1
|
Odendaal C, Reijngoud DJ, Bakker BM. How lipid transfer proteins and the mitochondrial membrane shape the kinetics of β-oxidation the liver. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2025; 1866:149519. [PMID: 39428049 DOI: 10.1016/j.bbabio.2024.149519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024]
Abstract
The mitochondrial fatty acid β-oxidation (mFAO) is important for producing ATP under conditions of energetic stress, such as fasting and cold exposure. The regulation of this pathway is dependent on the kinetic properties of the enzymes involved. To better understand pathway behaviour, accurate enzyme kinetics is required. Setting up and interpreting such proper assays requires a good understanding of what influences the enzymes' kinetics. Often, knowing the buffer composition, pH, and temperature is considered to be sufficient. Many mFAO enzymes are membrane-bound, however, and their kinetic properties depend on the composition and curvature of the mitochondrial membranes. These properties are, in turn, affected by metabolite concentrations, but are rarely accounted for in kinetic assays. Especially for carnitine palmitoyltransferase 1 (CPT1), this has been shown to be of great consequence. Moreover, the enzymes of the mFAO metabolise water-insoluble acyl-CoA derivatives, which become toxic at high concentrations. In vivo, these are carried across the cytosol by intracellular lipid transfer proteins (iLTPs), such as the fatty-acid and acyl-CoA-binding proteins (FABP and ACBP, respectively). In vitro, this is often mimicked by using bovine serum albumin (BSA), which differs from the iLPTs in terms of its binding behaviour and subcellular localisation patterns. In this review, we argue that the iLTPs and membrane properties cannot be ignored when measuring or interpreting the kinetics of mFAO enzymes. They should be considered fundamental to the activity of mFAO enzymes just as pH, buffer composition, and temperature are.
Collapse
Affiliation(s)
- Christoff Odendaal
- Laboratory of Paediatrics, University Medical Centre Groningen, University of Groningen, the Netherlands
| | - Dirk-Jan Reijngoud
- Laboratory of Paediatrics, University Medical Centre Groningen, University of Groningen, the Netherlands
| | - Barbara M Bakker
- Laboratory of Paediatrics, University Medical Centre Groningen, University of Groningen, the Netherlands.
| |
Collapse
|
2
|
Yuan S, Tian S, Meng C, Ji F, Zhou B, Rushdi HE, Ye M. The Identification of Functional Genes Affecting Fat-Related Meat Traits in Meat-Type Pigeons Using Double-Digest Restriction-Associated DNA Sequencing and Molecular Docking Analysis. Animals (Basel) 2023; 13:3256. [PMID: 37893980 PMCID: PMC10603692 DOI: 10.3390/ani13203256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/26/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
The Chinese indigenous Shiqi (SQ) pigeon and the imported White King (WK) pigeon are two meat-type pigeon breeds of economical and nutritional importance in China. They displayed significant differences in such meat quality traits as intramuscular fat (IMF) content and fatty acid (FA) compositions in the breast muscles. In this study, we aimed to screen candidate genes that could affect fat-related meat quality traits in meat-type pigeons. We investigated the polymorphic variations at the genomic level using double-digest restriction-associated DNA (ddRAD) sequencing in 12 squabs of SQ and WK pigeons that exhibited significant inter-breed differences in IMF content as well as FA and amino acid compositions in the breast muscles, and screened candidate genes influencing fat-related traits in squabs through gene ontology analysis and pathway analysis. By focusing on 6019 SNPs, which were located in genes with correct annotations and had the potential to induce changes in the encoded proteins, we identified 19 genes (ACAA1, ACAA2, ACACB, ACADS, ACAT1, ACOX3, ACSBG1, ACSBG2, ACSL1, ACSL4, ELOVL6, FADS1, FADS2, HACD4, HADH, HADHA, HADHB, MECR, OXSM) as candidate genes that could affect fat-related traits in squabs. They were significantly enriched in the pathways of FA metabolism, degradation, and biosynthesis (p < 0.05). Results from molecular docking analysis further revealed that three non-synonymous amino acid alterations, ACAA1(S357N), ACAA2(T234I), and ACACB(H1418N), could alter the non-bonding interactions between the enzymatic proteins and their substrates. Since ACAA1, ACAA2, and ACACB encode rate-limiting enzymes in FA synthesis and degradation, alterations in the enzyme-substrate binding affinity may subsequently affect the catalytic efficiency of enzymes. We suggested that SNPs in these three genes were worthy of further investigation into their roles in explaining the disparities in fat-related traits in squabs.
Collapse
Affiliation(s)
- Siyu Yuan
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China; (S.Y.); (S.T.); (C.M.)
| | - Shaoqi Tian
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China; (S.Y.); (S.T.); (C.M.)
| | - Chuang Meng
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China; (S.Y.); (S.T.); (C.M.)
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Feng Ji
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100089, China;
| | - Bin Zhou
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China;
| | - Hossam E. Rushdi
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
| | - Manhong Ye
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China; (S.Y.); (S.T.); (C.M.)
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
3
|
Odendaal C, Jager EA, Martines ACMF, Vieira-Lara MA, Huijkman NCA, Kiyuna LA, Gerding A, Wolters JC, Heiner-Fokkema R, van Eunen K, Derks TGJ, Bakker BM. Personalised modelling of clinical heterogeneity between medium-chain acyl-CoA dehydrogenase patients. BMC Biol 2023; 21:184. [PMID: 37667308 PMCID: PMC10478272 DOI: 10.1186/s12915-023-01652-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 06/21/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Monogenetic inborn errors of metabolism cause a wide phenotypic heterogeneity that may even differ between family members carrying the same genetic variant. Computational modelling of metabolic networks may identify putative sources of this inter-patient heterogeneity. Here, we mainly focus on medium-chain acyl-CoA dehydrogenase deficiency (MCADD), the most common inborn error of the mitochondrial fatty acid oxidation (mFAO). It is an enigma why some MCADD patients-if untreated-are at risk to develop severe metabolic decompensations, whereas others remain asymptomatic throughout life. We hypothesised that an ability to maintain an increased free mitochondrial CoA (CoASH) and pathway flux might distinguish asymptomatic from symptomatic patients. RESULTS We built and experimentally validated, for the first time, a kinetic model of the human liver mFAO. Metabolites were partitioned according to their water solubility between the bulk aqueous matrix and the inner membrane. Enzymes are also either membrane-bound or in the matrix. This metabolite partitioning is a novel model attribute and improved predictions. MCADD substantially reduced pathway flux and CoASH, the latter due to the sequestration of CoA as medium-chain acyl-CoA esters. Analysis of urine from MCADD patients obtained during a metabolic decompensation showed an accumulation of medium- and short-chain acylcarnitines, just like the acyl-CoA pool in the MCADD model. The model suggested some rescues that increased flux and CoASH, notably increasing short-chain acyl-CoA dehydrogenase (SCAD) levels. Proteome analysis of MCADD patient-derived fibroblasts indeed revealed elevated levels of SCAD in a patient with a clinically asymptomatic state. This is a rescue for MCADD that has not been explored before. Personalised models based on these proteomics data confirmed an increased pathway flux and CoASH in the model of an asymptomatic patient compared to those of symptomatic MCADD patients. CONCLUSIONS We present a detailed, validated kinetic model of mFAO in human liver, with solubility-dependent metabolite partitioning. Personalised modelling of individual patients provides a novel explanation for phenotypic heterogeneity among MCADD patients. Further development of personalised metabolic models is a promising direction to improve individualised risk assessment, management and monitoring for inborn errors of metabolism.
Collapse
Affiliation(s)
- Christoff Odendaal
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Emmalie A Jager
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Section of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Anne-Claire M F Martines
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Marcel A Vieira-Lara
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Nicolette C A Huijkman
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Ligia A Kiyuna
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Albert Gerding
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Justina C Wolters
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Rebecca Heiner-Fokkema
- Department of Laboratory Medicine, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Karen van Eunen
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Terry G J Derks
- Section of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands.
| | - Barbara M Bakker
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands.
| |
Collapse
|
4
|
Ikeda T, Ishikawa T, Ninagawa S, Okada T, Ono M, Mori K. Proteomic analysis of fatty liver induced by starvation of medaka fish larvae. Cell Struct Funct 2023; 48:123-133. [PMID: 37380437 PMCID: PMC10915113 DOI: 10.1247/csf.23014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/23/2023] [Indexed: 06/30/2023] Open
Abstract
When medaka fish (Oryzias latipes) larvae are grown in the absence of exogenous nutrition, the liver becomes dark and positive to Oil Red O staining from 7 days post-hatch (dph). We determined the mechanism of this starvation-induced development of fatty liver by proteomic analysis using livers obtained from larvae grown in the presence or absence of 2% glucose at 5 dph. Results showed that changes in the expression levels of enzymes involved in glycolysis or the tricarboxylic acid cycle were modest, whereas the expression levels of enzymes involved in amino acid catabolism or β-oxidation of fatty acids were significantly elevated, suggesting that they become major energy sources under starvation conditions. Expression levels of enzymes for the uptake and β-oxidation of fatty acids as well as synthesis of triacylglycerol were elevated, whereas those for the synthesis of cholesterol as well as export of cholesterol and triacylglycerol were decreased under starvation conditions, which explains the accumulation of triacylglycerol in the liver. Our results provide the basis for future research to understand how gene malfunction(s) affects the development of fatty liver, which can lead to nonalcoholic steatohepatitis and then to liver cirrhosis.Key words: amino acid catabolism, β-oxidation, triacylglycerol, cholesterol, export.
Collapse
Affiliation(s)
- Tomoyo Ikeda
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Tokiro Ishikawa
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Satoshi Ninagawa
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Tetsuya Okada
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Masaya Ono
- National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Kazutoshi Mori
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
5
|
Bromodomain-containing protein 4 (BRD4) as an epigenetic regulator of fatty acid metabolism genes and ferroptosis. Cell Death Dis 2022; 13:912. [PMID: 36309482 PMCID: PMC9617950 DOI: 10.1038/s41419-022-05344-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/28/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
Abstract
Reprogramming lipid metabolism is considered a fundamental step in tumourigenesis that influences ferroptosis. However, molecular mechanisms between lipid metabolism and ferroptosis remain largely unknown. Results from the drug screening of 464 inhibitors (for 164 targets) applied to ferroptosis cells indicated that 4 inhibitors targeted bromodomain-containing protein 4 (BRD4) significantly inhibiting erastin-induced ferroptosis. Functional studies proved that the loss of BRD4 weakened oxidative catabolism in mitochondria, protecting cells from the excessive accumulation of lipid peroxides. Mechanism research revealed that the transcriptional levels of fatty acid metabolism-related genes (HADH, ACSL1 and ACAA2) participating in the β-oxidation of fatty acids (FAO) and polyunsaturated fatty acids (PUFAs) synthesis depended on the activity of super-enhancers (SEs) formed by BRD4 and HMGB2 in their promoter regions. Conclusively, this study demonstrated that BRD4 was indispensable for fatty acid metabolism based on its epigenetic regulatory mechanisms and affecting erastin-induced ferroptosis, providing a new theoretical reference for understanding the relationship between lipid metabolism and ferroptosis deeply.
Collapse
|
6
|
Zhang H, Pan E, Zhang Y, Zhao C, Liu Q, Pu Y, Yin L. LncRNA RPL34-AS1 suppresses the proliferation, migration and invasion of esophageal squamous cell carcinoma via targeting miR-575/ACAA2 axis. BMC Cancer 2022; 22:1017. [PMID: 36162992 PMCID: PMC9511711 DOI: 10.1186/s12885-022-10104-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 09/19/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are abnormally expressed in a broad type of cancers and play significant roles that regulate tumor development and metastasis. However, the pathological roles of lncRNAs in esophageal squamous cell carcinoma (ESCC) remain largely unknown. Here we aimed to investigate the role and regulatory mechanism of the novel lncRNA RPL34-AS1 in the development and progression of ESCC. METHODS The expression level of RPL34-AS1 in ESCC tissues and cell lines was determined by RT-qPCR. Functional experiments in vitro and in vivo were employed to explore the effects of RPL34-AS1 on tumor growth in ESCC cells. Mechanistically, fluorescence in situ hybridization (FISH), bioinformatics analyses, luciferase reporter assay, RNA immunoprecipitation (RIP) assay and western blot assays were used to detect the regulatory relationship between RPL34-AS1, miR-575 and ACAA2. RESULTS RPL34-AS1 was significantly down-regulated in ESCC tissues and cells, which was negatively correlated with overall survival in ESCC patients. Functionally, upregulation of RPL34-AS1 dramatically suppressed ESCC cell proliferation, colony formation, invasion and migration in vitro, whereas knockdown of RPL34-AS1 elicited the opposite function. Consistently, overexpression of RPL34-AS1 inhibited tumor growth in vivo. Mechanistically, RPL34-AS1 acted as a competing endogenous RNA (ceRNA) of miR-575 to relieve the repressive effect of miR-575 on its target ACAA2, then suppressed the tumorigenesis of ESCC. CONCLUSIONS Our results reveal a role for RPL34-AS1 in ESCC tumorigenesis and may provide a strategy for using RPL34-AS1 as a potential biomarker and an effect target for patients with ESCC.
Collapse
Affiliation(s)
- Hu Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, 87 Dingjiaqiao Street, Nanjing, 210009, People's Republic of China
| | - Enchun Pan
- Huaian Center for Disease Control and Prevention, Huaian, 223001, People's Republic of China
| | - Ying Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, 87 Dingjiaqiao Street, Nanjing, 210009, People's Republic of China
| | - Chao Zhao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, 87 Dingjiaqiao Street, Nanjing, 210009, People's Republic of China
| | - Qiwei Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, 87 Dingjiaqiao Street, Nanjing, 210009, People's Republic of China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, 87 Dingjiaqiao Street, Nanjing, 210009, People's Republic of China
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, 87 Dingjiaqiao Street, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
7
|
Jiao M, He W, Ouyang Z, Shi Q, Wen Y. Progress in structural and functional study of the bacterial phenylacetic acid catabolic pathway, its role in pathogenicity and antibiotic resistance. Front Microbiol 2022; 13:964019. [PMID: 36160191 PMCID: PMC9493321 DOI: 10.3389/fmicb.2022.964019] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Phenylacetic acid (PAA) is a central intermediate metabolite involved in bacterial degradation of aromatic components. The bacterial PAA pathway mainly contains 12 enzymes and a transcriptional regulator, which are involved in biofilm formation and antimicrobial activity. They are present in approximately 16% of the sequenced bacterial genome. In this review, we have summarized the PAA distribution in microbes, recent structural and functional study progress of the enzyme families of the bacterial PAA pathway, and their role in bacterial pathogenicity and antibiotic resistance. The enzymes of the bacterial PAA pathway have shown potential as an antimicrobial drug target for biotechnological applications in metabolic engineering.
Collapse
Affiliation(s)
- Min Jiao
- Department of Critical Care Medicine, Center for Microbiome Research of Med-X Institute, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Wenbo He
- Department of Critical Care Medicine, Center for Microbiome Research of Med-X Institute, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Zhenlin Ouyang
- Department of Critical Care Medicine, Center for Microbiome Research of Med-X Institute, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Qindong Shi
- Department of Critical Care Medicine, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yurong Wen
- Department of Critical Care Medicine, Center for Microbiome Research of Med-X Institute, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Department of Critical Care Medicine, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education Health Science Center, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
8
|
Zhang H, Zhao C, Liu Q, Zhang Y, Luo K, Pu Y, Yin L. Dysregulation of fatty acid metabolism associated with esophageal inflammation of ICR mice induced by nitrosamines exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 297:118680. [PMID: 34915095 DOI: 10.1016/j.envpol.2021.118680] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/01/2021] [Accepted: 12/11/2021] [Indexed: 06/14/2023]
Abstract
Nitrosamines, as ubiquitous environmental carcinogens with adverse impact on human health, were crucial inducers of esophageal cancer (EC). Esophageal inflammation (EI) was an important biological process and considered to be associated with the progression of EC. However, the underlying regulatory mechanism of EI process caused by nitrosamines exposure remained largely unclear. In this study, a metabolomics approach based on mass spectrometry was utilized to explore the effect of nitrosamines exposure to ICR mice. Also, the changes of pivotal metabolic enzyme levels, urinary nitrosamines and histopathological analysis were evaluated. The results showed that nitrosamines exposure was intimately interrelated with EI process in mice. Metabolomics profiling analysis indicated that nitrosamines caused significant alterations of metabolic pathway predominantly enriched in fatty acid metabolism. Targeted metabolomics analysis revealed that nitrosamines promoted decomposition of fatty acids and facilitated fatty acid β-oxidation (FAO) of mice. The significant increase of carnitine palmitoyltransferase 1 (CPT1) and downregulation of acetyl-CoA acyltransferase 2 (ACAA2) would promote FAO in EI process induced by nitrosamines. Additionally, the exposure levels of more than half of nitrosamines in urine were correlated with inflammatory fatty acid biomarkers. Overall, this study found that EI triggered by nitrosamines may be associated with the promotion of FAO, and provided novel insights for evaluating the underlying mechanism of environmental pollutant-caused toxicity based on metabolomics.
Collapse
Affiliation(s)
- Hu Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, People's Republic of China
| | - Chao Zhao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, People's Republic of China
| | - Qiwei Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, People's Republic of China
| | - Ying Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, People's Republic of China
| | - Kai Luo
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, People's Republic of China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, People's Republic of China
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
9
|
Caswell BT, de Carvalho CC, Nguyen H, Roy M, Nguyen T, Cantu DC. Thioesterase enzyme families: Functions, structures, and mechanisms. Protein Sci 2022; 31:652-676. [PMID: 34921469 PMCID: PMC8862431 DOI: 10.1002/pro.4263] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022]
Abstract
Thioesterases are enzymes that hydrolyze thioester bonds in numerous biochemical pathways, for example in fatty acid synthesis. This work reports known functions, structures, and mechanisms of updated thioesterase enzyme families, which are classified into 35 families based on sequence similarity. Each thioesterase family is based on at least one experimentally characterized enzyme, and most families have enzymes that have been crystallized and their tertiary structure resolved. Classifying thioesterases into families allows to predict tertiary structures and infer catalytic residues and mechanisms of all sequences in a family, which is particularly useful because the majority of known protein sequence have no experimental characterization. Phylogenetic analysis of experimentally characterized thioesterases that have structures with the two main structural folds reveal convergent and divergent evolution. Based on tertiary structure superimposition, catalytic residues are predicted.
Collapse
Affiliation(s)
- Benjamin T. Caswell
- Department of Chemical and Materials EngineeringUniversity of Nevada, RenoRenoNevadaUSA
| | - Caio C. de Carvalho
- Department of Chemical and Materials EngineeringUniversity of Nevada, RenoRenoNevadaUSA
| | - Hung Nguyen
- Department of Computer Science and EngineeringUniversity of Nevada, RenoRenoNevadaUSA
| | - Monikrishna Roy
- Department of Computer Science and EngineeringUniversity of Nevada, RenoRenoNevadaUSA
| | - Tin Nguyen
- Department of Computer Science and EngineeringUniversity of Nevada, RenoRenoNevadaUSA
| | - David C. Cantu
- Department of Chemical and Materials EngineeringUniversity of Nevada, RenoRenoNevadaUSA
| |
Collapse
|
10
|
Kretzschmar T, Wu JMF, Schulze PC. Mitochondrial Homeostasis Mediates Lipotoxicity in the Failing Myocardium. Int J Mol Sci 2021; 22:1498. [PMID: 33540894 PMCID: PMC7867320 DOI: 10.3390/ijms22031498] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 01/17/2023] Open
Abstract
Heart failure remains the most common cause of death in the industrialized world. In spite of new therapeutic interventions that are constantly being developed, it is still not possible to completely protect against heart failure development and progression. This shows how much more research is necessary to understand the underlying mechanisms of this process. In this review, we give a detailed overview of the contribution of impaired mitochondrial dynamics and energy homeostasis during heart failure progression. In particular, we focus on the regulation of fatty acid metabolism and the effects of fatty acid accumulation on mitochondrial structural and functional homeostasis.
Collapse
Affiliation(s)
| | | | - P. Christian Schulze
- Department of Internal Medicine I, University Hospital Jena, 07747 Jena, Thüringen, Germany; (T.K.); (J.M.F.W.)
| |
Collapse
|
11
|
Hathazi D, Griffin H, Jennings MJ, Giunta M, Powell C, Pearce SF, Munro B, Wei W, Boczonadi V, Poulton J, Pyle A, Calabrese C, Gomez‐Duran A, Schara U, Pitceathly RDS, Hanna MG, Joost K, Cotta A, Paim JF, Navarro MM, Duff J, Mattman A, Chapman K, Servidei S, Della Marina A, Uusimaa J, Roos A, Mootha V, Hirano M, Tulinius M, Giri M, Hoffmann EP, Lochmüller H, DiMauro S, Minczuk M, Chinnery PF, Müller JS, Horvath R. Metabolic shift underlies recovery in reversible infantile respiratory chain deficiency. EMBO J 2020; 39:e105364. [PMID: 33128823 PMCID: PMC7705457 DOI: 10.15252/embj.2020105364] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/31/2020] [Accepted: 09/09/2020] [Indexed: 12/23/2022] Open
Abstract
Reversible infantile respiratory chain deficiency (RIRCD) is a rare mitochondrial myopathy leading to severe metabolic disturbances in infants, which recover spontaneously after 6-months of age. RIRCD is associated with the homoplasmic m.14674T>C mitochondrial DNA mutation; however, only ~ 1/100 carriers develop the disease. We studied 27 affected and 15 unaffected individuals from 19 families and found additional heterozygous mutations in nuclear genes interacting with mt-tRNAGlu including EARS2 and TRMU in the majority of affected individuals, but not in healthy carriers of m.14674T>C, supporting a digenic inheritance. Our transcriptomic and proteomic analysis of patient muscle suggests a stepwise mechanism where first, the integrated stress response associated with increased FGF21 and GDF15 expression enhances the metabolism modulated by serine biosynthesis, one carbon metabolism, TCA lipid oxidation and amino acid availability, while in the second step mTOR activation leads to increased mitochondrial biogenesis. Our data suggest that the spontaneous recovery in infants with digenic mutations may be modulated by the above described changes. Similar mechanisms may explain the variable penetrance and tissue specificity of other mtDNA mutations and highlight the potential role of amino acids in improving mitochondrial disease.
Collapse
|
12
|
Leandro J, Houten SM. The lysine degradation pathway: Subcellular compartmentalization and enzyme deficiencies. Mol Genet Metab 2020; 131:14-22. [PMID: 32768327 DOI: 10.1016/j.ymgme.2020.07.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 02/07/2023]
Abstract
Lysine degradation via formation of saccharopine is a pathway confined to the mitochondria. The second pathway for lysine degradation, the pipecolic acid pathway, is not yet fully elucidated and known enzymes are localized in the mitochondria, cytosol and peroxisome. The tissue-specific roles of these two pathways are still under investigation. The lysine degradation pathway is clinically relevant due to the occurrence of two severe neurometabolic disorders, pyridoxine-dependent epilepsy (PDE) and glutaric aciduria type 1 (GA1). The existence of three other disorders affecting lysine degradation without apparent clinical consequences opens up the possibility to find alternative therapeutic strategies for PDE and GA1 through pathway modulation. A better understanding of the mechanisms, compartmentalization and interplay between the different enzymes and metabolites involved in lysine degradation is of utmost importance.
Collapse
Affiliation(s)
- João Leandro
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sander M Houten
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
13
|
Liu L, Zhou S, Deng Y. The 3-ketoacyl-CoA thiolase: an engineered enzyme for carbon chain elongation of chemical compounds. Appl Microbiol Biotechnol 2020; 104:8117-8129. [PMID: 32830293 DOI: 10.1007/s00253-020-10848-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/09/2020] [Accepted: 08/17/2020] [Indexed: 01/03/2023]
Abstract
Because of their function of catalyzing the rearrangement of the carbon chains, thiolases have attracted increasing attentions over the past decades. The 3-ketoacyl-CoA thiolase (KAT) is a member of the thiolase, which is capable of catalyzing the Claisen condensation reaction between the two acyl-CoAs, thereby achieving carbon chain elongation. In this way, diverse value-added compounds might be synthesized starting from simple small CoA thioesters. However, most KATs are hampered by low stability and poor substrate specificity, which has hindered the development of large-scale biosynthesis. In this review, the common characteristics in the three-dimensional structure of KATs from different sources are summarized. Moreover, structure-guided rational engineering is discussed as a strategy for enhancing the performance of KATs. Finally, we reviewed the metabolic engineering applications of KATs for producing various energy-storage molecules, such as n-butanol, fatty acids, dicarboxylic acids, and polyhydroxyalkanoates. KEY POINTS: • Summarize the structural characteristics and catalyzation mechanisms of KATs. • Review on the rational engineering to enhance the performance of KATs. • Discuss the applications of KATs for producing energy-storage molecules.
Collapse
Affiliation(s)
- Lixia Liu
- National Engineering Laboratory for Cereal Fermentation Technology (NELCF), Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China.,Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China
| | - Shenghu Zhou
- National Engineering Laboratory for Cereal Fermentation Technology (NELCF), Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China.,Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China
| | - Yu Deng
- National Engineering Laboratory for Cereal Fermentation Technology (NELCF), Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China. .,Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China.
| |
Collapse
|
14
|
Aspergillus fumigatus Mitochondrial Acetyl Coenzyme A Acetyltransferase as an Antifungal Target. Appl Environ Microbiol 2020; 86:AEM.02986-19. [PMID: 32005728 PMCID: PMC7082573 DOI: 10.1128/aem.02986-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 01/15/2020] [Indexed: 12/27/2022] Open
Abstract
Ergosterol plays an important role in maintaining cell membrane sterol homeostasis in fungi, and as such, it is considered an effective target in antifungal chemotherapy. In yeast, the enzyme acetyl-coenzyme A (CoA) acetyltransferase (ERG10) catalyzes the Claisen condensation of two acetyl-CoA molecules to acetoacetyl-CoA in the ergosterol biosynthesis pathway and is reported as being critical for cell viability. Using yeast ERG10 for alignment, two orthologues, AfERG10A (AFUB_000550) and AfERG10B (AFUB_083570), were discovered in the opportunistic fungal pathogen Aspergillus fumigatus Despite the essentiality of AfERG10B having been previously validated, the biological function of AfERG10A remains unclear. In this study, we have characterized recombinant AfERG10A as a functional acetyl-CoA acetyltransferase catalyzing both synthetic and degradative reactions. Unexpectedly, AfERG10A localizes to the mitochondria in A. fumigatus, as shown by C-terminal green fluorescent protein (GFP) tag fusion. Both knockout and inducible promoter strategies demonstrate that Aferg10A is essential for the survival of A. fumigatus The reduced expression of Aferg10A leads to severe morphological defects and increased susceptibility to oxidative and cell wall stresses. Although the catalytic mechanism of acetyl-CoA acetyltransferase family is highly conserved, the crystal structure of AfERG10A and its complex with CoA are solved, revealing four substitutions within the CoA binding site that are different from human orthologues. Taken together, our combination of genetic and structural studies demonstrates that mitochondrial AfERG10A is essential for A. fumigatus cell viability and could be a potential drug target to feed the antifungal drug development pipeline.IMPORTANCE A growing number of people worldwide are suffering from invasive aspergillosis caused by the human opportunistic fungal pathogen A. fumigatus Current therapeutic options rely on a limited repertoire of antifungals. Ergosterol is an essential component of the fungal cell membrane as well as a target of current antifungals. Approximately 20 enzymes are involved in ergosterol biosynthesis, of which acetyl-CoA acetyltransferase (ACAT) is the first enzyme. Two ACATs in A. fumigatus are AfErg10A and AfErg10B. However, the biological function of AfErg10A is yet to be investigated. In this study, we showed that AfErg10A is localized in the mitochondria and is essential for A. fumigatus survival and morphological development. In combination with structural studies, we validated AfErg10A as a potential drug target that will facilitate the development of novel antifungals and improve the efficiency of existing drugs.
Collapse
|
15
|
Structural basis for differentiation between two classes of thiolase: Degradative vs biosynthetic thiolase. JOURNAL OF STRUCTURAL BIOLOGY-X 2020; 4:100018. [PMID: 32647822 PMCID: PMC7337054 DOI: 10.1016/j.yjsbx.2019.100018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/17/2019] [Accepted: 12/27/2019] [Indexed: 11/23/2022]
Abstract
Thiolases are a well characterized family of enzymes with two distinct categories: degradative, β-ketoadipyl-CoA thiolases and biosynthetic, acetoacetyl-CoA thiolases. Both classes share an identical catalytic triad but catalyze reactions in opposite directions. Moreover, it is established that in contrast to the biosynthetic thiolases the degradative thiolases can accept substrates with broad chain lengths. Hitherto, no residue or structural pattern has been recognized that might help to discern the two thiolases, here we exploit, a tetrameric degradative thiolase from Pseudomonas putida KT2440 annotated as PcaF, as a model system to understand features which distinguishes the two classes using structural studies and bioinformatics analyses. Degradative thiolases have different active site architecture when compared to biosynthetic thiolases, demonstrating the dissimilar chemical nature of the active site architecture. Both thiolases deploy different "anchoring residues" to tether the large Coenzyme A (CoA) or CoA derivatives. Interestingly, the H356 of the catalytic triad in PcaF is directly involved in tethering the CoA/CoA derivatives into the active site and we were able to trap a gridlocked thiolase structure of the H356A mutant, where the CoA was found to be covalently linked to the catalytic cysteine residue, inhibiting the overall reaction. Further, X-ray structures with two long chain CoA derivatives, hexanal-CoA and octanal-CoA helped in delineating the long tunnel of 235 Å2 surface area in PcaF and led to identification of a unique covering loop exclusive to degradative thiolases that plays an active role in determining the tunnel length and the nature of the binding substrate.
Collapse
Key Words
- A-mutant-HAL-CoA, A-mutant-hexanal CoA complex
- A-mutant-Hex-CoA, A-mutant-Hexanoyl CoA complex
- A-mutants, H356A Mutant
- AA-mutants, H356A-C386A Mutant
- AS-mutant-OAL-CoA, AS-mutant-octanal CoA complex
- AS-mutant-Oct-CoA, AS-mutant-Octanoyl CoA complex
- AS-mutants, H356A-C90S Mutant
- Covalent locking
- Covering loop
- HAL, hexanal
- Hex-CoA, Hexanoyl CoA
- Hexanoyl CoA
- Mtb-thiolase, Mycobacterium tuberculosis thiolase
- OAL, octanal
- Oct-CoA, Octanoyl CoA
- Octanoyl CoA
- PcaF, β-ketoadipyl-CoA thiolase
- Tunnel
- Zr-thiolase, Zoogleria ramigera thiolase
Collapse
|
16
|
Abdelkreem E, Harijan RK, Yamaguchi S, Wierenga RK, Fukao T. Mutation update on ACAT1 variants associated with mitochondrial acetoacetyl-CoA thiolase (T2) deficiency. Hum Mutat 2019; 40:1641-1663. [PMID: 31268215 PMCID: PMC6790690 DOI: 10.1002/humu.23831] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 05/27/2019] [Accepted: 05/31/2019] [Indexed: 02/05/2023]
Abstract
Mitochondrial acetoacetyl-CoA thiolase (T2, encoded by the ACAT1 gene) deficiency is an inherited disorder of ketone body and isoleucine metabolism. It typically manifests with episodic ketoacidosis. The presence of isoleucine-derived metabolites is the key marker for biochemical diagnosis. To date, 105 ACAT1 variants have been reported in 149 T2-deficient patients. The 56 disease-associated missense ACAT1 variants have been mapped onto the crystal structure of T2. Almost all these missense variants concern residues that are completely or partially buried in the T2 structure. Such variants are expected to cause T2 deficiency by having lower in vivo T2 activity because of lower folding efficiency and/or stability. Expression and activity data of 30 disease-associated missense ACAT1 variants have been measured by expressing them in human SV40-transformed fibroblasts. Only two variants (p.Cys126Ser and p.Tyr219His) appear to have equal stability as wild-type. For these variants, which are inactive, the side chains point into the active site. In patients with T2 deficiency, the genotype does not correlate with the clinical phenotype but exerts a considerable effect on the biochemical phenotype. This could be related to variable remaining residual T2 activity in vivo and has important clinical implications concerning disease management and newborn screening.
Collapse
Affiliation(s)
- Elsayed Abdelkreem
- Department of Pediatrics, Graduate School of MedicineGifu UniversityGifuJapan
- Department of Pediatrics, Faculty of MedicineSohag UniversitySohagEgypt
| | - Rajesh K. Harijan
- Department of BiochemistryAlbert Einstein College of MedicineNew YorkNew York
| | - Seiji Yamaguchi
- Department of PediatricsShimane University School of MedicineIzumoJapan
| | | | - Toshiyuki Fukao
- Department of Pediatrics, Graduate School of MedicineGifu UniversityGifuJapan
| |
Collapse
|
17
|
Tassoni R, Blok A, Pannu NS, Ubbink M. New Conformations of Acylation Adducts of Inhibitors of β-Lactamase from Mycobacterium tuberculosis. Biochemistry 2019; 58:997-1009. [PMID: 30632739 PMCID: PMC6383187 DOI: 10.1021/acs.biochem.8b01085] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
![]()
Mycobacterium tuberculosis (Mtb), the main causative
agent of tuberculosis (TB), is naturally resistant to β-lactam
antibiotics due to the production of the extended spectrum β-lactamase
BlaC. β-Lactam/β-lactamase inhibitor combination therapies
can circumvent the BlaC-mediated resistance of Mtb and are promising
treatment options against TB. However, still little is known of the
exact mechanism of BlaC inhibition by the β-lactamase inhibitors
currently approved for clinical use, clavulanic acid, sulbactam, tazobactam,
and avibactam. Here, we present the X-ray diffraction crystal structures
of the acyl-enzyme adducts of wild-type BlaC with the four inhibitors.
The +70 Da adduct derived from clavulanate and the trans-enamine acylation adducts of sulbactam and tazobactam are reported.
BlaC in complex with avibactam revealed two inhibitor conformations.
Preacylation binding could not be observed because inhibitor binding
was not detected in BlaC variants carrying a substitution of the active
site serine 70 to either alanine or cysteine, by crystallography,
ITC or NMR. These results suggest that the catalytic serine 70 is
necessary not only for enzyme acylation but also for increasing BlaC
affinity for inhibitors in the preacylation state. The structure of
BlaC with the serine to cysteine mutation showed a covalent linkage
of the cysteine 70 Sγ atom to the nearby amino group of lysine
73. The differences of adduct conformations between BlaC and other
β-lactamases are discussed.
Collapse
Affiliation(s)
- Raffaella Tassoni
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , Leiden 2333CC , The Netherlands
| | - Anneloes Blok
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , Leiden 2333CC , The Netherlands
| | - Navraj S Pannu
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , Leiden 2333CC , The Netherlands
| | - Marcellus Ubbink
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , Leiden 2333CC , The Netherlands
| |
Collapse
|
18
|
The peroxisomal zebrafish SCP2-thiolase (type-1) is a weak transient dimer as revealed by crystal structures and native mass spectrometry. Biochem J 2019; 476:307-332. [PMID: 30573650 DOI: 10.1042/bcj20180788] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/12/2018] [Accepted: 12/20/2018] [Indexed: 12/19/2022]
Abstract
The SCP2 (sterol carrier protein 2)-thiolase (type-1) functions in the vertebrate peroxisomal, bile acid synthesis pathway, converting 24-keto-THC-CoA and CoA into choloyl-CoA and propionyl-CoA. This conversion concerns the β-oxidation chain shortening of the steroid fatty acyl-moiety of 24-keto-THC-CoA. This class of dimeric thiolases has previously been poorly characterized. High-resolution crystal structures of the zebrafish SCP2-thiolase (type-1) now reveal an open catalytic site, shaped by residues of both subunits. The structure of its non-dimerized monomeric form has also been captured in the obtained crystals. Four loops at the dimer interface adopt very different conformations in the monomeric form. These loops also shape the active site and their structural changes explain why a competent active site is not present in the monomeric form. Native mass spectrometry studies confirm that the zebrafish SCP2-thiolase (type-1) as well as its human homolog are weak transient dimers in solution. The crystallographic binding studies reveal the mode of binding of CoA and octanoyl-CoA in the active site, highlighting the conserved geometry of the nucleophilic cysteine, the catalytic acid/base cysteine and the two oxyanion holes. The dimer interface of SCP2-thiolase (type-1) is equally extensive as in other thiolase dimers; however, it is more polar than any of the corresponding interfaces, which correlates with the notion that the enzyme forms a weak transient dimer. The structure comparison of the monomeric and dimeric forms suggests functional relevance of this property. These comparisons provide also insights into the structural rearrangements that occur when the folded inactive monomers assemble into the mature dimer.
Collapse
|
19
|
Mitochondrial β-oxidation of saturated fatty acids in humans. Mitochondrion 2018; 46:73-90. [PMID: 29551309 DOI: 10.1016/j.mito.2018.02.009] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/04/2017] [Accepted: 02/27/2018] [Indexed: 12/30/2022]
Abstract
Mitochondrial β-oxidation of fatty acids generates acetyl-coA, NADH and FADH2. Acyl-coA synthetases catalyze the binding of fatty acids to coenzyme A to form fatty acyl-coA thioesters, the first step in the intracellular metabolism of fatty acids. l-carnitine system facilitates the transport of fatty acyl-coA esters across the mitochondrial membrane. Carnitine palmitoyltransferase-1 transfers acyl groups from coenzyme A to l-carnitine, forming acyl-carnitine esters at the outer mitochondrial membrane. Carnitine acyl-carnitine translocase exchanges acyl-carnitine esters that enter the mitochondria, by free l-carnitine. Carnitine palmitoyltransferase-2 converts acyl-carnitine esters back to acyl-coA esters at the inner mitochondrial membrane. The β-oxidation pathway of fatty acyl-coA esters includes four reactions. Fatty acyl-coA dehydrogenases catalyze the introduction of a double bond at the C2 position, producing 2-enoyl-coA esters and reducing equivalents that are transferred to the respiratory chain via electron transferring flavoprotein. Enoyl-coA hydratase catalyzes the hydration of the double bond to generate a 3-l-hydroxyacyl-coA derivative. 3-l-hydroxyacyl-coA dehydrogenase catalyzes the formation of a 3-ketoacyl-coA intermediate. Finally, 3-ketoacyl-coA thiolase catalyzes the cleavage of the chain, generating acetyl-coA and a fatty acyl-coA ester two carbons shorter. Mitochondrial trifunctional protein catalyzes the three last steps in the β-oxidation of long-chain and medium-chain fatty acyl-coA esters while individual enzymes catalyze the β-oxidation of short-chain fatty acyl-coA esters. Clinical phenotype of fatty acid oxidation disorders usually includes hypoketotic hypoglycemia triggered by fasting or infections, skeletal muscle weakness, cardiomyopathy, hepatopathy, and neurological manifestations. Accumulation of non-oxidized fatty acids promotes their conjugation with glycine and l-carnitine and alternate ways of oxidation, such as ω-oxidation.
Collapse
|
20
|
Blaisse MR, Fu B, Chang MCY. Structural and Biochemical Studies of Substrate Selectivity in Ascaris suum Thiolases. Biochemistry 2018; 57:3155-3166. [PMID: 29381332 DOI: 10.1021/acs.biochem.7b01123] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Thiolases are a class of carbon-carbon bond forming enzymes with important applications in biotechnology and metabolic engineering as they provide a general method for the condensation of two acyl coenzyme A (CoA) substrates. As such, developing a greater understanding of their substrate selectivity would expand our ability to engineer the enzymatic or microbial production of a broad range of small-molecule targets. Here, we report the crystal structures and biochemical characterization of Acat2 and Acat5, two biosynthetic thiolases from Ascaris suum with varying selectivity toward branched compared to linear compounds. The structure of the Acat2-C91S mutant bound to propionyl-CoA shows that the terminal methyl group of the substrate, representing the α-branch point, is directed toward the conserved Phe 288 and Met 158 residues. In Acat5, the Phe ring is rotated to accommodate a hydroxyl-π interaction with an adjacent Thr side chain, decreasing space in the binding pocket and possibly accounting for its strong preference for linear substrates compared to Acat2. Comparison of the different Acat thiolase structures shows that Met 158 is flexible, adopting alternate conformations with the side chain rotated toward or away from a covering loop at the back of the active site. Mutagenesis of residues in the covering loop in Acat5 with the corresponding residues from Acat2 allows for highly increased accommodation of branched substrates, whereas the converse mutations do not significantly affect Acat2 substrate selectivity. Our results suggest an important contribution of second-shell residues to thiolase substrate selectivity and offer insights into engineering this enzyme class.
Collapse
|
21
|
Li XX, Wang ZJ, Zheng Y, Guan YF, Yang PB, Chen X, Peng C, He JP, Ai YL, Wu SF, Chien KY, Wu Q, Chen HZ. Nuclear Receptor Nur77 Facilitates Melanoma Cell Survival under Metabolic Stress by Protecting Fatty Acid Oxidation. Mol Cell 2018; 69:480-492.e7. [PMID: 29395065 DOI: 10.1016/j.molcel.2018.01.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 10/02/2017] [Accepted: 12/29/2017] [Indexed: 12/13/2022]
Abstract
Fatty acid oxidation (FAO) is crucial for cells to overcome metabolic stress by providing ATP and NADPH. However, the mechanism by which FAO is regulated in tumors remains elusive. Here we show that Nur77 is required for the metabolic adaptation of melanoma cells by protecting FAO. Glucose deprivation activates ERK2 to phosphorylate and induce Nur77 translocation to the mitochondria, where Nur77 binds to TPβ, a rate-limiting enzyme in FAO. Although TPβ activity is normally inhibited by oxidation under glucose deprivation, the Nur77-TPβ association results in Nur77 self-sacrifice to protect TPβ from oxidation. FAO is therefore able to maintain NADPH and ATP levels and prevent ROS increase and cell death. The Nur77-TPβ interaction further promotes melanoma metastasis by facilitating circulating melanoma cell survival. This study demonstrates a novel regulatory function of Nur77 with linkage of the FAO-NADPH-ROS pathway during metabolic stress, suggesting Nur77 as a potential therapeutic target in melanoma.
Collapse
Affiliation(s)
- Xiao-Xue Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Zhi-Jing Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Yu Zheng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Yun-Feng Guan
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Peng-Bo Yang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Jian-Ping He
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Yuan-Li Ai
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Sheng-Fu Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Kun-Yi Chien
- Molecular Medicine Research Center, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan 333, Taiwan
| | - Qiao Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, China.
| | - Hang-Zi Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, China.
| |
Collapse
|
22
|
Kel I, Chang Z, Galluccio N, Romeo M, Beretta S, Diomede L, Mezzelani A, Milanesi L, Dieterich C, Merelli I. SPIRE, a modular pipeline for eQTL analysis of RNA-Seq data, reveals a regulatory hotspot controlling miRNA expression in C. elegans. MOLECULAR BIOSYSTEMS 2017; 12:3447-3458. [PMID: 27722582 DOI: 10.1039/c6mb00453a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The interpretation of genome-wide association study is difficult, as it is hard to understand how polymorphisms can affect gene regulation, in particular for trans-regulatory elements located far from their controlling gene. Using RNA or protein expression data as phenotypes, it is possible to correlate their variations with specific genotypes. This technique is usually referred to as expression Quantitative Trait Loci (eQTLs) analysis and only few packages exist for the integration of genotype patterns and expression profiles. In particular, tools are needed for the analysis of next-generation sequencing (NGS) data on a genome-wide scale, which is essential to identify eQTLs able to control a large number of genes (hotspots). Here we present SPIRE (Software for Polymorphism Identification Regulating Expression), a generic, modular and functionally highly flexible pipeline for eQTL processing. SPIRE integrates different univariate and multivariate approaches for eQTL analysis, paying particular attention to the scalability of the procedure in order to support cis- as well as trans-mapping, thus allowing the identification of hotspots in NGS data. In particular, we demonstrated how SPIRE can handle big association study datasets, reproducing published results and improving the identification of trans-eQTLs. Furthermore, we employed the pipeline to analyse novel data concerning the genotypes of two different C. elegans strains (N2 and Hawaii) and related miRNA expression data, obtained using RNA-Seq. A miRNA regulatory hotspot was identified in chromosome 1, overlapping the transcription factor grh-1, known to be involved in the early phases of embryonic development of C. elegans. In a follow-up qPCR experiment we were able to verify most of the predicted eQTLs, as well as to show, for a novel miRNA, a significant difference in the sequences of the two analysed strains of C. elegans. SPIRE is publicly available as open source software at , together with some example data, a readme file, supplementary material and a short tutorial.
Collapse
Affiliation(s)
- Ivan Kel
- Instituto di Tecnologie Biomediche - Consiglio Nazionale delle Ricerche, via F.lli Cervi 93, 20090, Segrate, Milano, Italy.
| | - Zisong Chang
- Max Delbrück Center for Molecular Medicine, Berlin Institute for Medical Systems Biology, Robert-Rössle-Straße 10, 13125, Berlin, Germany.
| | - Nadia Galluccio
- Instituto di Tecnologie Biomediche - Consiglio Nazionale delle Ricerche, via F.lli Cervi 93, 20090, Segrate, Milano, Italy.
| | - Margherita Romeo
- Dipartimento di Biochimica e Farmacologia Molecolare, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Via Giuseppe La Masa 19, Milan, Italy.
| | - Stefano Beretta
- Dipartimento di Informatica Sistemistica e Comunicazione, Università degli studi di Milano-Biccoca, Viale Sarca 336, 20125 Milano, Italy.
| | - Luisa Diomede
- Dipartimento di Biochimica e Farmacologia Molecolare, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Via Giuseppe La Masa 19, Milan, Italy.
| | - Alessandra Mezzelani
- Instituto di Tecnologie Biomediche - Consiglio Nazionale delle Ricerche, via F.lli Cervi 93, 20090, Segrate, Milano, Italy.
| | - Luciano Milanesi
- Instituto di Tecnologie Biomediche - Consiglio Nazionale delle Ricerche, via F.lli Cervi 93, 20090, Segrate, Milano, Italy.
| | - Christoph Dieterich
- Section of Bioinformatics and Systems Cardiology, Klaus Tschira Institute for Integrative Computational Cardiology and Department of Internal Medicine III, University of Heidelberg, Grabengasse 1, 69117 Heidelberg, Germany.
| | - Ivan Merelli
- Instituto di Tecnologie Biomediche - Consiglio Nazionale delle Ricerche, via F.lli Cervi 93, 20090, Segrate, Milano, Italy.
| |
Collapse
|
23
|
Ithayaraja M, Janardan N, Wierenga RK, Savithri HS, Murthy MRN. Crystal structure of a thiolase from Escherichia coli at 1.8 Å resolution. Acta Crystallogr F Struct Biol Commun 2016; 72:534-44. [PMID: 27380370 PMCID: PMC4933003 DOI: 10.1107/s2053230x16008451] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 05/25/2016] [Indexed: 11/10/2022] Open
Abstract
Thiolases catalyze the Claisen condensation of two acetyl-CoA molecules to give acetoacetyl-CoA, as well as the reverse degradative reaction. Four genes coding for thiolases or thiolase-like proteins are found in the Escherichia coli genome. In this communication, the successful cloning, purification, crystallization and structure determination at 1.8 Å resolution of a homotetrameric E. coli thiolase are reported. The structure of E. coli thiolase co-crystallized with acetyl-CoA at 1.9 Å resolution is also reported. As observed in other tetrameric thiolases, the present E. coli thiolase is a dimer of two tight dimers and probably functions as a biodegradative enzyme. Comparison of the structure and biochemical properties of the E. coli enzyme with those of other well studied thiolases reveals certain novel features of this enzyme, such as the modification of a lysine in the dimeric interface, the possible oxidation of the catalytic Cys88 in the structure of the enzyme obtained in the presence of CoA and active-site hydration. The tetrameric enzyme also displays an interesting departure from exact 222 symmetry, which is probably related to the deformation of the tetramerization domain that stabilizes the oligomeric structure of the protein. The current study allows the identification of substrate-binding amino-acid residues and water networks at the active site and provides the structural framework required for understanding the biochemical properties as well as the physiological function of this E. coli thiolase.
Collapse
Affiliation(s)
- M. Ithayaraja
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka 560 012, India
| | - N. Janardan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka 560 012, India
| | - Rik K. Wierenga
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, FIN-90014 Oulu, Finland
| | - H. S. Savithri
- Biochemistry Department, Indian Institute of Science, Bangalore, Karnataka 560 012, India
| | - M. R. N. Murthy
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka 560 012, India
| |
Collapse
|
24
|
Harijan RK, Mazet M, Kiema TR, Bouyssou G, Alexson SEH, Bergmann U, Moreau P, Michels PAM, Bringaud F, Wierenga RK. The SCP2-thiolase-like protein (SLP) of Trypanosoma brucei is an enzyme involved in lipid metabolism. Proteins 2016; 84:1075-96. [PMID: 27093562 DOI: 10.1002/prot.25054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 04/03/2016] [Accepted: 04/08/2016] [Indexed: 11/06/2022]
Abstract
Bioinformatics studies have shown that the genomes of trypanosomatid species each encode one SCP2-thiolase-like protein (SLP), which is characterized by having the YDCF thiolase sequence fingerprint of the Cβ2-Cα2 loop. SLPs are only encoded by the genomes of these parasitic protists and not by those of mammals, including human. Deletion of the Trypanosoma brucei SLP gene (TbSLP) increases the doubling time of procyclic T. brucei and causes a 5-fold reduction of de novo sterol biosynthesis from glucose- and acetate-derived acetyl-CoA. Fluorescence analyses of EGFP-tagged TbSLP expressed in the parasite located the TbSLP in the mitochondrion. The crystal structure of TbSLP (refined at 1.75 Å resolution) confirms that TbSLP has the canonical dimeric thiolase fold. In addition, the structures of the TbSLP-acetoacetyl-CoA (1.90 Å) and TbSLP-malonyl-CoA (2.30 Å) complexes reveal that the two oxyanion holes of the thiolase active site are preserved. TbSLP binds malonyl-CoA tightly (Kd 90 µM), acetoacetyl-CoA moderately (Kd 0.9 mM) and acetyl-CoA and CoA very weakly. TbSLP possesses low malonyl-CoA decarboxylase activity. Altogether, the data show that TbSLP is a mitochondrial enzyme involved in lipid metabolism. Proteins 2016; 84:1075-1096. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Rajesh K Harijan
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, FIN-90014, Finland.,Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, 10461, USA
| | - Muriel Mazet
- Centre De Résonance Magnétique Des Systèmes Biologiques (RMSB), UMR5536, Université De Bordeaux, CNRS, 146 Rue Léo Saignat, Bordeaux, 33076, France.,Laboratoire De Microbiologie Fondamentale Et Pathogénicité (MFP), UMR5234, Université De Bordeaux, CNRS, 146 Rue Léo Saignat, Bordeaux, 33076, France
| | - Tiila R Kiema
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, FIN-90014, Finland
| | - Guillaume Bouyssou
- Laboratoire De Biogenèse Membranaire, UMR-5200, Université De Bordeaux, CNRS, Bâtiment A3 - 1er Étage, INRA Bordeaux Aquitaine BP81, 71 Avenue Edouard Bourlaux, Villenave D'Ornon Cedex, 33883, France
| | - Stefan E H Alexson
- Karolinska Institutet, Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska University Hospital, Stockholm, SE 141 86, Sweden
| | - Ulrich Bergmann
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, FIN-90014, Finland
| | - Patrick Moreau
- Laboratoire De Biogenèse Membranaire, UMR-5200, Université De Bordeaux, CNRS, Bâtiment A3 - 1er Étage, INRA Bordeaux Aquitaine BP81, 71 Avenue Edouard Bourlaux, Villenave D'Ornon Cedex, 33883, France
| | - Paul A M Michels
- Centre for Immunity, Infection and Evolution and Centre for Translational and Chemical Biology, School of Biological Sciences, the King's Buildings, University of Edinburgh, Charlotte Auerbach Road, Edinburgh, EH9 3FL, United Kingdom
| | - Frédéric Bringaud
- Centre De Résonance Magnétique Des Systèmes Biologiques (RMSB), UMR5536, Université De Bordeaux, CNRS, 146 Rue Léo Saignat, Bordeaux, 33076, France.,Laboratoire De Microbiologie Fondamentale Et Pathogénicité (MFP), UMR5234, Université De Bordeaux, CNRS, 146 Rue Léo Saignat, Bordeaux, 33076, France
| | - Rik K Wierenga
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, FIN-90014, Finland
| |
Collapse
|
25
|
Tomilov A, Tomilova N, Shan Y, Hagopian K, Bettaieb A, Kim K, Pelicci PG, Haj F, Ramsey J, Cortopassi G. p46Shc Inhibits Thiolase and Lipid Oxidation in Mitochondria. J Biol Chem 2016; 291:12575-12585. [PMID: 27059956 DOI: 10.1074/jbc.m115.695577] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Indexed: 01/27/2023] Open
Abstract
Although the p46Shc isoform has been known to be mitochondrially localized for 11 years, its function in mitochondria has been a mystery. We confirmed p46Shc to be mitochondrially localized and showed that the major mitochondrial partner of p46Shc is the lipid oxidation enzyme 3-ketoacylCoA thiolase ACAA2, to which p46Shc binds directly and with a strong affinity. Increasing p46Shc expression inhibits, and decreasing p46Shc stimulates enzymatic activity of thiolase in vitro Thus, we suggest p46Shc to be a negative mitochondrial thiolase activity regulator, and reduction of p46Shc expression activates thiolase. This is the first demonstration of a protein that directly binds and controls thiolase activity. Thiolase was thought previously only to be regulated by metabolite balance and steady-state flux control. Thiolase is the last enzyme of the mitochondrial fatty acid beta-oxidation spiral, and thus is important for energy metabolism. Mice with reduction of p46Shc are lean, resist obesity, have higher lipid oxidation capacity, and increased thiolase activity. The thiolase-p46Shc connection shown here in vitro and in organello may be an important underlying mechanism explaining the metabolic phenotype of Shc-depleted mice in vivo.
Collapse
Affiliation(s)
- Alexey Tomilov
- Department of ‡Molecular Biosciences, University of California Davis, California 95616
| | - Natalia Tomilova
- Department of ‡Molecular Biosciences, University of California Davis, California 95616
| | - Yuxi Shan
- Department of ‡Molecular Biosciences, University of California Davis, California 95616
| | - Kevork Hagopian
- Department of ‡Molecular Biosciences, University of California Davis, California 95616; Department of Nutrition, University of California Davis, California 95616 and
| | - Ahmed Bettaieb
- Department of ‡Molecular Biosciences, University of California Davis, California 95616
| | - Kyoungmi Kim
- Department of ‡Molecular Biosciences, University of California Davis, California 95616; Department of Public Health Sciences, University of California Davis, California 95616
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology, IFOM-IEO, Via Adamello, 1620139 Milan, Italy
| | - Fawaz Haj
- Department of Nutrition, University of California Davis, California 95616 and
| | - Jon Ramsey
- Department of ‡Molecular Biosciences, University of California Davis, California 95616; Department of Nutrition, University of California Davis, California 95616 and
| | - Gino Cortopassi
- Department of ‡Molecular Biosciences, University of California Davis, California 95616.
| |
Collapse
|
26
|
Janardan N, Harijan RK, Kiema TR, Wierenga RK, Murthy MRN. Structural characterization of a mitochondrial 3-ketoacyl-CoA (T1)-like thiolase fromMycobacterium smegmatis. ACTA ACUST UNITED AC 2015; 71:2479-93. [DOI: 10.1107/s1399004715019331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 10/12/2015] [Indexed: 11/10/2022]
Abstract
Thiolases catalyze the degradation and synthesis of 3-ketoacyl-CoA molecules. Here, the crystal structures of a T1-like thiolase (MSM-13 thiolase) fromMycobacterium smegmatisin apo and liganded forms are described. Systematic comparisons of six crystallographically independent unliganded MSM-13 thiolase tetramers (dimers of tight dimers) from three different crystal forms revealed that the two tight dimers are connected to a rigid tetramerization domainviaflexible hinge regions, generating an asymmetric tetramer. In the liganded structure, CoA is bound to those subunits that are rotated towards the tip of the tetramerization loop of the opposing dimer, suggesting that this loop is important for substrate binding. The hinge regions responsible for this rotation occur near Val123 and Arg149. The Lα1–covering loop–Lα2 region, together with the Nβ2–Nα2 loop of the adjacent subunit, defines a specificity pocket that is larger and more polar than those of other tetrameric thiolases, suggesting that MSM-13 thiolase has a distinct substrate specificity. Consistent with this finding, only residual activity was detected with acetoacetyl-CoA as the substrate in the degradative direction. No activity was observed with acetyl-CoA in the synthetic direction. Structural comparisons with other well characterized thiolases suggest that MSM-13 thiolase is probably a degradative thiolase that is specific for 3-ketoacyl-CoA molecules with polar, bulky acyl chains.
Collapse
|