Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 161(5): 1152-1163, 2015 May 21.
Article in English | MEDLINE | ID: mdl-25981666

ABSTRACT

Cells adapt to temperature shifts by adjusting levels of lipid desaturation and membrane fluidity. This fundamental process occurs in nearly all forms of life, but its mechanism in eukaryotes is unknown. We discovered that the evolutionarily conserved Caenorhabditis elegans gene acdh-11 (acyl-CoA dehydrogenase [ACDH]) facilitates heat adaptation by regulating the lipid desaturase FAT-7. Human ACDH deficiency causes the most common inherited disorders of fatty acid oxidation, with syndromes that are exacerbated by hyperthermia. Heat upregulates acdh-11 expression to decrease fat-7 expression. We solved the high-resolution crystal structure of ACDH-11 and established the molecular basis of its selective and high-affinity binding to C11/C12-chain fatty acids. ACDH-11 sequesters C11/C12-chain fatty acids and prevents these fatty acids from activating nuclear hormone receptors and driving fat-7 expression. Thus, the ACDH-11 pathway drives heat adaptation by linking temperature shifts to regulation of lipid desaturase levels and membrane fluidity via an unprecedented mode of fatty acid signaling.


Subject(s)
Acyl-CoA Dehydrogenase/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , Fatty Acids/metabolism , Acyl-CoA Dehydrogenase/chemistry , Adaptation, Physiological , Amino Acid Sequence , Animals , Caenorhabditis elegans Proteins/chemistry , Hot Temperature , Models, Molecular , Molecular Sequence Data , Sequence Alignment
2.
J Biol Phys ; 50(1): 89-118, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38103157

ABSTRACT

Acyl-CoA dehydrogenase deficiency (ACAD) is an inherited and potentially fatal disorder with variable clinical symptoms. The relationship between pathogenicity and deleterious point mutations is investigated here in ACAD structures of short (SCAD) and medium-chain (MCAD) types. Structures and dynamic features of native and mutant forms of enzymes models were compared. A total of 2.88 µs molecular dynamics simulations were performed at four different temperatures. Total energy, RMSD, protein ligand interactions and affinity, RMSF measures, secondary structure changes, and important interactions were studied. Mutations in the three main domains of ACADs are pathogenic, while those located at linker turns are not. Mutations affect mostly tetramer formations, secondary structures, and many contacts and interactions. In R206H (MCAD mutant) which is experimentally known to cause a huge turnover decrease, the lack of a single H-bond between substrate and FAD was observed. Secondary structures showed temperature-dependent changes, and SCAD activity was found to be highly correlated to the enzyme helix 3-10 content. Finally, RMSF patterns pointed to one important loop that maintains the substrate close to the active site and is a cause of substrate wobbling upon mutation. Despite similar structure, function, and cellular location, SCAD and MCAD may have different optimum temperatures that are related to the structure taken at that specific temperature. In conclusion, new insight has been provided on the effect of various SCAD and MCAD pathogenic mutations on the structure and dynamical features of the enzymes.


Subject(s)
Lipid Metabolism, Inborn Errors , Point Mutation , Humans , Virulence , Acyl-CoA Dehydrogenase/chemistry , Acyl-CoA Dehydrogenase/genetics , Lipid Metabolism, Inborn Errors/genetics , Protein Structure, Secondary
3.
Clin Genet ; 103(6): 644-654, 2023 06.
Article in English | MEDLINE | ID: mdl-36840705

ABSTRACT

Biallelic variants in the ACADM gene cause medium-chain acyl-CoA dehydrogenase deficiency (MCADD). This study reports on differences in the occurrence of secondary free carnitine (C0) deficiency and different biochemical phenotypes related to genotype and age in 109 MCADD patients followed-up at a single tertiary care center during 22 years. C0 deficiency occurred earlier and more frequently in c.985A>G homozygotes (genotype A) compared to c.985A>G compound heterozygotes (genotype B) and individuals carrying variants other than c.985A>G and c.199C>T (genotype D) (median age 4.2 vs. 6.6 years; p < 0.001). No patient carrying c.199C>T (genotype C) developed C0 deficiency. A daily dosage of 20-40 mg/kg carnitine was sufficient to maintain normal C0 concentrations. Compared to genotype A as reference group, octanoylcarnitine (C8) was significantly lower in genotypes B and C, whereas C0 was significantly higher by 8.28 µmol/L in genotype C (p < 0.05). In conclusion, C0 deficiency is mainly found in patients with pathogenic genotypes associated with high concentrations of presumably toxic acylcarnitines, while individuals carrying the variant c.199C>T are spared and show consistently mild biochemical phenotypes into adulthood. Low-dose carnitine supplementation maintains normal C0 concentrations. However, future studies need to evaluate clinical benefits on acute and chronic manifestations of MCADD.


Subject(s)
Lipid Metabolism, Inborn Errors , Neonatal Screening , Humans , Infant, Newborn , Genotype , Lipid Metabolism, Inborn Errors/genetics , Carnitine , Amino Acids , Genetic Association Studies , Acyl-CoA Dehydrogenase/chemistry , Acyl-CoA Dehydrogenase/genetics
4.
Proc Natl Acad Sci U S A ; 117(28): 16324-16332, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32601219

ABSTRACT

FadE, an acyl-CoA dehydrogenase, introduces unsaturation to carbon chains in lipid metabolism pathways. Here, we report that FadE5 from Mycobacterium tuberculosis (MtbFadE5) and Mycobacterium smegmatis (MsFadE5) play roles in drug resistance and exhibit broad specificity for linear acyl-CoA substrates but have a preference for those with long carbon chains. Here, the structures of MsFadE5 and MtbFadE5, in the presence and absence of substrates, have been determined. These reveal the molecular basis for the broad substrate specificity of these enzymes. FadE5 interacts with the CoA region of the substrate through a large number of hydrogen bonds and an unusual π-π stacking interaction, allowing these enzymes to accept both short- and long-chain substrates. Residues in the substrate binding cavity reorient their side chains to accommodate substrates of various lengths. Longer carbon-chain substrates make more numerous hydrophobic interactions with the enzyme compared with the shorter-chain substrates, resulting in a preference for this type of substrate.


Subject(s)
Acyl-CoA Dehydrogenase/chemistry , Acyl-CoA Dehydrogenase/metabolism , Mycobacterium/enzymology , Acyl Coenzyme A/metabolism , Acyl-CoA Dehydrogenase/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Catalytic Domain , Drug Resistance, Bacterial/genetics , Fatty Acids/chemistry , Fatty Acids/metabolism , Models, Molecular , Mutation , Mycobacterium/drug effects , Mycobacterium/genetics , Protein Conformation , Structure-Activity Relationship , Substrate Specificity
5.
Int J Mol Sci ; 22(18)2021 Sep 13.
Article in English | MEDLINE | ID: mdl-34576026

ABSTRACT

Quorum sensing (QS) is a microbial cell-cell communication mechanism and plays an important role in bacterial infections. QS-mediated bacterial infections can be blocked through quorum quenching (QQ), which hampers signal accumulation, recognition, and communication. The pathogenicity of numerous bacteria, including Xanthomonas campestris pv. campestris (Xcc), is regulated by diffusible signal factor (DSF), a well-known fatty acid signaling molecule of QS. Cupriavidus pinatubonensis HN-2 could substantially attenuate the infection of XCC through QQ by degrading DSF. The QQ mechanism in strain HN-2, on the other hand, is yet to be known. To understand the molecular mechanism of QQ in strain HN-2, we used whole-genome sequencing and comparative genomics studies. We discovered that the fadT gene encodes acyl-CoA dehydrogenase as a novel QQ enzyme. The results of site-directed mutagenesis demonstrated the requirement of fadT gene for DSF degradation in strain HN-2. Purified FadT exhibited high enzymatic activity and outstanding stability over a broad pH and temperature range with maximal activity at pH 7.0 and 35 °C. No cofactors were required for FadT enzyme activity. The enzyme showed a strong ability to degrade DSF. Furthermore, the expression of fadT in Xcc results in a significant reduction in the pathogenicity in host plants, such as Chinese cabbage, radish, and pakchoi. Taken together, our results identified a novel DSF-degrading enzyme, FadT, in C. pinatubonensis HN-2, which suggests its potential use in the biological control of DSF-mediated pathogens.


Subject(s)
Acyl-CoA Dehydrogenase/genetics , Bacterial Infections/genetics , Fatty Acids/genetics , Plant Diseases/genetics , Xanthomonas campestris/genetics , Acyl-CoA Dehydrogenase/chemistry , Acyl-CoA Dehydrogenase/isolation & purification , Bacterial Infections/microbiology , Brassica/growth & development , Brassica/microbiology , Cell Communication/genetics , Fatty Acids/metabolism , Gene Expression Regulation, Enzymologic , Genome, Bacterial/genetics , Genomics , Mutagenesis, Site-Directed , Plant Diseases/microbiology , Quorum Sensing/genetics , Raphanus/genetics , Raphanus/microbiology , Signal Transduction/genetics , Virulence Factors/genetics , Whole Genome Sequencing , Xanthomonas campestris/enzymology
6.
Biochemistry ; 59(42): 4081-4092, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33040522

ABSTRACT

A wide variety of steroid metabolites synthesized by eukaryotes are all ultimately catabolized by bacteria; while generally saprophytic, pathogenic Mycobacteria have repurposed these pathways to utilize host intracellular cholesterol pools. Steroid degradation is complex, but a recurring theme is that cycles of ß-oxidation are used to iteratively remove acetyl- or propanoyl-CoA groups. These ß-oxidation cycles are initiated by the FAD-dependent oxidation of acyl groups, catalyzed by acyl-CoA dehydrogenases (ACADs). We show here that the tcur3481 and tcur3483 genes of Thermomonospora curvata encode subunits of a single ACAD that degrades steroid side chains with a preference for three-carbon over five-carbon substituents. The structure confirms that this enzyme is heterotetrameric, with active sites only in the Tcur3483 subunits. In comparison with the steroid ACAD FadE26-FadE27 from Mycobacterium tuberculosis, the active site is narrower and closed at the steroid-binding end, suggesting that Tcur3481-Tcur3483 is in a catalytically productive state, while FadE26-FadE27 is opened up to allow substrate entry. The flavin rings in Tcur3481-Tcur3483 sit in an unusual pocket created by Gly363, a residue conserved as Ala in steroid ACADs narrowly specific for five-carbon side chains, including FadE34. A Gly363Ala variant of Tcur3481-Tcur3483 prefers five-carbon side chains, while an inverse Ala691Gly FadE34 variant enables three-carbon side chain steroid oxidation. We determined the structure of the Tcur3483 Gly363Ala variant, showing that the flavin rings shift into the more conventional position. Modeling suggests that the shifted flavin position made possible by Gly363 is required to allow the bulky, inflexible three-carbon steroid to bind productively in the active site.


Subject(s)
Acyl-CoA Dehydrogenase/metabolism , Glycine/metabolism , Acyl-CoA Dehydrogenase/chemistry , Catalytic Domain , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/metabolism , Steroids/metabolism , Substrate Specificity
7.
J Am Chem Soc ; 142(2): 835-846, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31793780

ABSTRACT

Terminal alkenes are easily derivatized, making them desirable functional group targets for polyketide synthase (PKS) engineering. However, they are rarely encountered in natural PKS systems. One mechanism for terminal alkene formation in PKSs is through the activity of an acyl-CoA dehydrogenase (ACAD). Herein, we use biochemical and structural analysis to understand the mechanism of terminal alkene formation catalyzed by an γ,δ-ACAD from the biosynthesis of the polyketide natural product FK506, TcsD. While TcsD is homologous to canonical α,ß-ACADs, it acts regioselectively at the γ,δ-position and only on α,ß-unsaturated substrates. Furthermore, this regioselectivity is controlled by a combination of bulky residues in the active site and a lateral shift in the positioning of the FAD cofactor within the enzyme. Substrate modeling suggests that TcsD utilizes a novel set of hydrogen bond donors for substrate activation and positioning, preventing dehydrogenation at the α,ß position of substrates. From the structural and biochemical characterization of TcsD, key residues that contribute to regioselectivity and are unique to the protein family were determined and used to identify other putative γ,δ-ACADs that belong to diverse natural product biosynthetic gene clusters. These predictions are supported by the demonstration that a phylogenetically distant homologue of TcsD also regioselectively oxidizes α,ß-unsaturated substrates. This work exemplifies a powerful approach to understand unique enzymatic reactions and will facilitate future enzyme discovery, inform enzyme engineering, and aid natural product characterization efforts.


Subject(s)
Acyl-CoA Dehydrogenase/chemistry , Bacteria/enzymology , Protein Conformation
8.
Int J Mol Sci ; 21(15)2020 Jul 29.
Article in English | MEDLINE | ID: mdl-32751237

ABSTRACT

Several alkylating agents that either occur in the environment or are self-produced can cause DNA-damaging injuries in bacterial cells. Therefore, all microorganisms have developed repair systems that are able to counteract DNA alkylation damage. The adaptive response to alkylation stress in Escherichia coli consists of the Ada operon, which has been widely described; however, the homologous system in Mycobacterium tuberculosis (MTB) has been shown to have a different genetic organization but it is still largely unknown. In order to describe the defense system of MTB, we first investigated the proteins involved in the repair mechanism in the homologous non-pathogenic mycobacterium M. smegmatis. Ogt, Ada-AlkA and FadE8 proteins were recombinantly produced, purified and characterized. The biological role of Ogt was examined using proteomic experiments to identify its protein partners in vivo under stress conditions. Our results suggested the formation of a functional complex between Ogt and Ada-AlkA, which was confirmed both in silico by docking calculations and by gel filtration chromatography. We propose that this stable association allows the complex to fulfill the biological roles exerted by Ada in the homologous E. coli system. Finally, FadE8 was demonstrated to be structurally and functionally related to its E. coli homologous, AidB.


Subject(s)
Acyl-CoA Dehydrogenase/chemistry , Bacterial Proteins/chemistry , DNA Repair , DNA, Bacterial/genetics , Methyltransferases/chemistry , Mycobacterium smegmatis/genetics , Acyl-CoA Dehydrogenase/genetics , Acyl-CoA Dehydrogenase/metabolism , Alkylating Agents/pharmacology , Alkylation , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Chromosomes, Bacterial/chemistry , Cloning, Molecular , DNA Damage , DNA, Bacterial/chemistry , DNA, Bacterial/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Molecular Docking Simulation , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Proteomics/methods , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
9.
PLoS Comput Biol ; 14(11): e1006626, 2018 11.
Article in English | MEDLINE | ID: mdl-30496278

ABSTRACT

The conformational dynamics of proteins is rarely used in methodologies used to predict the impact of genetic mutations due to the paucity of three-dimensional protein structures as compared to the vast number of available sequences. Until now a three-dimensional (3D) structure has been required to predict the conformational dynamics of a protein. We introduce an approach that estimates the conformational dynamics of a protein, without relying on structural information. This de novo approach utilizes coevolving residues identified from a multiple sequence alignment (MSA) using Potts models. These coevolving residues are used as contacts in a Gaussian network model (GNM) to obtain protein dynamics. B-factors calculated using sequence-based GNM (Seq-GNM) are in agreement with crystallographic B-factors as well as theoretical B-factors from the original GNM that utilizes the 3D structure. Moreover, we demonstrate the ability of the calculated B-factors from the Seq-GNM approach to discriminate genomic variants according to their phenotypes for a wide range of proteins. These results suggest that protein dynamics can be approximated based on sequence information alone, making it possible to assess the phenotypes of nSNVs in cases where a 3D structure is unknown. We hope this work will promote the use of dynamics information in genetic disease prediction at scale by circumventing the need for 3D structures.


Subject(s)
Acyl-CoA Dehydrogenase/chemistry , Computational Biology/methods , Disease Susceptibility , Neurons/metabolism , Protein Isoforms , Proteins/chemistry , Animals , Computer Simulation , Cytochrome Reductases/chemistry , Genomics , Humans , Imaging, Three-Dimensional , Molecular Conformation , Muramidase/chemistry , Normal Distribution , Phenotype , Protein Conformation , ROC Curve , Rats
10.
Biochemistry ; 55(51): 7086-7098, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27976856

ABSTRACT

Medium-chain acyl-CoA dehydrogenase deficiency (MCADD) is the most common genetic disorder affecting the mitochondrial fatty acid ß-oxidation pathway. The mature and functional form of human MCAD (hMCAD) is a homotetramer assembled as a dimer of dimers (monomers A/B and C/D). Each monomer binds a FAD cofactor, necessary for the enzyme's activity. The most frequent mutation in MCADD results from the substitution of a lysine with a glutamate in position 304 of mature hMCAD (p.K329E in the precursor protein). Here, we combined in vitro and in silico approaches to assess the impact of the p.K329E mutation on the protein's structure and function. Our in silico results demonstrated for the first time that the p.K329E mutation, despite lying at the dimer-dimer interface and being deeply buried inside the tetrameric core, seems to affect the tetramer surface, especially the ß-domain that forms part of the catalytic pocket wall. Additionally, the molecular dynamics data indicate a stronger impact of the mutation on the protein's motions in dimer A/B, while dimer C/D remains similar to the wild type. For dimer A/B, severe disruptions in the architecture of the pockets and in the FAD and octanoyl-CoA binding affinities were also observed. The presence of unaffected pockets (C/D) in the in silico studies may explain the decreased enzymatic activity determined for the variant protein (46% residual activity). Moreover, the in silico structural changes observed for the p.K329E variant protein provide an explanation for the structural instability observed experimentally, namely, the disturbed oligomeric profile, thermal stability, and conformational flexibility, with respect to the wild-type.


Subject(s)
Acyl-CoA Dehydrogenase/genetics , Computer Simulation , Lipid Metabolism, Inborn Errors/genetics , Mutation, Missense , Acyl-CoA Dehydrogenase/chemistry , Acyl-CoA Dehydrogenase/deficiency , Biocatalysis , Enzyme Stability , Glutamic Acid/genetics , Humans , Kinetics , Lipid Metabolism, Inborn Errors/enzymology , Lysine/genetics , Models, Molecular , Motion , Principal Component Analysis , Protein Binding , Protein Domains , Protein Multimerization , Temperature
11.
J Biol Chem ; 290(16): 10486-94, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25737446

ABSTRACT

Very long acyl-CoA dehydrogenase (VLCAD) deficiency is a genetic pediatric disorder presenting with a spectrum of phenotypes that remains for the most part untreatable. Here, we present a novel strategy for the correction of VLCAD deficiency by increasing mutant VLCAD enzymatic activity. Treatment of VLCAD-deficient fibroblasts, which express distinct mutant VLCAD protein and exhibit deficient fatty acid ß-oxidation, with S-nitroso-N-acetylcysteine induced site-specific S-nitrosylation of VLCAD mutants at cysteine residue 237. Cysteine 237 S-nitrosylation was associated with an 8-17-fold increase in VLCAD-specific activity and concomitant correction of acylcarnitine profile and ß-oxidation capacity, two hallmarks of the disorder. Overall, this study provides biochemical evidence for a potential therapeutic modality to correct ß-oxidation deficiencies.


Subject(s)
Acetylcysteine/analogs & derivatives , Acyl-CoA Dehydrogenase/metabolism , Carnitine/analogs & derivatives , Fibroblasts/drug effects , Acetylcysteine/pharmacology , Acyl-CoA Dehydrogenase/chemistry , Acyl-CoA Dehydrogenase/genetics , Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Amino Acid Sequence , Carnitine/metabolism , Congenital Bone Marrow Failure Syndromes , Cysteine/metabolism , Dose-Response Relationship, Drug , Fatty Acids/metabolism , Fibroblasts/enzymology , Fibroblasts/pathology , Genetic Therapy/methods , Humans , Kinetics , Lipid Metabolism, Inborn Errors/drug therapy , Lipid Metabolism, Inborn Errors/enzymology , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Molecular Sequence Data , Muscular Diseases/drug therapy , Muscular Diseases/enzymology , Muscular Diseases/genetics , Muscular Diseases/pathology , Mutation , Oxidation-Reduction , Primary Cell Culture , Skin/drug effects , Skin/enzymology , Skin/pathology
12.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 6): 1360-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26057676

ABSTRACT

3-Sulfinopropionyl-coenzyme A (3SP-CoA) desulfinase (AcdDPN7; EC 3.13.1.4) was identified during investigation of the 3,3'-dithiodipropionic acid (DTDP) catabolic pathway in the betaproteobacterium Advenella mimigardefordensis strain DPN7(T). DTDP is an organic disulfide and a precursor for the synthesis of polythioesters (PTEs) in bacteria, and is of interest for biotechnological PTE production. AcdDPN7 catalyzes sulfur abstraction from 3SP-CoA, a key step during the catabolism of DTDP. Here, the crystal structures of apo AcdDPN7 at 1.89 Å resolution and of its complex with the CoA moiety from the substrate analogue succinyl-CoA at 2.30 Å resolution are presented. The apo structure shows that AcdDPN7 belongs to the acyl-CoA dehydrogenase superfamily fold and that it is a tetramer, with each subunit containing one flavin adenine dinucleotide (FAD) molecule. The enzyme does not show any dehydrogenase activity. Dehydrogenase activity would require a catalytic base (Glu or Asp residue) at either position 246 or position 366, where a glutamine and a glycine are instead found, respectively, in this desulfinase. The positioning of CoA in the crystal complex enabled the modelling of a substrate complex containing 3SP-CoA. This indicates that Arg84 is a key residue in the desulfination reaction. An Arg84Lys mutant showed a complete loss of enzymatic activity, suggesting that the guanidinium group of the arginine is essential for desulfination. AcdDPN7 is the first desulfinase with an acyl-CoA dehydrogenase fold to be reported, which underlines the versatility of this enzyme scaffold.


Subject(s)
Acyl-CoA Dehydrogenase/chemistry , Alcaligenaceae/enzymology , Coenzyme A/chemistry , Enzymes/chemistry , Propionates/chemistry , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Folding
13.
Microbiology (Reading) ; 160(Pt 8): 1760-1771, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24794972

ABSTRACT

Diverse and elaborate pathways for nutrient utilization, as well as mechanisms to combat unfavourable nutrient conditions make Pseudomonas putida KT2440 a versatile micro-organism able to occupy a range of ecological niches. The fatty acid degradation pathway of P. putida is complex and correlated with biopolymer medium chain length polyhydroxyalkanoate (mcl-PHA) biosynthesis. Little is known about the second step of fatty acid degradation (ß-oxidation) in this strain. In silico analysis of its genome sequence revealed 21 putative acyl-CoA dehydrogenases (ACADs), four of which were functionally characterized through mutagenesis studies. Four mutants with insertionally inactivated ACADs (PP_1893, PP_2039, PP_2048 and PP_2437) grew and accumulated mcl-PHA on a range of fatty acids as the sole source of carbon and energy. Their ability to grow and accumulate biopolymer was differentially negatively affected on various fatty acids, in comparison to the wild-type strain. Inactive PP_2437 exhibited a pattern of reduced growth and PHA accumulation when fatty acids with lengths of 10 to 14 carbon chains were used as substrates. Recombinant expression and biochemical characterization of the purified protein allowed functional annotation in P. putida KT2440 as an ACAD showing clear preference for dodecanoyl-CoA ester as a substrate and optimum activity at 30 °C and pH 6.5-7.


Subject(s)
Acyl-CoA Dehydrogenase/chemistry , Acyl-CoA Dehydrogenase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Pseudomonas putida/enzymology , Acyl-CoA Dehydrogenase/genetics , Amino Acid Sequence , Bacterial Proteins/genetics , Molecular Sequence Data , Pseudomonas putida/chemistry , Pseudomonas putida/genetics , Pseudomonas putida/metabolism , Sequence Alignment , Substrate Specificity
14.
Mol Genet Metab ; 112(1): 30-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24685553

ABSTRACT

Short-chain acyl-CoA dehydrogenase (SCAD) deficiency is an autosomal recessive inborn error of metabolism that leads to the impaired mitochondrial fatty acid ß-oxidation of short chain fatty acids. It is heterogeneous in clinical presentation including asymptomatic in most patients identified by newborn screening. Multiple mutations have been identified in patients; however, neither clear genotype-phenotype relationships nor a good correlation between genotype and current biochemical markers for diagnosis has been identified. The definition and pathophysiology of this deficiency remain unclear. To better understand this disorder at a global level, quantitative alterations in the mitochondrial proteome in SCAD deficient mice were examined using a combined proteomics approach: two-dimensional gel difference electrophoresis (2DIGE) followed by protein identification with MALDI-TOF/TOF and iTRAQ labeling followed by nano-LC/MALDI-TOF/TOF. We found broad mitochondrial dysfunction in SCAD deficiency. Changes in the levels of multiple energy metabolism related proteins were identified indicating that a more complex mechanism for development of symptoms may exist. Affected pathways converge on disorders with neurologic symptoms, suggesting that even asymptomatic individuals with SCAD deficiency may be at risk to develop more severe disease. Our results also identified a pattern associated with hepatotoxicity implicated in mitochondrial dysfunction, fatty acid metabolism, decrease of depolarization of mitochondria and mitochondrial membranes, and swelling of mitochondria, demonstrating that SCAD deficiency relates more directly to mitochondrial dysfunction and alteration of fatty acid metabolism. We propose several candidate molecules that may serve as markers for recognition of clinical risk associated with this disorder.


Subject(s)
Acyl-CoA Dehydrogenase/deficiency , Liver/chemistry , Mitochondria/metabolism , Mitochondrial Proteins/analysis , Proteome/analysis , Acyl-CoA Dehydrogenase/chemistry , Animals , Biomarkers/metabolism , Energy Metabolism , Fatty Acids/metabolism , Gene Expression Regulation , Lipid Metabolism, Inborn Errors/pathology , Lipid Metabolism, Inborn Errors/physiopathology , Liver/physiopathology , Mice , Mice, Inbred BALB C , Mitochondria/genetics , Oxidation-Reduction
15.
J Bacteriol ; 195(7): 1538-51, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23354747

ABSTRACT

3-Sulfinopropionyl coenzyme A (3SP-CoA) desulfinase (AcdDPN7) is a new desulfinase that catalyzes the sulfur abstraction from 3SP-CoA in the betaproteobacterium Advenella mimigardefordensis strain DPN7(T). During investigation of a Tn5::mob-induced mutant defective in growth on 3,3'-dithiodipropionate (DTDP) and also 3-sulfinopropionate (3SP), the transposon insertion was mapped to an open reading frame with the highest homology to an acyl-CoA dehydrogenase (Acd) from Burkholderia phenoliruptrix strain BR3459a (83% identical and 91% similar amino acids). An A. mimigardefordensis Δacd mutant was generated and verified the observed phenotype of the Tn5::mob-induced mutant. For enzymatic studies, AcdDPN7 was heterologously expressed in Escherichia coli BL21(DE3)/pLysS by using pET23a::acdDPN7. The purified protein is yellow and contains a noncovalently bound flavin adenine dinucleotide (FAD) cofactor, as verified by high-performance liquid chromatography-electrospray ionization mass spectrometry (HPLC-ESI-MS) analyses. Size-exclusion chromatography revealed a native molecular mass of about 173 kDa, indicating a homotetrameric structure (theoretically 179 kDa), which is in accordance with other members of the acyl-CoA dehydrogenase superfamily. In vitro assays unequivocally demonstrated that the purified enzyme converted 3SP-CoA into propionyl-CoA and sulfite (SO3(2-)). Kinetic studies of AcdDPN7 revealed a Vmax of 4.19 µmol min(-1) mg(-1), an apparent Km of 0.013 mM, and a kcat/Km of 240.8 s(-1) mM(-1) for 3SP-CoA. However, AcdDPN7 is unable to perform a dehydrogenation, which is the usual reaction catalyzed by members of the acyl-CoA dehydrogenase superfamily. Comparison to other known desulfinases showed a comparably high catalytic efficiency of AcdDPN7 and indicated a novel reaction mechanism. Hence, AcdDPN7 encodes a new desulfinase based on an acyl-CoA dehydrogenase (EC 1.3.8.x) scaffold. Concomitantly, we identified the gene product that is responsible for the final desulfination step during catabolism of 3,3'-dithiodipropionate (DTDP), a sulfur-containing precursor substrate for biosynthesis of polythioesters.


Subject(s)
Acyl-CoA Dehydrogenase/metabolism , Alcaligenaceae/enzymology , Alcaligenaceae/metabolism , Propionates/metabolism , Acyl-CoA Dehydrogenase/chemistry , Acyl-CoA Dehydrogenase/genetics , Burkholderia/genetics , Cloning, Molecular , Coenzymes/metabolism , DNA Transposable Elements , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Escherichia coli/genetics , Flavin-Adenine Dinucleotide/metabolism , Gene Expression , Gene Knockout Techniques , Kinetics , Molecular Sequence Data , Molecular Weight , Mutagenesis, Insertional , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Analysis, DNA , Sequence Homology, Amino Acid
16.
Biochim Biophys Acta ; 1812(12): 1658-63, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21968293

ABSTRACT

Protein misfolding is a hallmark of a number of metabolic diseases, in which fatty acid oxidation defects are included. The latter result from genetic deficiencies in transport proteins and enzymes of the mitochondrial ß-oxidation, and milder disease conditions frequently result from conformational destabilization and decreased enzymatic function of the affected proteins. Small molecules which have the ability to raise the functional levels of the affected protein above a certain disease threshold are thus valuable tools for effective drug design. In this work we have investigated the effect of mitochondrial cofactors and metabolites as potential stabilizers in two ß-oxidation acyl-CoA dehydrogenases: short chain acyl-CoA dehydrogenase and the medium chain acyl-CoA dehydrogenase as well as glutaryl-CoA dehydrogenase, which is involved in lysine and tryptophan metabolism. We found that near physiological concentrations (low micromolar) of FAD resulted in a spectacular enhancement of the thermal stabilities of these enzymes and prevented enzymatic activity loss during a 1h incubation at 40°C. A clear effect of the respective substrate, which was additive to that of the FAD effect, was also observed for short- and medium-chain acyl-CoA dehydrogenase but not for glutaryl-CoA dehydrogenase. In conclusion, riboflavin may be beneficial during feverish crises in patients with short- and medium-chain acyl-CoA dehydrogenase as well as in glutaryl-CoA dehydrogenase deficiencies, and treatment with substrate analogs to butyryl- and octanoyl-CoAs could theoretically enhance enzyme activity for some enzyme proteins with inherited folding difficulties.


Subject(s)
Acyl-CoA Dehydrogenase/chemistry , Butyryl-CoA Dehydrogenase/chemistry , Coenzymes/chemistry , Glutaryl-CoA Dehydrogenase/chemistry , Mitochondrial Proteins/chemistry , Acyl Coenzyme A/chemistry , Calorimetry, Differential Scanning , Catalytic Domain , Enzyme Assays , Enzyme Stability , Flavin-Adenine Dinucleotide/chemistry , Humans , Protein Binding , Protein Unfolding , Riboflavin/chemistry , Transition Temperature
17.
Mol Genet Metab ; 107(4): 684-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23141465

ABSTRACT

Sodium phenylbutyrate is used for treating urea cycle disorders, providing an alternative for ammonia excretion. Following conversion to its CoA ester, phenylbutyryl-CoA is postulated to undergo one round of ß-oxidation to phenylacetyl-CoA, the active metabolite. Molecular modeling suggests that medium chain acyl-CoA dehydrogenase (MCAD; EC 1.3.99.3), a key enzyme in straight chain fatty acid ß-oxidation, could utilize phenylbutyryl-CoA as substrate. Moreover, phenylpropionyl-CoA has been shown to be a substrate for MCAD and its intermediates accumulate in patients with MCAD deficiency. We have examined the involvement of MCAD and other acyl-CoA dehydrogenases (ACADs) in the metabolism of phenylbutyryl-CoA. Anaerobic titration of purified recombinant human MCAD with phenylbutyryl-CoA caused changes in the MCAD spectrum that are similar to those induced by octanoyl-CoA, its bona fide substrate, and unique to the development of the charge transfer ternary complex. The calculated apparent dissociation constant (K(D app)) for these substrates was 2.16 µM and 0.12 µM, respectively. The MCAD reductive and oxidative half reactions were monitored using the electron transfer flavoprotein (ETF) fluorescence reduction assay. The catalytic efficiency and the K(m) for phenylbutyryl-CoA were 0.2 mM 34(-1)·sec(-1) and 5.3 µM compared to 4.0 mM(-1)·sec(-1) and 2.8 µM for octanoyl-CoA. Extracts of wild type and MCAD-deficient lymphoblast cells were tested for the ability to reduce ETF using phenylbutyryl-CoA as substrate. While ETF reduction activity was detected in extracts of wild type cells, it was undetectable in extracts of cells deficient in MCAD. The results are consistent with MCAD playing a key role in phenylbutyrate metabolism.


Subject(s)
Acyl-CoA Dehydrogenase/metabolism , Phenylbutyrates/metabolism , Acyl-CoA Dehydrogenase/chemistry , Catalytic Domain , Electron-Transferring Flavoproteins/metabolism , Humans , Kinetics , Metabolic Networks and Pathways , Molecular Docking Simulation , Oxidation-Reduction , Protein Conformation , Recombinant Proteins/metabolism , Substrate Specificity
18.
Hum Mol Genet ; 18(9): 1612-23, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19224950

ABSTRACT

Newborn screening (NBS) for medium-chain acyl-CoA dehydrogenase deficiency (MCADD) revealed a higher birth prevalence and genotypic variability than previously estimated, including numerous novel missense mutations in the ACADM gene. On average, these mutations are associated with milder biochemical phenotypes raising the question about their pathogenic relevance. In this study, we analyzed the impact of 10 ACADM mutations identified in NBS (A27V, Y42H, Y133H, R181C, R223G, D241G, K304E, R309K, I331T and R388S) on conformation, stability and enzyme kinetics of the corresponding proteins. Partial to total rescue of aggregation by co-overexpression of GroESL indicated protein misfolding. This was confirmed by accelerated thermal unfolding in all variants, as well as decreased proteolytic stability and accelerated thermal inactivation in most variants. Catalytic function varied from high residual activity to markedly decreased activity or substrate affinity. Mutations mapping to the beta-domain of the protein predisposed to severe destabilization. In silico structural analyses of the affected amino acid residues revealed involvement in functionally relevant networks. Taken together, our results substantiate the hypothesis of protein misfolding with loss-of-function being the common molecular basis in MCADD. Moreover, considerable structural alterations in all analyzed variants do not support the view that novel mutations found in NBS bear a lower risk of metabolic decompensation than that associated with mutations detected in clinically ascertained patients. Finally, the detailed insight into how ACADM missense mutations induce loss of MCAD function may provide guidance for risk assessment and counseling of patients, and in future may assist delineation of novel pharmacological strategies.


Subject(s)
Acyl-CoA Dehydrogenase/chemistry , Acyl-CoA Dehydrogenase/deficiency , Lipid Metabolism, Inborn Errors/enzymology , Neonatal Screening , Protein Folding , Acyl-CoA Dehydrogenase/genetics , Amino Acid Substitution , Enzyme Stability , Female , Humans , Infant, Newborn , Kinetics , Lipid Metabolism, Inborn Errors/genetics , Male , Molecular Conformation , Molecular Sequence Data , Mutation, Missense
19.
Mol Genet Metab ; 102(4): 418-29, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21237683

ABSTRACT

Long-chain fatty acids are an important source of energy in muscle and heart where the acyl-CoA dehydrogenases (ACADs) participate in consecutive cycles of ß-oxidation to generate acetyl-CoA and reducing equivalents for generating energy. However, the role of long-chain fatty acid oxidation in the brain and other tissues that do not rely on fat for energy is poorly understood. Here we characterize two new ACADs, ACAD10 and ACAD11, both with significant expression in human brain. ACAD11 utilizes substrates with primary carbon chain lengths between 20 and 26, with optimal activity towards C22CoA. The combination of ACAD11 with the newly characterized ACAD9 accommodates the full spectrum of long chain fatty acid substrates presented to mitochondrial ß-oxidation in human cerebellum. ACAD10 has significant activity towards the branched-chain substrates R and S, 2 methyl-C15-CoA and is highly expressed in fetal but not adult brain. This pattern of expression is similar to that of LCAD, another ACAD previously shown to be involved in long branched chain fatty acid metabolism. Interestingly, the ACADs in human cerebellum were found to have restricted cellular distribution. ACAD9 was most highly expressed in the granular layer, ACAD11 in the white matter, and MCAD in the molecular layer and axons of specific neurons. This compartmentalization of ACADs in the human central nerve system suggests that ß-oxidation in cerebellum participates in different functions other than generating energy, for example, the synthesis and/or degradation of unique cellular lipids and catabolism of aromatic amino acids, compounds that are vital to neuronal function.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Acyl-CoA Dehydrogenase/metabolism , Brain/metabolism , Acyl-CoA Dehydrogenase/chemistry , Acyl-CoA Dehydrogenase/genetics , Acyl-CoA Dehydrogenase, Long-Chain/chemistry , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Alternative Splicing , Amino Acid Sequence , Brain/embryology , Brain/growth & development , Cells, Cultured , Cerebellum/anatomy & histology , Cerebellum/cytology , Cerebellum/metabolism , Computer Simulation , Enzyme Assays , Gene Components , Humans , Isoenzymes/metabolism , Kidney/metabolism , Mitochondria/metabolism , Molecular Sequence Data , Muscle, Skeletal/metabolism , Organ Specificity , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Sequence Alignment , Substrate Specificity , Transcription, Genetic
20.
Biochemistry ; 49(17): 3517-24, 2010 May 04.
Article in English | MEDLINE | ID: mdl-20334431

ABSTRACT

D-kijanose is an unusual nitrosugar found attached to the antibiotic kijanimicin. Ten enzymes are required for its production in Actinomadura kijaniata, a soil-dwelling actinomycete. The focus of this investigation is on the protein encoded by the kijd3 gene and hereafter referred to as KijD3. On the basis of amino acid sequence analyses, KijD3 has been proposed to be an FAD-dependent oxidoreductase, which catalyzes the sixth step in d-kijanose biosynthesis by converting dTDP-3-amino-2,3,6-trideoxy-4-keto-3-methyl-d-glucose into its C-3' nitro derivative. This putative activity, however, has never been demonstrated in vivo or in vitro. Here we report the first structural study of this enzyme. For our investigation, crystals of KijD3 were grown in the presence of dTDP, and the structure was solved to 2.05-A resolution. The enzyme is a tetramer with each subunit folding into three distinct regions: a five alpha-helical bundle, an eight-stranded beta-sheet, and a second five alpha-helical bundle. The dTDP moiety is anchored to the protein via the side chains of Glu 113, Gln 254, and Arg 330. The overall fold of KijD3 places it into the well-characterized fatty acyl-CoA dehydrogenase superfamily. There is a decided cleft in each subunit with the appropriate dimensions to accommodate a dTDP-linked sugar. Strikingly, the loop defined by Phe 383 to Ala 388, which projects into the active site, contains two adjacent cis-peptide bonds, Pro 386 and Tyr 387. Activity assays demonstrate that KijD3 requires FAD for activity and that it produces a hydroxylamino product. The molecular architecture of KijD3 described in this report serves as a paradigm for a new family of enzymes that function on dTDP-linked sugar substrates.


Subject(s)
Acyl-CoA Dehydrogenase/chemistry , Acyl-CoA Dehydrogenase/metabolism , Aminoglycosides/metabolism , Flavin-Adenine Dinucleotide/metabolism , Nitroreductases/chemistry , Nitroreductases/metabolism , Nocardiaceae/enzymology , Acyl-CoA Dehydrogenase/genetics , Catalysis , Catalytic Domain , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Dimerization , Models, Molecular , Nitroreductases/genetics , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL