera/pathology , Crystallography, X-Ray , DNA, Bacterial/metabolism , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Mice , Models, Molecular , Protein Binding , Protein Conformation , Surface Plasmon Resonance , Vibrio alginolyticus/metabolism , Vibrio cholerae/enzymology , Vibrio cholerae/pathogenicity , Virulence
20.
Nat Commun ; 7: 10906, 2016 Mar 11.
Article En | MEDLINE | ID: mdl-26965057

The biosynthesis of phospholipids and glycolipids are critical pathways for virtually all cell membranes. PatA is an essential membrane associated acyltransferase involved in the biosynthesis of mycobacterial phosphatidyl-myo-inositol mannosides (PIMs). The enzyme transfers a palmitoyl moiety from palmitoyl-CoA to the 6-position of the mannose ring linked to 2-position of inositol in PIM1/PIM2. We report here the crystal structures of PatA from Mycobacterium smegmatis in the presence of its naturally occurring acyl donor palmitate and a nonhydrolyzable palmitoyl-CoA analog. The structures reveal an α/ß architecture, with the acyl chain deeply buried into a hydrophobic pocket that runs perpendicular to a long groove where the active site is located. Enzyme catalysis is mediated by an unprecedented charge relay system, which markedly diverges from the canonical HX4D motif. Our studies establish the mechanistic basis of substrate/membrane recognition and catalysis for an important family of acyltransferases, providing exciting possibilities for inhibitor design.


Acyltransferases/metabolism , Mycobacterium smegmatis/metabolism , Acyltransferases/chemistry , Catalysis , Catalytic Domain , Cell Membrane/metabolism , Crystallography, X-Ray , Mannosides/biosynthesis , Mycobacterium smegmatis/chemistry , Palmitates/metabolism , Palmitoyl Coenzyme A/metabolism , Phosphatidylinositols/biosynthesis , Protein Structure, Secondary , Protein Structure, Tertiary
...