1
|
Guo L, Du Y, Li H, He T, Yao L, Yang G, Yang X. Metabolites-mediated posttranslational modifications in cardiac metabolic remodeling: Implications for disease pathology and therapeutic potential. Metabolism 2025; 165:156144. [PMID: 39864796 DOI: 10.1016/j.metabol.2025.156144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
The nonenergy - producing functions of metabolism are attracting increasing attention, as metabolic changes are involved in discrete pathways modulating enzyme activity and gene expression. Substantial evidence suggests that myocardial metabolic remodeling occurring during diabetic cardiomyopathy, heart failure, and cardiac pathological stress (e.g., myocardial ischemia, pressure overload) contributes to the progression of pathology. Within the rewired metabolic network, metabolic intermediates and end-products can directly alter protein function and/or regulate epigenetic modifications by providing acyl groups for posttranslational modifications, thereby affecting the overall cardiac stress response and providing a direct link between cellular metabolism and cardiac pathology. This review provides a comprehensive overview of the functional diversity and mechanistic roles of several types of metabolite-mediated histone and nonhistone acylation, namely O-GlcNAcylation, lactylation, crotonylation, β-hydroxybutyrylation, and succinylation, as well as fatty acid-mediated modifications, in regulating physiological processes and contributing to the progression of heart disease. Furthermore, it explores the potential of these modifications as therapeutic targets for disease intervention.
Collapse
Affiliation(s)
- Lifei Guo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China; The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China; Cadet Team 6 of School of Basic Medicine, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China
| | - Yuting Du
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China; The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China
| | - Heng Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China
| | - Ting He
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China
| | - Li Yao
- Department of Pathology, Xi' an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi' an 710018, China
| | - Guodong Yang
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China.
| | - Xuekang Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China.
| |
Collapse
|
2
|
Sharma S, McKenzie M. The Pathogenesis of Very Long-Chain Acyl-CoA Dehydrogenase Deficiency. Biomolecules 2025; 15:416. [PMID: 40149952 PMCID: PMC11940467 DOI: 10.3390/biom15030416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Living systems require energy to maintain their existence and perform tasks such as cell division. This energy is stored in several molecular forms in nature, specifically lipids, carbohydrates, and amino acids. At a cellular level, energy is extracted from these complex molecules and transferred to adenosine triphosphate (ATP) in the cytoplasm and mitochondria. Within the mitochondria, fatty acid β-oxidation (FAO) and oxidative phosphorylation (OXPHOS) are crucial metabolic processes involved in generating ATP, with defects in these pathways causing mitochondrial disease. Very long-chain acyl-CoA dehydrogenase deficiency (VLCADD) is a fatty acid β-oxidation disorder (FAOD) affecting 1 to 2 individuals per 100,000. Similar to other mitochondrial disorders, there is no cure for VLCADD, with symptomatic treatment comprising dietary management and supplementation with medium-chain fatty acids to bypass the enzyme deficiency. While this addresses the primary defect in VLCADD, there is growing evidence that other aspects of mitochondrial function are also affected in VLCADD, including secondary defects in OXPHOS function. Here, we review our current understanding of VLCADD with a focus on the associated biochemical and molecular defects that can disrupt multiple aspects of mitochondrial function. We describe the interactions between FAO proteins and the OXPHOS complexes and how these interactions are critical for maintaining the activity of both metabolic pathways. In particular, we describe what is now known about the protein-protein interactions between VLCAD and the OXPHOS supercomplex and how their disruption contributes to overall VLCADD pathogenesis.
Collapse
Affiliation(s)
- Shashwat Sharma
- School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Waurn Ponds, VIC 3216, Australia;
| | - Matthew McKenzie
- School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Waurn Ponds, VIC 3216, Australia;
- Institute for Physical Activity and Nutrition, Deakin University, 75 Pigdons Road, Waurn Ponds, VIC 3216, Australia
| |
Collapse
|
3
|
Wang Y, Hu M, Cao J, Wang F, Han JR, Wu TW, Li L, Yu J, Fan Y, Xie G, Lian H, Cao Y, Naowarojna N, Wang X, Zou Y. ACSL4 and polyunsaturated lipids support metastatic extravasation and colonization. Cell 2025; 188:412-429.e27. [PMID: 39591965 DOI: 10.1016/j.cell.2024.10.047] [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: 09/26/2023] [Revised: 04/13/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024]
Abstract
Metastatic dissemination to distant organs demands that cancer cells possess high morphological and metabolic adaptability. However, contributions of the cellular lipidome to metastasis remain elusive. Here, we uncover a correlation between metastasis potential and ferroptosis susceptibility in multiple cancers. Metastases-derived cancer cells exhibited higher ferroptosis sensitivity and polyunsaturated fatty acyl (PUFA)-lipid contents than primary-tumor-derived cells from ovarian cancer patients. Metabolism-focused CRISPR screens in a mouse model for ovarian cancer distant metastasis established via two rounds of in vivo selection revealed the PUFA-lipid biosynthesis enzyme acyl-coenzyme A (CoA) synthetase long-chain family member 4 (ACSL4) as a pro-hematogenous metastasis factor. ACSL4 promotes metastatic extravasation by enhancing membrane fluidity and cellular invasiveness. While promoting metastasis, the high PUFA-lipid state creates dependencies on abhydrolase-domain-containing 6, acylglycerol lipase (ABHD6), enoyl-CoA delta isomerase 1 (ECI1), and enoyl-CoA hydratase 1 (ECH1)-rate-limiting enzymes preparing unsaturated fatty acids (UFAs) for β-oxidation. ACSL4/ECH1 co-inhibition achieved potent suppression of metastasis. Our work establishes the dual functions of PUFA-lipids in tumor progression and metastasis that may be exploitable for therapeutic development.
Collapse
Affiliation(s)
- Yuqi Wang
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Mangze Hu
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Jian Cao
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China
| | - Fengxiang Wang
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingrong Regina Han
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; School of Life Sciences, Fudan University, Shanghai, China
| | - Tianshu William Wu
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Luxiao Li
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Jinshi Yu
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Yujing Fan
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Guanglei Xie
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Bioinformatics and Genomics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Heyuan Lian
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Yueying Cao
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China
| | - Nathchar Naowarojna
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Xi Wang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Westlake Bioinformatics and Genomics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| | - Yilong Zou
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
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
|
5
|
Eaton S. I Walk the Line: Between Basic Science and Paediatric Surgery. J Pediatr Surg 2024; 59:172-176. [PMID: 37940464 DOI: 10.1016/j.jpedsurg.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023]
Abstract
The role of a basic scientist working with paediatric surgeons is not an obvious one. However, there are several levels at which science can contribute to the speciality, and also ways that scientists can learn useful lessons from paediatric surgery. As most conditions treated by paediatric surgeons have low case numbers, we need to find ways of defining optimal treatment and developing novel therapies within a challenging number of patients.
Collapse
Affiliation(s)
- Simon Eaton
- UCL Great Ormond Street Institute of Child Health, London, UK.
| |
Collapse
|
6
|
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
|
7
|
Singh R, Kundu P, Mishra VK, Singh BK, Bhattacharyya S, Das AK. Crystal structure of FadA2 thiolase from Mycobacterium tuberculosis and prediction of its substrate specificity and membrane-anchoring properties. FEBS J 2023; 290:3997-4022. [PMID: 37026388 DOI: 10.1111/febs.16792] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/17/2023] [Accepted: 04/05/2023] [Indexed: 04/08/2023]
Abstract
Tuberculosis (TB) is one of the leading causes of human death caused by Mycobacterium tuberculosis (Mtb). Mtb can enter into a long-lasting persistence where it can utilize fatty acids as the carbon source. Hence, fatty acid metabolism pathway enzymes are considered promising and pertinent mycobacterial drug targets. FadA2 (thiolase) is one of the enzymes involved in Mtb's fatty acid metabolism pathway. FadA2 deletion construct (ΔL136-S150) was designed to produce soluble protein. The crystal structure of FadA2 (ΔL136-S150) at 2.9 Å resolution was solved and analysed for membrane-anchoring region. The four catalytic residues of FadA2 are Cys99, His341, His390 and Cys427, and they belong to four loops with characteristic sequence motifs, i.e., CxT, HEAF, GHP and CxA. FadA2 is the only thiolase of Mtb which belongs to the CHH category containing the HEAF motif. Analysing the substrate-binding channel, it has been suggested that FadA2 is involved in the β-oxidation pathway, i.e., the degradative pathway, as the long-chain fatty acid can be accommodated in the channel. The catalysed reaction is favoured by the presence of two oxyanion holes, i.e., OAH1 and OAH2. OAH1 formation is unique in FadA2, formed by the NE2 of His390 present in the GHP motif and NE2 of His341 present in the HEAF motif, whereas OAH2 formation is similar to CNH category thiolase. Sequence and structural comparison with the human trifunctional enzyme (HsTFE-β) suggests the membrane-anchoring region in FadA2. Molecular dynamics simulations of FadA2 with a membrane containing POPE lipid were conducted to understand the role of a long insertion sequence of FadA2 in membrane anchoring.
Collapse
Affiliation(s)
- Rashika Singh
- Department of Biotechnology, Indian Institute of Technology Kharagpur, India
| | - Prasun Kundu
- Department of Biotechnology, Indian Institute of Technology Kharagpur, India
| | | | - Bina Kumari Singh
- School of Bioscience, Indian Institute of Technology Kharagpur, India
| | - Sudipta Bhattacharyya
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, India
| | - Amit Kumar Das
- Department of Biotechnology, Indian Institute of Technology Kharagpur, India
| |
Collapse
|
8
|
Du J, Sudlow LC, Shahverdi K, Zhou H, Michie M, Schindler TH, Mitchell JD, Mollah S, Berezin MY. Oxaliplatin-induced cardiotoxicity in mice is connected to the changes in energy metabolism in the heart tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.542198. [PMID: 37292714 PMCID: PMC10245950 DOI: 10.1101/2023.05.24.542198] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Oxaliplatin is a platinum-based alkylating chemotherapeutic agent used for cancer treatment. At high cumulative dosage, the negative effect of oxaliplatin on the heart becomes evident and is linked to a growing number of clinical reports. The aim of this study was to determine how chronic oxaliplatin treatment causes the changes in energy-related metabolic activity in the heart that leads to cardiotoxicity and heart damage in mice. C57BL/6 male mice were treated with a human equivalent dosage of intraperitoneal oxaliplatin (0 and 10 mg/kg) once a week for eight weeks. During the treatment, mice were followed for physiological parameters, ECG, histology and RNA sequencing of the heart. We identified that oxaliplatin induces strong changes in the heart and affects the heart's energy-related metabolic profile. Histological post-mortem evaluation identified focal myocardial necrosis infiltrated with a small number of associated neutrophils. Accumulated doses of oxaliplatin led to significant changes in gene expression related to energy related metabolic pathways including fatty acid (FA) oxidation, amino acid metabolism, glycolysis, electron transport chain, and NAD synthesis pathway. At high accumulative doses of oxaliplatin, the heart shifts its metabolism from FAs to glycolysis and increases lactate production. It also leads to strong overexpression of genes in NAD synthesis pathways such as Nmrk2. Changes in gene expression associated with energy metabolic pathways can be used to develop diagnostic methods to detect oxaliplatin-induced cardiotoxicity early on as well as therapy to compensate for the energy deficit in the heart to prevent heart damage.
Collapse
Affiliation(s)
- Junwei Du
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
- Institute of Materials Science & Engineering Washington University, St. Louis, MO 63130, USA
| | - Leland C Sudlow
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Kiana Shahverdi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Haiying Zhou
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Megan Michie
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Thomas H Schindler
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
| | - Joshua D Mitchell
- Cardio-Oncology Center of Excellence, Washington University School of Medicine, St. Louis, MO 63110
| | - Shamim Mollah
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
| | - Mikhail Y Berezin
- Mallinckrodt Institute of Radiology, Washington University School of Medicine St. Louis, MO 63110, USA
- Institute of Materials Science & Engineering Washington University, St. Louis, MO 63130, USA
| |
Collapse
|
9
|
Kablau A, Erler S, Eckert JH, Pistorius J, Sharbati S, Einspanier R. Effects of Flupyradifurone and Two Reference Insecticides Commonly Used in Toxicological Studies on the Larval Proteome of the Honey bee Apis mellifera. INSECTS 2023; 14:77. [PMID: 36662005 PMCID: PMC9862931 DOI: 10.3390/insects14010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/21/2022] [Accepted: 01/11/2023] [Indexed: 06/17/2023]
Abstract
The western honey bee Apis mellifera is globally distributed due to its beekeeping advantages and plays an important role in the global ecology and economy. In recent decades, several studies have raised concerns about bee decline. Discussed are multiple reasons such as increased pathogen pressure, malnutrition or pesticide use. Insecticides are considered to be one of the major factors. In 2013, the use of three neonicotinoids in the field was prohibited in the EU. Flupyradifurone was introduced as a potential successor; it has a comparable mode of action as the banned neonicotinoids. However, there is a limited number of studies on the effects of sublethal concentrations of flupyradifurone on honey bees. Particularly, the larval physiological response by means of protein expression has not yet been studied. Hence, the larval protein expression was investigated via 2D gel electrophoresis after following a standardised protocol to apply sublethal concentrations of the active substance (flupyradifurone 10 mg/kg diet) to larval food. The treated larvae did not show increased mortality or an aberrant development. Proteome comparisons showed clear differences concerning the larval metabolism, immune response and energy supply. Further field studies are needed to validate the in vitro results at a colony level.
Collapse
Affiliation(s)
- Arne Kablau
- Institute of Veterinary Biochemistry, Freie Universität Berlin, 14163 Berlin, Germany
- LABOKLIN GmbH and Co. KG, 97688 Bad Kissingen, Germany
| | - Silvio Erler
- Institute for Bee Protection, Julius Kühn Institute (JKI)—Federal Research Centre for Cultivated Plants, 38104 Braunschweig, Germany
- Zoological Institute, Technische Universität Braunschweig, 38106 Brauschweig, Germany
| | - Jakob H. Eckert
- Institute for Bee Protection, Julius Kühn Institute (JKI)—Federal Research Centre for Cultivated Plants, 38104 Braunschweig, Germany
- Institute for Microbiology, Technische Universität Braunschweig, 38106 Brauschweig, Germany
| | - Jens Pistorius
- Institute for Bee Protection, Julius Kühn Institute (JKI)—Federal Research Centre for Cultivated Plants, 38104 Braunschweig, Germany
| | - Soroush Sharbati
- Institute of Veterinary Biochemistry, Freie Universität Berlin, 14163 Berlin, Germany
| | - Ralf Einspanier
- Institute of Veterinary Biochemistry, Freie Universität Berlin, 14163 Berlin, Germany
| |
Collapse
|
10
|
Stolwijk NN, Langeveld M, Jacobs BAW, Vogt L, Haverkamp JA, Ferdinandusse S, Hollak CEM. Recurrent metabolic alkalosis following ketone body treatment of adult mitochondrial trifunctional protein deficiency: A case report. JIMD Rep 2022; 63:407-413. [PMID: 36101817 PMCID: PMC9458612 DOI: 10.1002/jmd2.12309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/19/2022] [Accepted: 06/07/2022] [Indexed: 12/05/2022] Open
Abstract
Recent studies have reported the potential for the therapeutic use of ketones in the form of ketone salts (KSs) in pediatric patients with fatty acid oxidation disorders (FAODs). We report a case of ketone salt administration in an adult patient with mitochondrial trifunctional protein deficiency (MTPD), an ultra-rare inborn error of the fatty acid metabolism. This patient was treated with oral KSs during an episode of sepsis of unknown origin. Before KS supplementation was initiated, he had developed severe rhabdomyolysis as well as a respiratory insufficiency that did not respond to emergency treatment aimed at stabilizing the metabolic decompensation by promoting anabolism. Therefore, KS supplementation was attempted twice to support his energy production and help regain metabolic stability. In both instances, KS supplementation led to a considerable metabolic alkalosis, which prompted its discontinuation. This adverse event could have been caused by an increase in extracellular sodium load due to KS administration. Therefore, the clinical applicability of KSs in adults may be limited. Alternative chemical forms of beta-hydroxybutyrate (βHB), such as ketone esters, might provide a more acceptable safety profile for future research into the therapeutic benefits of ketone body supplementation in adult patients with FAODs.
Collapse
Affiliation(s)
- Nina N. Stolwijk
- Medicine for SocietyAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Department of Endocrinology and MetabolismAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
| | - Mirjam Langeveld
- Department of Endocrinology and MetabolismAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
| | - Bart A. W. Jacobs
- Medicine for SocietyAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Department of Pharmacy and Clinical PharmacologyAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
| | - Liffert Vogt
- Division of Nephrology, Department of Internal MedicineAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
| | - Jorien A. Haverkamp
- Department of Endocrinology and MetabolismAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Department of Clinical ChemistryAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
| | - Carla E. M. Hollak
- Medicine for SocietyAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Department of Endocrinology and MetabolismAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
11
|
Schwantje M, Fuchs SA, de Boer L, Bosch AM, Cuppen I, Dekkers E, Derks TGJ, Ferdinandusse S, Ijlst L, Houtkooper RH, Maase R, van der Pol WL, de Vries MC, Verschoof‐Puite RK, Wanders RJA, Williams M, Wijburg F, Visser G. Genetic, biochemical, and clinical spectrum of patients with mitochondrial trifunctional protein deficiency identified after the introduction of newborn screening in the Netherlands. J Inherit Metab Dis 2022; 45:804-818. [PMID: 35383965 PMCID: PMC9546250 DOI: 10.1002/jimd.12502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/23/2022] [Accepted: 04/04/2022] [Indexed: 11/24/2022]
Abstract
Long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD) is included in many newborn screening (NBS) programs. Acylcarnitine-based NBS for LCHADD not only identifies LCHADD, but also the other deficiencies of the mitochondrial trifunctional protein (MTP), a multi-enzyme complex involved in long-chain fatty acid β-oxidation. Besides LCHAD, MTP harbors two additional enzyme activities: long-chain enoyl-CoA hydratase (LCEH) and long-chain ketoacyl-CoA thiolase (LCKAT). Deficiency of one or more MTP activities causes generalized MTP deficiency (MTPD), LCHADD, LCEH deficiency (not yet reported), or LCKAT deficiency (LCKATD). To gain insight in the outcomes of MTP-deficient patients diagnosed after the introduction of NBS for LCHADD in the Netherlands, a retrospective evaluation of genetic, biochemical, and clinical characteristics of MTP-deficient patients, identified since 2007, was carried out. Thirteen patients were identified: seven with LCHADD, five with MTPD, and one with LCKATD. All LCHADD patients (one missed by NBS, clinical diagnosis) and one MTPD patient (clinical diagnosis) were alive. Four MTPD patients and one LCKATD patient developed cardiomyopathy and died within 1 month and 13 months of life, respectively. Surviving patients did not develop symptomatic hypoglycemia, but experienced reversible cardiomyopathy and rhabdomyolysis. Five LCHADD patients developed subclinical neuropathy and/or retinopathy. In conclusion, patient outcomes were highly variable, stressing the need for accurate classification of and discrimination between the MTP deficiencies to improve insight in the yield of NBS for LCHADD. NBS allowed the prevention of symptomatic hypoglycemia, but current treatment options failed to treat cardiomyopathy and prevent long-term complications. Moreover, milder patients, who might benefit from NBS, were missed due to normal acylcarnitine profiles.
Collapse
Affiliation(s)
- Marit Schwantje
- Department of Metabolic DiseasesWilhelmina Children's Hospital, University Medical Center UtrechtUtrechtThe Netherlands
- Laboratory Genetic Metabolic Diseases, and Metabolism Institute, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Sabine A. Fuchs
- Department of Metabolic DiseasesWilhelmina Children's Hospital, University Medical Center UtrechtUtrechtThe Netherlands
| | - Lonneke de Boer
- Department of Metabolic Diseases, Amalia Children's HospitalRadboud University Medical CentreNijmegenThe Netherlands
| | - Annet M. Bosch
- Department of Metabolic Diseases, Emma Children's Hospital, and Metabolism Institute, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Inge Cuppen
- Department of Neurology and NeurosurgeryWilhelmina Children's Hospital, University Medical Center UtrechtUtrechtThe Netherlands
| | - Eugenie Dekkers
- National Institute for Public Health and the Environment (RIVM) Reference Laboratory for Pre‐ and Neonatal Screening, Center for Health Protection (R.M.) and Center for Population Screening (E.D)BilthovenThe Netherlands
| | - Terry G. J. Derks
- Department of Metabolic DiseasesBeatrix Children's Hospital, University Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, and Metabolism Institute, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Lodewijk Ijlst
- Laboratory Genetic Metabolic Diseases, and Metabolism Institute, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Riekelt H. Houtkooper
- Laboratory Genetic Metabolic Diseases, and Metabolism Institute, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Rose Maase
- National Institute for Public Health and the Environment (RIVM) Reference Laboratory for Pre‐ and Neonatal Screening, Center for Health Protection (R.M.) and Center for Population Screening (E.D)BilthovenThe Netherlands
| | - W. Ludo van der Pol
- Department of Neurology and NeurosurgeryWilhelmina Children's Hospital, University Medical Center UtrechtUtrechtThe Netherlands
| | - Maaike C. de Vries
- Department of Metabolic Diseases, Amalia Children's HospitalRadboud University Medical CentreNijmegenThe Netherlands
| | - Rendelien K. Verschoof‐Puite
- Department for Vaccine Supply and Prevention ProgramsNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands
| | - Ronald J. A. Wanders
- Laboratory Genetic Metabolic Diseases, and Metabolism Institute, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Monique Williams
- Department of PediatricsCenter for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center RotterdamRotterdamNetherlands
| | - Frits Wijburg
- Department of Metabolic Diseases, Emma Children's Hospital, and Metabolism Institute, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Gepke Visser
- Department of Metabolic DiseasesWilhelmina Children's Hospital, University Medical Center UtrechtUtrechtThe Netherlands
- Laboratory Genetic Metabolic Diseases, and Metabolism Institute, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
12
|
Schwantje M, Ebberink MS, Doolaard M, Ruiter JPN, Fuchs SA, Darin N, Hedberg‐Oldfors C, Régal L, Donker Kaat L, Huidekoper HH, Olpin S, Cole D, Moat SJ, Visser G, Ferdinandusse S. Thermo-sensitive mitochondrial trifunctional protein deficiency presenting with episodic myopathy. J Inherit Metab Dis 2022; 45:819-831. [PMID: 35403730 PMCID: PMC9542805 DOI: 10.1002/jimd.12503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/24/2022] [Accepted: 04/07/2022] [Indexed: 11/29/2022]
Abstract
Mitochondrial trifunctional protein (MTP) is involved in long-chain fatty acid β-oxidation (lcFAO). Deficiency of one or more of the enzyme activities as catalyzed by MTP causes generalized MTP deficiency (MTPD), long-chain hydroxyacyl-CoA dehydrogenase deficiency (LCHADD), or long-chain ketoacyl-CoA thiolase deficiency (LCKATD). When genetic variants result in thermo-sensitive enzymes, increased body temperature (e.g. fever) can reduce enzyme activity and be a risk factor for clinical decompensation. This is the first description of five patients with a thermo-sensitive MTP deficiency. Clinical and genetic information was obtained from clinical files. Measurement of LCHAD and LCKAT activities, lcFAO-flux studies and palmitate loading tests were performed in skin fibroblasts cultured at 37°C and 40°C. In all patients (four MTPD, one LCKATD), disease manifested during childhood (manifestation age: 2-10 years) with myopathic symptoms triggered by fever or exercise. In four patients, signs of retinopathy or neuropathy were present. Plasma long-chain acylcarnitines were normal or slightly increased. HADHB variants were identified (at age: 6-18 years) by whole exome sequencing or gene panel analyses. At 37°C, LCHAD and LCKAT activities were mildly impaired and lcFAO-fluxes were normal. Remarkably, enzyme activities and lcFAO-fluxes were markedly diminished at 40°C. Preventive (dietary) measures improved symptoms for most. In conclusion, all patients with thermo-sensitive MTP deficiency had a long diagnostic trajectory and both genetic and enzymatic testing were required for diagnosis. The frequent absence of characteristic acylcarnitine abnormalities poses a risk for a diagnostic delay. Given the positive treatment effects, upfront genetic screening may be beneficial to enhance early recognition.
Collapse
Affiliation(s)
- Marit Schwantje
- Department of Metabolic DiseasesWilhelmina Children's Hospital, University Medical Center UtrechtUtrechtThe Netherlands
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Merel S. Ebberink
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Mirjam Doolaard
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Jos P. N. Ruiter
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Sabine A. Fuchs
- Department of Metabolic DiseasesWilhelmina Children's Hospital, University Medical Center UtrechtUtrechtThe Netherlands
| | - Niklas Darin
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska AcademyUniversity of Gothenburg, Sahlgrenska University HospitalGothenburgSweden
| | - Carola Hedberg‐Oldfors
- Department of Laboratory Medicine, Institute of BiomedicineUniversity of GothenburgGothenburgSweden
| | - Luc Régal
- Pediatric Neurology and Metabolism Department of PediatricsUZ BrusselJetteBelgium
| | - Laura Donker Kaat
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Hidde H. Huidekoper
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Simon Olpin
- Department of Clinical ChemistrySheffield Children's HospitalSheffieldUK
| | - Duncan Cole
- Wales Newborn Screening Laboratory, Department of Medical Biochemistry, Immunology and ToxicologyUniversity Hospital of WalesCardiffUK
- School of MedicineCardiff UniversityCardiffUK
| | - Stuart J. Moat
- Department of Clinical ChemistrySheffield Children's HospitalSheffieldUK
- Wales Newborn Screening Laboratory, Department of Medical Biochemistry, Immunology and ToxicologyUniversity Hospital of WalesCardiffUK
| | - Gepke Visser
- Department of Metabolic DiseasesWilhelmina Children's Hospital, University Medical Center UtrechtUtrechtThe Netherlands
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
13
|
Avilkina V, Chauveau C, Ghali Mhenni O. Sirtuin function and metabolism: Role in pancreas, liver, and adipose tissue and their crosstalk impacting bone homeostasis. Bone 2022; 154:116232. [PMID: 34678494 DOI: 10.1016/j.bone.2021.116232] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022]
Abstract
Mammalian sirtuins (SIRT1-7) are members of the nicotine adenine dinucleotide (NAD+)-dependent family of enzymes critical for histone deacetylation and posttranslational modification of proteins. Sirtuin family members regulate a wide spectrum of biological processes and are best known for maintaining longevity. Sirtuins are well characterized in metabolic tissues such as the pancreas, liver and adipose tissue (AT). They are regulated by a diverse range of stimuli, including nutrients and metabolic changes within the organism. Indeed, nutrient-associated conditions, such as obesity and anorexia nervosa (AN), were found to be associated with bone fragility development in osteoporosis. Interestingly, it has also been demonstrated that sirtuins, more specifically SIRT1, can regulate bone activity. Various studies have demonstrated the importance of sirtuins in bone in the regulation of bone homeostasis and maintenance of the balance between bone resorption and bone formation. However, to understand the molecular mechanisms involved in the negative regulation of bone homeostasis during overnutrition (obesity) or undernutrition, it is crucial to examine a wider picture and to determine the pancreatic, liver and adipose tissue pathway crosstalk responsible for bone loss. Particularly, under AN conditions, sirtuin family members are highly expressed in metabolic tissue, but this phenomenon is reversed in bone, and severe bone loss has been observed in human subjects. AN-associated bone loss may be connected to SIRT1 deficiency; however, additional factors may interfere with bone homeostasis. Thus, in this review, we focus on sirtuin activity in the pancreas, liver and AT in cases of over- and undernutrition, especially the regulation of their secretome by sirtuins. Furthermore, we examine how the secretome of the pancreas, liver and AT affects bone homeostasis, focusing on undernutrition. This review aims to lead to a better understanding of the crosstalk between sirtuins, metabolic organs and bone. In long term prospective it should contribute to promote improvement of therapeutic strategies for the prevention of metabolic diseases and the development of osteoporosis.
Collapse
Affiliation(s)
- Viktorija Avilkina
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Univ. Littoral Côte d'Opale, F-62200, Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Christophe Chauveau
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Univ. Littoral Côte d'Opale, F-62200, Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Olfa Ghali Mhenni
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Univ. Littoral Côte d'Opale, F-62200, Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France.
| |
Collapse
|
14
|
Yang J, Yuan D, Tan X, Zeng Y, Tang N, Chen D, Tan J, Cai R, Huang J, Yan T. Analysis of a family with mitochondrial trifunctional protein deficiency caused by HADHA gene mutations. Mol Med Rep 2021; 25:47. [PMID: 34878152 PMCID: PMC8674702 DOI: 10.3892/mmr.2021.12563] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/22/2021] [Indexed: 11/21/2022] Open
Abstract
Mitochondrial trifunctional protein (MTP) deficiency (MTPD; MIM 609015) is a metabolic disease of fatty acid oxidation. MTPD is an autosomal recessive disorder caused by mutations in the HADHA gene, encoding the α-subunit of a trifunctional protease, or in the HADHB gene, encoding the β-subunit of a trifunctional protease. To the best of our knowledge, only two cases of families with MTPD due to HADHB gene mutations have been reported in China, and the HADHA gene mutation has not been reported in a Chinese family with MTPD. The present study reported the clinical characteristics and compound heterozygous HADHA gene mutations of two patients with MTPD in the Chinese population. The medical history, routine examination data, blood acyl-carnitine analysis results, results of pathological examination after autopsy and family pedigree map were collected for patients with MTPD. The HADHA gene was analyzed by Sanger sequencing or high-throughput sequencing, the pathogenicity of the newly discovered variant was interpreted by bioinformatics analysis, and the function of the mutated protein was modeled and analyzed according to 3D structure. The two patients with MTPD experienced metabolic crises and died following an infectious disease. Lactate dehydrogenase, creatine kinase (CK), CK-MB and liver enzyme abnormalities were observed in routine examinations. Tandem mass spectrometry revealed that long-chain acyl-carnitine was markedly elevated in blood samples from the patients with MTPD. The autopsy results for one child revealed fat accumulation in the liver and heart. Next-generation sequencing detected compound heterozygous c.703C>T (p.R235W) and c.2107G>A (p.G703R) mutations in the HADHA gene. The mother did not have acute fatty liver during pregnancy with the two patients. Using amniotic fluid prenatal diagnostic testing, the unborn child was confirmed to carry only c.2107G>A (p.G703R). Molecular mechanistic analysis indicated that the two variants affected the conformation of the α-subunit of the MTP enzyme complex, and consequently affected the stability and function of the enzyme complex. The present study comprehensively analyzed the cases, including exome sequencing and protein structure analysis and, to the best of our knowledge, describes the first observation of compound heterozygous mutations in the HADHA gene underlying this disorder in China. The clinical phenotypes of the two heterozygous variants of the HADHA gene are non-lethal. The present study may improve understanding of the HADHA gene mutation spectrum and clinical phenotype in the Chinese population.
Collapse
Affiliation(s)
- Jinling Yang
- Newborn Screening Center, Department of Medical Genetics, Key Laboratory of Prevention and Control of Birth Defects, Liuzhou Maternity and Child Health Care Hospital, Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi 545000, P.R. China
| | - Dejian Yuan
- Newborn Screening Center, Department of Medical Genetics, Key Laboratory of Prevention and Control of Birth Defects, Liuzhou Maternity and Child Health Care Hospital, Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi 545000, P.R. China
| | - Xiaohui Tan
- School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yexi Zeng
- Newborn Screening Center, Huizhou Second Maternity and Child Health Care Hospital, Huizhou, Guangdong 516001, P.R. China
| | - Ning Tang
- Newborn Screening Center, Department of Medical Genetics, Key Laboratory of Prevention and Control of Birth Defects, Liuzhou Maternity and Child Health Care Hospital, Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi 545000, P.R. China
| | - Dayu Chen
- Newborn Screening Center, Department of Medical Genetics, Key Laboratory of Prevention and Control of Birth Defects, Liuzhou Maternity and Child Health Care Hospital, Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi 545000, P.R. China
| | - Jianqiang Tan
- Newborn Screening Center, Department of Medical Genetics, Key Laboratory of Prevention and Control of Birth Defects, Liuzhou Maternity and Child Health Care Hospital, Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi 545000, P.R. China
| | - Ren Cai
- Newborn Screening Center, Department of Medical Genetics, Key Laboratory of Prevention and Control of Birth Defects, Liuzhou Maternity and Child Health Care Hospital, Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi 545000, P.R. China
| | - Jun Huang
- Newborn Screening Center, Department of Medical Genetics, Key Laboratory of Prevention and Control of Birth Defects, Liuzhou Maternity and Child Health Care Hospital, Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi 545000, P.R. China
| | - Tizhen Yan
- Newborn Screening Center, Department of Medical Genetics, Key Laboratory of Prevention and Control of Birth Defects, Liuzhou Maternity and Child Health Care Hospital, Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi 545000, P.R. China
| |
Collapse
|
15
|
Guan Y, Zhang Y, Shen XM, Zhou L, Shang X, Peng Y, Hu Y, Li W. Charcot-Marie-Tooth Disease With Episodic Rhabdomyolysis Due to Two Novel Mutations in the β Subunit of Mitochondrial Trifunctional Protein and Effective Response to Modified Diet Therapy. Front Neurol 2021; 12:694966. [PMID: 34712195 PMCID: PMC8546186 DOI: 10.3389/fneur.2021.694966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/30/2021] [Indexed: 11/29/2022] Open
Abstract
A 29-year-old female experienced chronic progressive peripheral neuropathy since childhood and was diagnosed with Charcot–Marie–Tooth disease (CMT) at age 15. She developed recurrent, fever-induced rhabdomyolysis (RM) at age 24. EMG studies showed decreased amplitude of compound muscle action potential, declined motor conductive velocity, and absence of sensor nerve action potential. Acylcarnitine analysis revealed elevated C16-OH, C18-OH, and C18:1-OH. Muscle biopsy showed scattered foci of necrotic myofibers invaded by macrophages, occasional regenerating fibers, and remarkable muscle fiber type grouping. Whole-exome sequencing identified two novel heterozygous mutations: c.490G>A (p.G164S) and c.686G>A (p.R229Q) in HADHB gene encoding the β-subunit of mitochondrial trifunctional protein (MTP). Reduction of long-chain fatty acid via dietary restrictions alleviated symptoms effectively. Our study indicates that the defect of the MTP β-subunit accounts for both CMT and RM in the same patient and expands the clinical spectrum of disorders caused by the HADHB mutations. Our systematic review of all MTPD patients with dietary treatment indicates that the effect of dietary treatment is related to the age of onset and the severity of symptoms.
Collapse
Affiliation(s)
- Yuqing Guan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanxia Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xin-Ming Shen
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Liang Zhou
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuan Shang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yu Peng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yafang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Li
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
16
|
Sekine Y, Yamamoto K, Kurata M, Honda A, Onishi I, Kinowaki Y, Kawade G, Watabe S, Nomura S, Fukuda S, Ishibashi S, Ikeda M, Yamamoto M, Kitagawa M. HADHB, a fatty acid beta-oxidation enzyme, is a potential prognostic predictor in malignant lymphoma. Pathology 2021; 54:286-293. [PMID: 34531036 DOI: 10.1016/j.pathol.2021.06.119] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 06/05/2021] [Accepted: 06/10/2021] [Indexed: 02/08/2023]
Abstract
In haematological malignancies, such as malignant lymphoma, reprogramming of fatty acid metabolism favours tumour cell survival and drug resistance. Hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit alpha (HADHA), an enzyme involved in fatty acid beta-oxidation (FAO), is overexpressed in high-grade lymphoma and is a predictor of poor prognosis in diffuse large B-cell lymphoma (DLBCL). HADHB forms a heterodimer with HADHA and functions as an FAO enzyme together with HADHA; however, the relevance of its expression in malignant lymphoma is unknown. In this study, we investigated the roles and antitumour effects of HADHB expression in malignant lymphoma. Immunohistochemical analysis showed that HADHB was frequently overexpressed in the high-grade lymphoma subtype. HADHB overexpression was observed in 68% (87/128) of DLBCL cases and was an independent predictor of poor prognosis (p=0.001). In vitro analysis demonstrated that HADHB knockdown suppressed cell proliferation in LCL-K and MD901 cells (p<0.05). Additionally, treatment with the FAO inhibitor, ranolazine, increased cell death in control cells compared with that in HADHB knockdown LCL-K and MD901 cells (p<0.01). Cell death was also suppressed by the ferroptosis inhibitor, ferrosatin-1, in LCL-K and MD901 cells (p<0.05). Collectively, these findings provide basic evidence for the development of new cell death-based therapies for refractory malignant lymphoma. We plan to perform prospective studies and preclinical studies using animal models to confirm these results.
Collapse
Affiliation(s)
- Yuji Sekine
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Department of Cardiovascular Surgery, Nara Prefecture General Medical Centre, Nara, Japan
| | - Kouhei Yamamoto
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Morito Kurata
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ayaka Honda
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Iichiroh Onishi
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuko Kinowaki
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Genji Kawade
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shiori Watabe
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Serina Nomura
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sho Fukuda
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sachiko Ishibashi
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masumi Ikeda
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masahide Yamamoto
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masanobu Kitagawa
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
17
|
Pemafibrate Protects against Fatty Acid-Induced Nephropathy by Maintaining Renal Fatty Acid Metabolism. Metabolites 2021; 11:metabo11060372. [PMID: 34207854 PMCID: PMC8230306 DOI: 10.3390/metabo11060372] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/25/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
As classical agonists for peroxisomal proliferator-activated receptor alpha (PPARα), fibrates activate renal fatty acid metabolism (FAM) and provide renoprotection. However, fibrate prescription is limited in patients with kidney disease, since impaired urinary excretion of the drug causes serious adverse effects. Pemafibrate (PEM), a novel selective PPARα modulator, is mainly excreted in bile, and, thus, may be safe and effective in kidney disease patients. It remains unclear, however, whether PEM actually exhibits renoprotective properties. We investigated this issue using mice with fatty acid overload nephropathy (FAON). PEM (0.5 mg/kg body weight/day) or a vehicle was administered for 20 days to 13-week-old wild-type male mice, which were simultaneously injected with free fatty acid (FFA)-binding bovine serum albumin from day 7 to day 20 to induce FAON. All mice were sacrificed on day 20 for assessment of the renoprotective effect of PEM against FAON. PEM significantly attenuated the histological findings of tubular injury caused by FAON, increased the renal expressions of mRNA and proteins related to FAM, and decreased renal FFA content and oxidative stress. Taken together, PEM exhibits renoprotective effects through the activation and maintenance of renal FAM and represents a promising drug for kidney disease.
Collapse
|
18
|
Rigaudière F, Delouvrier E, Le Gargasson JF, Milani P, Ogier de Baulny H, Schiff M. Long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency and progressive retinopathy: one case report followed by ERGs, VEPs, EOG over a 17-year period. Doc Ophthalmol 2021; 142:371-380. [PMID: 33392894 DOI: 10.1007/s10633-020-09802-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 10/27/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND LCHAD (long-chain 3-hydroxyacyl-CoA dehydrogenase) deficiency is a rare genetic disorder of mitochondrial long-chain fatty acid oxidation inherited as a recessive trait. Affected patients can present with hypoglycaemia, rhabdomyolysis and cardiomyopathy. About half of the patients may suffer from retinopathy. CASE REPORT A 19-year-old girl was diagnosed as suffering from LCHAD deficiency with recurrent rhabdomyolysis episodes at the age of 7 months by an inaugural coma with hypoglycaemia and hepatomegaly. Appropriate dietary management with carnitine supplementation was initiated. Retinopathy was diagnosed at age two. Ophthalmological assessments including visual acuity, visual field, OCT, flash ERGs, P-ERG, flash VEPs and EOG recordings were conducted over a 17-year period. RESULTS Visual acuity was decreased. Fundi showed a progressive retinopathy and chorioretinopathy. Photophobia was noticed 2 years before the decrease in photopic-ERG amplitude with normal scotopic-ERGs. Scotopic-ERG amplitude decreased 10 years after the decrease in photopic-ERG amplitude. No EOG light rise was observed. Flash VEPs remained normal. These results suggest that the cone system dysfunction occurs largely prior to the rod system dysfunction with a relative preservation of the macula function. COMMENTS This dysfunction of cones prior to the dysfunction of rods was not reported previously. This could be related to mitochondrial energy failure in cones as cones are greater consumers of ATP than rods. This hypothesis needs to be further confirmed as other long-chain fatty oxidation defective patients (VLCAD and CPT2 deficiencies) do not exhibit retinopathy.
Collapse
Affiliation(s)
- Florence Rigaudière
- Service de Physiologie Clinique, Exploration Fonctionnelle, Hôpital Lariboisière, AP-HP, Paris, France. .,Faculté de Médecine Paris-Diderot, Université de Paris, Paris, France.
| | | | - Jean-François Le Gargasson
- Service de Physiologie Clinique, Exploration Fonctionnelle, Hôpital Lariboisière, AP-HP, Paris, France.,Faculté de Médecine Paris-Diderot, Université de Paris, Paris, France
| | - Paolo Milani
- Service de Physiologie Clinique, Exploration Fonctionnelle, Hôpital Lariboisière, AP-HP, Paris, France
| | - Hélène Ogier de Baulny
- Faculté de Médecine Paris-Diderot, Université de Paris, Paris, France.,Reference Center for Inborn Errors of Metabolism, Robert Debré Hospital, AP-HP, Paris, France
| | - Manuel Schiff
- Reference Center for Inborn Errors of Metabolism, Robert Debré Hospital, AP-HP, Paris, France.,Reference Center for Inborn Errors of Metabolism, Faculté de Médecine Paris-Descartes, Necker University Hospital, AP-HP, Université de Paris, Paris, France.,Institut Imagine, Inserm UMRS_1163, Paris, France
| |
Collapse
|
19
|
Wongkittichote P, Watson JR, Leonard JM, Toolan ER, Dickson PI, Grange DK. Fatal COVID-19 infection in a patient with long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency: A case report. JIMD Rep 2020; 56:40-45. [PMID: 33204595 PMCID: PMC7653242 DOI: 10.1002/jmd2.12165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/07/2020] [Accepted: 08/25/2020] [Indexed: 12/30/2022] Open
Abstract
Long-chain fatty-acyl CoA dehydrogenase deficiency (LCHADD) is an inborn error of long chain fatty acid oxidation with various features including hypoketotic hypoglycemia, recurrent rhabdomyolysis, pigmentary retinopathy, peripheral neuropathy, cardiomyopathy, and arrhythmias. Various stresses trigger metabolic decompensation. Coronavirus disease 2019 (COVID-19) is a pandemic caused by the RNA virus SARS-CoV-2 with diverse presentations ranging from respiratory symptoms to myocarditis. We report a case of a patient with LCHADD who initially presented with typical metabolic decompensation symptoms including nausea, vomiting, and rhabdomyolysis in addition to mild cough, and was found to have COVID-19. She developed acute respiratory failure and refractory hypotension from severe cardiomyopathy which progressed to multiple organ failure and death. Our case illustrates the need for close monitoring of cardiac function in patients with a long-chain fatty acid oxidation disorder.
Collapse
Affiliation(s)
- Parith Wongkittichote
- Division of Genetics and Genomic Medicine, Department of PediatricsWashington University School of MedicineSt LouisMissouriUSA
| | - James R. Watson
- Division of Hospital Medicine, Department of MedicineWashington University School of MedicineSt LouisMissouriUSA
| | - Jennifer M. Leonard
- Department of SurgeryWashington University School of MedicineSt LouisMissouriUSA
| | - Elizabeth R. Toolan
- Division of Genetics and Genomic Medicine, Department of PediatricsWashington University School of MedicineSt LouisMissouriUSA
| | - Patricia I. Dickson
- Division of Genetics and Genomic Medicine, Department of PediatricsWashington University School of MedicineSt LouisMissouriUSA
| | - Dorothy K. Grange
- Division of Genetics and Genomic Medicine, Department of PediatricsWashington University School of MedicineSt LouisMissouriUSA
| |
Collapse
|
20
|
Kablau A, Eckert JH, Pistorius J, Sharbati S, Einspanier R. Effects of selected insecticidal substances on mRNA transcriptome in larvae of Apis mellifera. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2020; 170:104703. [PMID: 32980071 DOI: 10.1016/j.pestbp.2020.104703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/03/2020] [Accepted: 09/06/2020] [Indexed: 06/11/2023]
Abstract
For the last decade, scientists have reported a loss of honeybee colonies. Multiple factors like parasites, pathogens and pesticides are dealt as possible drivers of honeybee losses. In particular, insecticides are considered as a major factor of pollinator poisoning. We applied sublethal concentrations of four insecticidal substances to honeybee larval food and analyzed the effects on transcriptome. The aim was to identify candidate genes indicating early negative impacts after application of insecticidal substances. Honeybee larvae were kept in-vitro under hive conditions (34-35 °C) and fed with dimethoate, fenoxycarb, chlorantraniliprole and flupyradifurone in sublethal concentrations between day 3-6 after grafting. Larvae at day 4, 6 and 8 were sampled and their transcriptome analyzed. By use of a RT-qPCR array differences in gene expression of selected gene families (immune system, development detoxification) were measured. Targets mainly involved in development, energy metabolism and the immune system were significantly affected by the insecticidal substances tested, selectively inducing genes of the detoxification system, immune response and nutritional stress.
Collapse
Affiliation(s)
- Arne Kablau
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Germany
| | - Jakob H Eckert
- Julius Kühn-Institute, Institute for Bee Protection, Braunschweig, Germany
| | - Jens Pistorius
- Julius Kühn-Institute, Institute for Bee Protection, Braunschweig, Germany
| | - Soroush Sharbati
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Germany
| | - Ralf Einspanier
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Germany.
| |
Collapse
|
21
|
Khani M, Taheri H, Shamshiri H, Moazzeni H, Hardy J, Bras JT, InanlooRahatloo K, Alavi A, Nafissi S, Elahi E. Deep geno- and phenotyping in two consanguineous families with CMT2 reveals HADHA as an unusual disease-causing gene and an intronic variant in GDAP1 as an unusual mutation. J Neurol 2020; 268:640-650. [PMID: 32897397 DOI: 10.1007/s00415-020-10171-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Charcot-Marie-Tooth (CMT) disease is a prevalent and heterogeneous peripheral neuropathy. Most patients affected with the axonal form of CMT (CMT2) do not harbor mutations in the approximately 90 known CMT-associated genes. We aimed to identify causative genes in two CMT2 pedigrees. METHODS Neurologic examination, laboratory tests and brain MRIs were performed. Genetic analysis included exome sequencing of four patients from the two pedigrees. The predicted effect of a deep intronic mutation on splicing was tested by regular and real-time PCR and sequencing. RESULTS Clinical data were consistent with CMT2 diagnosis. Inheritance patterns were autosomal recessive. Exome data of CMT2-101 did not include mutations in known CMT-associated genes. Sequence data, segregation analysis, bioinformatics analysis, evolutionary conservation, and information in the literature strongly implicated HADHA as the causative gene. An intronic variation positioned 23 nucleotides away from following intron/exon border in GDAP1 was ultimately identified as cause of CMT in CMT2-102. It was shown to affect splicing. CONCLUSION The finding of a HADHA mutation as a cause of CMT is of interest because its encoded protein is a subunit of the mitochondrial trifunctional protein (MTP) complex, a mitochondrial enzyme involved in long chain fatty acid oxidation. Long chain fatty acid oxidation is an important source of energy for skeletal muscles. The mutation found in CMT2-102 is only the second intronic mutation reported in GDAP1. The mutation in the CMT2-102 pedigree was outside the canonical splice site sequences, emphasizing the importance of careful examination of available intronic sequences in exome sequence data.
Collapse
Affiliation(s)
- Marzieh Khani
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Hanieh Taheri
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Hosein Shamshiri
- Department of Neurology, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Moazzeni
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - Jose Tomas Bras
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | | | - Afagh Alavi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Shahriar Nafissi
- Department of Neurology, Tehran University of Medical Sciences, Tehran, Iran.
| | - Elahe Elahi
- School of Biology, College of Science, University of Tehran, Tehran, Iran.
| |
Collapse
|
22
|
Trichloroethylene injures rat liver and elevates the level of peroxisomal bifunctional enzyme (Ehhadh). Mol Cell Toxicol 2020. [DOI: 10.1007/s13273-020-00075-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
23
|
Ruan S, Xiang S, Wu W, Cao S, Du Q, Wang Z, Chen T, Shen Q, Liu L, Chen H, Weng L, Zhu H, Liu Q. Potential role of mTORC1 and the PI3K-Akt pathway in anti-acne properties of licorice flavonoids. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.103968] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
24
|
Donald E, Leb J, Bialer M, Axsom K. A Rare Presentation of Cardiomyopathy in Pregnancy. JACC Case Rep 2020; 2:1066-1069. [PMID: 34317416 PMCID: PMC8302089 DOI: 10.1016/j.jaccas.2020.05.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/29/2020] [Accepted: 05/06/2020] [Indexed: 11/25/2022]
Abstract
Our patient presented in her third trimester of pregnancy with new onset of heart failure. A thorough workup in the initial postpartum period with detailed past medical history, advanced imaging modalities, and a multidisciplinary approach revealed a rare and treatable etiology of cardiomyopathy. (Level of Difficulty: Intermediate.)
Collapse
Affiliation(s)
- Elena Donald
- New York Presbyterian Hospital, Columbia University Irving Medical Center, New York, New York
| | - Jay Leb
- New York Presbyterian Hospital, Columbia University Irving Medical Center, New York, New York
| | - Martin Bialer
- Cohen Children's Medical Center, Northwell Health System, Great Neck, New York
| | - Kelly Axsom
- New York Presbyterian Hospital, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
25
|
Tan C, Liu X, Peng W, Wang H, Zhou W, Jiang J, Wei X, Mo L, Chen Y, Chen L. Seizure-induced impairment in neuronal ketogenesis: Role of zinc-α2-glycoprotein in mitochondria. J Cell Mol Med 2020; 24:6833-6845. [PMID: 32340079 PMCID: PMC7299723 DOI: 10.1111/jcmm.15337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 01/11/2020] [Accepted: 04/12/2020] [Indexed: 12/14/2022] Open
Abstract
Ketone bodies (KBs) were known to suppress seizure. Untraditionally, neurons were recently reported to utilize fatty acids and produce KBs, but the effect of seizure on neuronal ketogenesis has not been researched. Zinc‐α2‐glycoprotein (ZAG) was reported to suppress seizure via unclear mechanism. Interestingly, ZAG was involved in fatty acid β‐oxidation and thus may exert anti‐epileptic effect by promoting ketogenesis. However, this promotive effect of ZAG on neuronal ketogenesis has not been clarified. In this study, we performed immunoprecipitation and mass spectrometry to identify potential interaction partners with ZAG. The mechanisms of how ZAG translocated into mitochondria were determined by quantitative coimmunoprecipitation after treatment with apoptozole, a heat shock cognate protein 70 (HSC70) inhibitor. ZAG level was modulated by lentivirus in neurons or adeno‐associated virus in rat brains. Seizure models were induced by magnesium (Mg2+)‐free artificial cerebrospinal fluid in neurons or intraperitoneal injection of pentylenetetrazole kindling in rats. Ketogenesis was determined by cyclic thio‐NADH method in supernatant of neurons or brain homogenate. The effect of peroxisome proliferator–activated receptor γ (PPARγ) on ZAG expression was examined by Western blot, quantitative real‐time polymerase chain reaction (qRT‐PCR) and chromatin immunoprecipitation qRT‐PCR. We found that seizure induced ketogenesis deficiency via a ZAG‐dependent mechanism. ZAG entered mitochondria through a HSC70‐dependent mechanism, promoted ketogenesis by binding to four β‐subunits of long‐chain L‐3‐hydroxyacyl‐CoA dehydrogenase (HADHB) and alleviated ketogenesis impairment in a neuronal seizure model and pentylenetetrazole‐kindled epileptic rats. Additionally, PPARγ activation up‐regulated ZAG expression by binding to promoter region of AZGP1 gene and promoted ketogenesis through a ZAG‐dependent mechanism.
Collapse
Affiliation(s)
- Changhong Tan
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xi Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wuxue Peng
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Wang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wen Zhou
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Jiang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Wei
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lijuan Mo
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yangmei Chen
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lifen Chen
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
26
|
Nakama M, Sasai H, Kubota M, Hasegawa Y, Fujiki R, Okuyama T, Ohara O, Fukao T. Novel HADHB mutations in a patient with mitochondrial trifunctional protein deficiency. Hum Genome Var 2020; 7:10. [PMID: 32257295 PMCID: PMC7118068 DOI: 10.1038/s41439-020-0097-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/07/2020] [Accepted: 02/21/2020] [Indexed: 01/02/2023] Open
Abstract
We encountered a patient with mitochondrial trifunctional protein deficiency in whom the corresponding mutations were not identified by a DNA panel for newborn screening for targeted diseases. After diagnosis confirmation by an enzyme assay and immunoblotting using the autopsied liver, the re-evaluation of the panel data indicated a heterozygous deletion of exons 6-9 that was later confirmed at the genomic level. cDNA analysis also identified exonization of the 5' region of intron 9 caused by a deep intronic mutation, c.811 + 82A>G.
Collapse
Affiliation(s)
- Mina Nakama
- Clinical Genetics Center, Gifu University Hospital, Gifu, Japan
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Hideo Sasai
- Clinical Genetics Center, Gifu University Hospital, Gifu, Japan
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Mitsuru Kubota
- Department of General Pediatrics & Interdisciplinary Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Yuki Hasegawa
- Department of Pediatrics, Faculty of Medicine, Shimane University, Shimane, Japan
| | - Ryoji Fujiki
- Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan
| | - Torayuki Okuyama
- Department of Clinical Laboratory Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Osamu Ohara
- Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan
| | - Toshiyuki Fukao
- Clinical Genetics Center, Gifu University Hospital, Gifu, Japan
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| |
Collapse
|
27
|
Karri K, Waxman DJ. Widespread Dysregulation of Long Noncoding Genes Associated With Fatty Acid Metabolism, Cell Division, and Immune Response Gene Networks in Xenobiotic-exposed Rat Liver. Toxicol Sci 2020; 174:291-310. [PMID: 31926019 PMCID: PMC7098378 DOI: 10.1093/toxsci/kfaa001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Xenobiotic exposure dysregulates hundreds of protein-coding genes in mammalian liver, impacting many physiological processes and inducing diverse toxicological responses. Little is known about xenobiotic effects on long noncoding RNAs (lncRNAs), many of which have important regulatory functions. Here, we present a computational framework to discover liver-expressed, xenobiotic-responsive lncRNAs (xeno-lncs) with strong functional, gene regulatory potential and elucidate the impact of xenobiotic exposure on their gene regulatory networks. We assembled the long noncoding transcriptome of xenobiotic-exposed rat liver using RNA-seq datasets from male rats treated with 27 individual chemicals, representing 7 mechanisms of action (MOAs). Ortholog analysis was combined with coexpression data and causal inference methods to infer lncRNA function and deduce gene regulatory networks, including causal effects of lncRNAs on protein-coding gene expression and biological pathways. We discovered > 1400 liver-expressed xeno-lncs, many with human and/or mouse orthologs. Xenobiotics representing different MOAs often regulated common xeno-lnc targets: 123 xeno-lncs were dysregulated by ≥ 10 chemicals, and 5 xeno-lncs responded to ≥ 20 of the 27 chemicals investigated; 81 other xeno-lncs served as MOA-selective markers of xenobiotic exposure. Xeno-lnc-protein-coding gene coexpression regulatory network analysis identified xeno-lncs closely associated with exposure-induced perturbations of hepatic fatty acid metabolism, cell division, or immune response pathways, and with apoptosis or cirrhosis. We also identified hub and bottleneck lncRNAs, which are expected to be key regulators of gene expression. This work elucidates extensive networks of xeno-lnc-protein-coding gene interactions and provides a framework for understanding the widespread transcriptome-altering actions of foreign chemicals in a key-responsive mammalian tissue.
Collapse
Affiliation(s)
- Kritika Karri
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, Massachusetts
| |
Collapse
|
28
|
Khare T, Khare S, Angdisen JJ, Zhang Q, Stuckel A, Mooney BP, Ridenhour SE, Gitan RS, Hammoud GM, Ibdah JA. Defects in long-chain 3-hydroxy acyl-CoA dehydrogenase lead to hepatocellular carcinoma: A novel etiology of hepatocellular carcinoma. Int J Cancer 2020; 147:1461-1473. [PMID: 32115688 DOI: 10.1002/ijc.32943] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 01/02/2020] [Accepted: 02/17/2020] [Indexed: 12/14/2022]
Abstract
The incidence of both nonalcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma (HCC) have been increasing at an alarming rate. Little is known about NAFLD without cirrhosis as a risk for HCC. Here we report, for the first time, generation of a mouse model with a defect in long-chain 3-hydoxy acyl-CoA dehydrogenase (LCHAD). The LCHAD exon 15 deletion was embryonic lethal to the homozygous mice whereas heterozygous mice (HT) develop significant hepatic steatosis starting at young age (3 months old) and HCC at older age (>13 months old) without any evidence of fibrosis or cirrhosis. None of the wild-type (WT) mice developed steatosis and HCC (n = 39), whereas HT-LCHAD mice (n = 41) showed steatosis and ~20% (8/41) developed liver masses with histological features of HCC. Proteomic analysis of liver tissues from WT-mice and HT-mice with no signs of HCC was conducted. Proteins with significant changes in abundance were identified by mass spectrometry. Abundance of 24 proteins was significantly different (p < 0.01) between WT and HT-LCHAD mice. The proteins found to vary in abundance are associated with different cellular response processes ranging from intermediary metabolism of carbohydrate, protein and lipid to oxidative stress, signal transduction and the process of tumorigenesis. Protein expression pattern of the HT-LCHAD mouse liver indicates predisposition to HCC and suggests that impaired hepatic mitochondrial fatty acid oxidation plays an important role in the development and progression of HCC. To assess the implication of these studies in human disease, we demonstrated significant downregulation of HADHA transcripts in HCC patients.
Collapse
Affiliation(s)
- Tripti Khare
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Sharad Khare
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA.,Harry S Truman Veterans' Hospital, Columbia, MO, USA
| | - Jerry J Angdisen
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Qiong Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Alexei Stuckel
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Brian P Mooney
- Gehrke Proteomics center, University of Missouri, Columbia, MO, USA
| | - Suzanne E Ridenhour
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Raad S Gitan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Ghassan M Hammoud
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Jamal A Ibdah
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA.,Harry S Truman Veterans' Hospital, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
29
|
Yang Y, Tao J, Zong S. Identification of putative Type-I sex pheromone biosynthesis-related genes expressed in the female pheromone gland of Streltzoviella insularis. PLoS One 2020; 15:e0227666. [PMID: 31945099 PMCID: PMC6964838 DOI: 10.1371/journal.pone.0227666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 12/24/2019] [Indexed: 11/18/2022] Open
Abstract
Species-specific sex pheromones play key roles in moth sexual communication. Although the general pathway of Type-I sex pheromone biosynthesis is well established, only a handful of genes encoding enzymes involved in this pathway have been characterized. Streltzoviella insularis is a destructive wood-boring pest of many street trees in China, and the female sex pheromone of this species comprises a blend of (Z)-3-tetradecenyl acetate, (E)-3-tetradecenyl acetate, and (Z)-5-dodecenyl acetate. This organism therefore provides an excellent model for research on the diversity of genes and molecular mechanisms involved in pheromone production. Herein, we assembled the pheromone gland transcriptome of S. insularis by next-generation sequencing and identified 74 genes encoding candidate key enzymes involved in the fatty acid biosynthesis, β-oxidation, and functional group modification. In addition, tissue expression patterns further showed that an acetyl-CoA carboxylase and two desaturases were highly expressed in the pheromone glands compared with the other tissues, indicating possible roles in S. insularis sex pheromone biosynthesis. Finally, we proposed putative S. insularis biosynthetic pathways for sex pheromone components and highlighted candidate genes. Our findings lay a solid foundation for understanding the molecular mechanisms underpinning S. insularis sex pheromone biosynthesis, and provide potential targets for disrupting chemical communication that could assist the development of novel pest control methods.
Collapse
Affiliation(s)
- Yuchao Yang
- Beijing Key Laboratory for Forest Pest Control, School of Forestry, Beijing Forestry University, Beijing, China
| | - Jing Tao
- Beijing Key Laboratory for Forest Pest Control, School of Forestry, Beijing Forestry University, Beijing, China
| | - Shixiang Zong
- Beijing Key Laboratory for Forest Pest Control, School of Forestry, Beijing Forestry University, Beijing, China
| |
Collapse
|
30
|
Ravera S, Ferrando S, Agas D, De Angelis N, Raffetto M, Sabbieti MG, Signore A, Benedicenti S, Amaroli A. 1064 nm Nd:YAG laser light affects transmembrane mitochondria respiratory chain complexes. JOURNAL OF BIOPHOTONICS 2019; 12:e201900101. [PMID: 31033186 DOI: 10.1002/jbio.201900101] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 04/23/2019] [Accepted: 04/26/2019] [Indexed: 06/09/2023]
Abstract
Photobiomodulation (PBM) is a non-plant-cell manipulation through a transfer of energy by means of light sources at the non-ablative or thermal intensity. Authors showed that cytochrome-c-oxidase (complex IV) is the specific chromophore's target of PBM at the red (600-700 nm) and NIR (760-900 nm) wavelength regions. Recently, it was suggested that the infrared region of the spectrum could influence other chromospheres, despite the interaction by wavelengths higher than 900 nm with mitochondrial chromophores was not clearly demonstrated. We characterized the interaction between mitochondria respiratory chain, malate dehydrogenase, a key enzyme of Krebs cycle, and 3-hydroxyacyl-CoA dehydrogenase, an enzyme involved in the β-oxidation (two mitochondrial matrix enzymes) with the 1064 nm Nd:YAG (100mps and 10 Hz frequency mode) irradiated at the average power density of 0.50, 0.75, 1.00, 1.25 and 1.50 W/cm2 to generate the respective fluences of 30, 45, 60, 75 and 90 J/cm2 . Our results show the effect of laser light on the transmembrane mitochondrial complexes I, III, IV and V (adenosine triphosphate synthase) (window effects), but not on the extrinsic mitochondrial membrane complex II and mitochondria matrix enzymes. The effect is not due to macroscopical thermal change. An interaction of this wavelength with the Fe-S proteins and Cu-centers of respiratory complexes and with the water molecules could be supposed.
Collapse
Affiliation(s)
- Silvia Ravera
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Sara Ferrando
- Laboratory of New Model Organism (NeMo LAB), Department of Earth, Environmental and Life Sciences, University of Genoa, Genoa, Italy
| | - Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (Macerata), Italy
| | - Nicola De Angelis
- Laser Therapy Centre, Department of Surgical and Diagnostic Sciences (D.I.S.C), University of Genoa, Genoa, Italy
- Department of Dentistry, University of Technology MARA, Sungai Buloh, Malaysia
| | - Mirco Raffetto
- Department of Electrical, Electronic, Telecommunications Engineering and Naval Architecture, University of Genoa, Genoa, Italy
| | - Maria G Sabbieti
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (Macerata), Italy
| | - Antonio Signore
- Laser Therapy Centre, Department of Surgical and Diagnostic Sciences (D.I.S.C), University of Genoa, Genoa, Italy
- Faculty of Therapeutic Stomatology, Institute of Dentistry, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Stefano Benedicenti
- Laser Therapy Centre, Department of Surgical and Diagnostic Sciences (D.I.S.C), University of Genoa, Genoa, Italy
| | - Andrea Amaroli
- Laser Therapy Centre, Department of Surgical and Diagnostic Sciences (D.I.S.C), University of Genoa, Genoa, Italy
| |
Collapse
|
31
|
Ji C, Lu Z, Xu L, Li F, Cong M, Shan X, Wu H. Evaluation of mitochondrial toxicity of cadmium in clam Ruditapes philippinarum using iTRAQ-based proteomics. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 251:802-810. [PMID: 31125810 DOI: 10.1016/j.envpol.2019.05.046] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 04/30/2019] [Accepted: 05/10/2019] [Indexed: 06/09/2023]
Abstract
Cadmium is one of the most serious metal pollutants in the Bohai Sea. Previous studies revealed that mitochondrion might be the target organelle of Cd toxicity. However, there is a lack of a global view on the mitochondrial responses in marine animals to Cd. In this work, the mitochondrial responses were characterized in clams Ruditapes philippinarum treated with two concentrations (5 and 50 μg/L) of Cd for 5 weeks using tetraethylbenzimidazolylcarbocyanine iodide (JC-1) staining, ultrastructural observation and quantitative proteomic analysis. Basically, a significant decrease of mitochondrial membrane potential (△Ψm) was observed in clams treated with the high concentration (50 μg/L) of Cd. Cd treatments also induced specific morphological changes indicated by elongated mitochondria. Furthermore, iTRAQ-based mitochondrial proteomics showed that a total of 97 proteins were significantly altered in response to Cd treatment. These proteins were closely associated with multiple biological processes in mitochondria, including tricarboxylic acid (TCA) cycle, oxidative phosphorylation, fatty acid β-oxidation, stress resistance and apoptosis, and mitochondrial fission. These findings confirmed that mitochondrion was one of the key targets of Cd toxicity. Moreover, dynamical regulations, such as reconstruction of energy homeostasis, induction of stress resistance and apoptosis, and morphological alterations, in mitochondria might play essential roles in Cd tolerance. Overall, this work provided a deep insight into the mitochondrial toxicity of Cd in clams based on a global mitochondrial proteomic analysis.
Collapse
Affiliation(s)
- Chenglong Ji
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai, 264003, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, PR China
| | - Zhen Lu
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai, 264003, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Lanlan Xu
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai, 264003, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Fei Li
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai, 264003, PR China
| | - Ming Cong
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai, 264003, PR China
| | - Xiujuan Shan
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, PR China
| | - Huifeng Wu
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai, 264003, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, PR China.
| |
Collapse
|
32
|
Complementary substrate specificity and distinct quaternary assembly of the Escherichia coli aerobic and anaerobic β-oxidation trifunctional enzyme complexes. Biochem J 2019; 476:1975-1994. [DOI: 10.1042/bcj20190314] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/20/2019] [Accepted: 06/24/2019] [Indexed: 02/03/2023]
Abstract
AbstractThe trifunctional enzyme (TFE) catalyzes the last three steps of the fatty acid β-oxidation cycle. Two TFEs are present in Escherichia coli, EcTFE and anEcTFE. EcTFE is expressed only under aerobic conditions, whereas anEcTFE is expressed also under anaerobic conditions, with nitrate or fumarate as the ultimate electron acceptor. The anEcTFE subunits have higher sequence identity with the human mitochondrial TFE (HsTFE) than with the soluble EcTFE. Like HsTFE, here it is found that anEcTFE is a membrane-bound complex. Systematic enzyme kinetic studies show that anEcTFE has a preference for medium- and long-chain enoyl-CoAs, similar to HsTFE, whereas EcTFE prefers short chain enoyl-CoA substrates. The biophysical characterization of anEcTFE and EcTFE shows that EcTFE is heterotetrameric, whereas anEcTFE is purified as a complex of two heterotetrameric units, like HsTFE. The tetrameric assembly of anEcTFE resembles the HsTFE tetramer, although the arrangement of the two anEcTFE tetramers in the octamer is different from the HsTFE octamer. These studies demonstrate that EcTFE and anEcTFE have complementary substrate specificities, allowing for complete degradation of long-chain enoyl-CoAs under aerobic conditions. The new data agree with the notion that anEcTFE and HsTFE are evolutionary closely related, whereas EcTFE belongs to a separate subfamily.
Collapse
|
33
|
Crystal structure of human mitochondrial trifunctional protein, a fatty acid β-oxidation metabolon. Proc Natl Acad Sci U S A 2019; 116:6069-6074. [PMID: 30850536 DOI: 10.1073/pnas.1816317116] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Membrane-bound mitochondrial trifunctional protein (TFP) catalyzes β-oxidation of long chain fatty acyl-CoAs, employing 2-enoyl-CoA hydratase (ECH), 3-hydroxyl-CoA dehydrogenase (HAD), and 3-ketothiolase (KT) activities consecutively. Inherited deficiency of TFP is a recessive genetic disease, manifesting in hypoketotic hypoglycemia, cardiomyopathy, and sudden death. We have determined the crystal structure of human TFP at 3.6-Å resolution. The biological unit of the protein is α2β2 The overall structure of the heterotetramer is the same as that observed by cryo-EM methods. The two β-subunits make a tightly bound homodimer at the center, and two α-subunits are bound to each side of the β2 dimer, creating an arc, which binds on its concave side to the mitochondrial innermembrane. The catalytic residues in all three active sites are arranged similarly to those of the corresponding, soluble monofunctional enzymes. A structure-based, substrate channeling pathway from the ECH active site to the HAD and KT sites is proposed. The passage from the ECH site to the HAD site is similar to those found in the two bacterial TFPs. However, the passage from the HAD site to the KT site is unique in that the acyl-CoA intermediate can be transferred between the two sites by passing along the mitochondrial inner membrane using the hydrophobic nature of the acyl chain. The 3'-AMP-PPi moiety is guided by the positively charged residues located along the "ceiling" of the channel, suggesting that membrane integrity is an essential part of the channel and is required for the activity of the enzyme.
Collapse
|
34
|
Zhang Y, Zhou F, Bai M, Liu Y, Zhang L, Zhu Q, Bi Y, Ning G, Zhou L, Wang X. The pivotal role of protein acetylation in linking glucose and fatty acid metabolism to β-cell function. Cell Death Dis 2019; 10:66. [PMID: 30683850 PMCID: PMC6347623 DOI: 10.1038/s41419-019-1349-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/12/2018] [Accepted: 01/02/2019] [Indexed: 01/16/2023]
Abstract
Protein acetylation has a crucial role in energy metabolism. Here we performed the first large-scale profiling of acetylome in rat islets, showing that almost all enzymes in core metabolic pathways related to insulin secretion were acetylated. Label-free quantitative acetylome of islets in response to high glucose revealed hyperacetylation of enzymes involved in fatty acid β-oxidation (FAO), including trifunctional enzyme subunit alpha (ECHA). Acetylation decreased the protein stability of ECHA and its ability to promote FAO. The overexpression of SIRT3, a major mitochondrial deacetylase, prevented the degradation of ECHA via decreasing its acetylation level in β-cells. SIRT3 expression was upregulated in rat islets upon exposure to low glucose or fasting. SIRT3 overexpression in islets markedly decreased palmitate-potentiated insulin secretion, whereas islets from SIRT3 knockout mice secreted more insulin, with an opposite action on FAO. ECHA overexpression partially reversed SIRT3 deficiency-elicited insulin hypersecretion. Our study highlights the potential role of protein acetylation in insulin secretion.
Collapse
Affiliation(s)
- Yuqing Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.,Center for Reproductive Medicine, Shandong University, Jinan, 250000, China.,Key Laboratory of Reproductive Endocrinology, Ministry of Education, Shandong University, Jinan, 250000, China
| | - Feiye Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Mengyao Bai
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Yun Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Linlin Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Qin Zhu
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| | - Libin Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| | - Xiao Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
35
|
Abstract
The mitochondrial trifunctional protein (TFP) catalyzes three reactions in the fatty acid β-oxidation process. Mutations in the two TFP subunits cause mitochondrial trifunctional protein deficiency and acute fatty liver of pregnancy that can lead to death. Here we report a 4.2-Å cryo-electron microscopy α2β2 tetrameric structure of the human TFP. The tetramer has a V-shaped architecture that displays a distinct assembly compared with the bacterial TFPs. A concave surface of the TFP tetramer interacts with the detergent molecules in the structure, suggesting that this region is involved in associating with the membrane. Deletion of a helical hairpin in TFPβ decreases its binding to the liposomes in vitro and reduces its membrane targeting in cells. Our results provide the structural basis for TFP function and have important implications for fatty acid oxidation related diseases.
Collapse
|
36
|
Nassir F, Arndt JJ, Johnson SA, Ibdah JA. Regulation of mitochondrial trifunctional protein modulates nonalcoholic fatty liver disease in mice. J Lipid Res 2018; 59:967-973. [PMID: 29581157 PMCID: PMC5983392 DOI: 10.1194/jlr.m080952] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 03/21/2018] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial trifunctional protein (MTP) plays a critical role in the oxidation of long-chain fatty acids. We previously reported that aging mice (>9 months old) heterozygous for an MTP defect (MTP+/-) develop nonalcoholic fatty liver disease (NAFLD). We tested whether a high-fat diet (HFD) accelerates NAFLD in young MTP+/-mice, and whether overexpression of the nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase sirtuin 3 (SIRT3) deacetylates MTP and improves mitochondrial function and NAFLD. Three-month-old WT and MTP+/- mice were fed HFD (60% cal fat) for 16 weeks and livers were assessed for fatty acid oxidation (FAO) and NAFLD. Compared with WT, MTP+/- mice displayed reduced hepatic SIRT3 levels and reduced FAO, with increased hepatic steatosis and the inflammatory marker CD68. Hepatic overexpression of SIRT3 in HFD-fed MTP+/- mice increased hepatic MTP protein levels at the posttranscriptional level. Immunoprecipitation of MTP from liver mitochondria followed by Western blot with acetyl-lysine antibody showed higher acetylation of MTP in MTP+/- compared with WT mice. Overexpression of SIRT3 in MTP+/- mice significantly reduced the acetylation of MTP compared with β-galactosidase controls, increased mitochondrial FAO, and reduced hepatic steatosis, CD68, and serum ALT levels. Taken together, our data indicate that deacetylation of MTP by SIRT3 improves mitochondrial function and rescues NAFLD in MTP+/- mice.
Collapse
Affiliation(s)
- Fatiha Nassir
- Departments of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO; Nutrition and Exercise Physiology, University of Missouri, Columbia, MO; United States Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
| | - Justin J Arndt
- Departments of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO
| | - Sarah A Johnson
- Departments of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO; United States Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO
| | - Jamal A Ibdah
- Departments of Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO; Nutrition and Exercise Physiology, University of Missouri, Columbia, MO; Medical Pharmacology & Physiology, University of Missouri, Columbia, MO; United States Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO.
| |
Collapse
|
37
|
A Nonsense Variant in the ACADVL Gene in German Hunting Terriers with Exercise Induced Metabolic Myopathy. G3-GENES GENOMES GENETICS 2018; 8:1545-1554. [PMID: 29491033 PMCID: PMC5940147 DOI: 10.1534/g3.118.200084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Several enzymes are involved in fatty acid oxidation, which is a key process in mitochondrial energy production. Inherited defects affecting any step of fatty acid oxidation can result in clinical disease. We present here an extended family of German Hunting Terriers with 10 dogs affected by clinical signs of exercise induced weakness, muscle pain, and suspected rhabdomyolysis. The combination of clinical signs, muscle histopathology and acylcarnitine analysis with an elevated tetradecenoylcarnitine (C14:1) peak suggested a possible diagnosis of acyl-CoA dehydrogenase very long chain deficiency (ACADVLD). Whole genome sequence analysis of one affected dog and 191 controls revealed a nonsense variant in the ACADVL gene encoding acyl-CoA dehydrogenase very long chain, c.1728C>A or p.(Tyr576*). The variant showed perfect association with the phenotype in the 10 affected and more than 500 control dogs of various breeds. Pathogenic variants in the ACADVL gene have been reported in humans with similar myopathic phenotypes. We therefore considered the detected variant to be the most likely candidate causative variant for the observed exercise induced myopathy. To our knowledge, this is the first description of this disease in dogs, which we propose to name exercise induced metabolic myopathy (EIMM), and the identification of the first canine pathogenic ACADVL variant. Our findings provide a large animal model for a known human disease and will enable genetic testing to avoid the unintentional breeding of affected offspring.
Collapse
|
38
|
El-Gharbawy A, Vockley J. Inborn Errors of Metabolism with Myopathy: Defects of Fatty Acid Oxidation and the Carnitine Shuttle System. Pediatr Clin North Am 2018; 65:317-335. [PMID: 29502916 PMCID: PMC6566095 DOI: 10.1016/j.pcl.2017.11.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Fatty acid oxidation disorders (FAODs) and carnitine shuttling defects are inborn errors of energy metabolism with associated mortality and morbidity due to cardiomyopathy, exercise intolerance, rhabdomyolysis, and liver disease with physiologic stress. Hypoglycemia is characteristically hypoketotic. Lactic acidemia and hyperammonemia may occur during decompensation. Recurrent rhabdomyolysis is debilitating. Expanded newborn screening can detect most of these disorders, allowing early, presymptomatic treatment. Treatment includes avoiding fasting and sustained extraneous exercise and providing high-calorie hydration during illness to prevent lipolysis, and medium-chain triglyceride oil supplementation in long-chain FAODs. Carnitine supplementation may be helpful. However, conventional treatment does not prevent all symptoms.
Collapse
Affiliation(s)
- Areeg El-Gharbawy
- Department of Pediatrics, Division of Medical Genetics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA; Cairo University, Kasr Al-Aini, Cairo, Egypt
| | - Jerry Vockley
- Department of Pediatrics, Division of Medical Genetics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA.
| |
Collapse
|
39
|
In vivo studies on the mechanism of methylene cyclopropyl acetic acid and methylene cyclopropyl glycine-induced hypoglycemia. Biochem J 2018; 475:1063-1074. [PMID: 29483297 DOI: 10.1042/bcj20180063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/24/2018] [Accepted: 02/26/2018] [Indexed: 01/20/2023]
Abstract
Exposure to the toxins methylene cyclopropyl acetic acid (MCPA) and methylene cyclopropyl glycine (MCPG) of unripe ackee and litchi fruit can lead to hypoglycemia and death; however, the molecular mechanisms by which MCPA and MCPG cause hypoglycemia have not been established in vivo To determine the in vivo mechanisms of action of these toxins, we infused them into conscious rodents and assessed rates of hepatic gluconeogenesis and ketogenesis, hepatic acyl-CoA and hepatic acetyl-CoA content, and hepatocellular energy charge. MCPG suppressed rates of hepatic β-oxidation as reflected by reductions in hepatic ketogenesis, reducing both short- and medium-chain hepatic acyl-CoA concentrations. Hepatic acetyl-CoA content decreased, and hepatic glucose production was inhibited. MCPA also suppressed β-oxidation of short-chain acyl-CoAs, rapidly inhibiting hepatic ketogenesis and hepatic glucose production, depleting hepatic acetyl-CoA content and ATP content, while increasing other short-chain acyl-CoAs. Utilizing a recently developed positional isotopomer NMR tracer analysis method, we demonstrated that MCPA-induced reductions in hepatic acetyl-CoA content were associated with a marked reduction of hepatic pyruvate carboxylase (PC) flux. Taken together, these data reveal the in vivo mechanisms of action of MCPA and MCPG: the hypoglycemia associated with ingestion of these toxins can be ascribed mostly to MCPA- or MCPG-induced reductions in hepatic PC flux due to inhibition of β-oxidation of short-chain acyl-CoAs by MCPA or inhibition of both short- and medium-chain acyl-CoAs by MCPG with resultant reductions in hepatic acetyl-CoA content, with an additional contribution to hypoglycemia through reduced hepatic ATP stores by MCPA.
Collapse
|
40
|
Bursle C, Weintraub R, Ward C, Justo R, Cardinal J, Coman D. Mitochondrial Trifunctional Protein Deficiency: Severe Cardiomyopathy and Cardiac Transplantation. JIMD Rep 2017; 40:91-95. [PMID: 29124685 PMCID: PMC6122028 DOI: 10.1007/8904_2017_68] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/17/2017] [Accepted: 10/19/2017] [Indexed: 12/14/2022] Open
Abstract
We describe mitochondrial trifunctional protein deficiency (MTPD) in two male siblings who presented with severe cardiomyopathy in infancy. The first sibling presented in severe cardiac failure at 6 months of age and succumbed soon after. The second sibling came to attention after newborn screening identified a possible fatty acid oxidation defect. Dietary therapy and carnitine supplementation commenced in the neonatal period. Despite this the second child required cardiac transplantation at 3 years of age after a sudden and rapid decline in cardiac function. The outcome has been excellent, with no apparent extra-cardiac manifestations of a fatty acid oxidation disorder at the age of 7. Pathogenic HADHA mutations were subsequently identified via genome wide exome sequencing. This is the first reported case of MTPD to undergo cardiac transplantation. We suggest that cardiac transplantation could be considered in the treatment of cardiomyopathy in MTPD.
Collapse
Affiliation(s)
- C Bursle
- Department of Metabolic Medicine, The Lady Cilento Children's Hospital, Brisbane, QLD, Australia
- School of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - R Weintraub
- Department of Cardiology, The Royal Children's Hospital, Melbourne, VIC, Australia
- School of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - C Ward
- School of Medicine, University of Queensland, Brisbane, QLD, Australia
- Department of Cardiology, The Lady Cilento Children's Hospital, Brisbane, QLD, Australia
| | - R Justo
- School of Medicine, University of Queensland, Brisbane, QLD, Australia
- Department of Cardiology, The Lady Cilento Children's Hospital, Brisbane, QLD, Australia
| | - J Cardinal
- Cardinal Bioresearch, Brisbane, QLD, Australia
| | - D Coman
- Department of Metabolic Medicine, The Lady Cilento Children's Hospital, Brisbane, QLD, Australia.
- School of Medicine, University of Queensland, Brisbane, QLD, Australia.
- Department of Paediatrics, The Wesley Hospital, Brisbane, QLD, Australia.
- School of Medicine, Griffith University, Gold Coast, QLD, Australia.
| |
Collapse
|
41
|
Zeng J, Deng S, Wang Y. Identification of the Catalytic Residue of Rat Acyl-CoA Dehydrogenase 9 by Site-Directed Mutagenesis. Appl Biochem Biotechnol 2017; 182:1198-1207. [DOI: 10.1007/s12010-016-2392-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 12/29/2016] [Indexed: 10/20/2022]
|
42
|
Clinical and molecular investigation of 14 Japanese patients with complete TFP deficiency: a comparison with Caucasian cases. J Hum Genet 2017; 62:809-814. [DOI: 10.1038/jhg.2017.52] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 04/12/2017] [Accepted: 04/12/2017] [Indexed: 12/30/2022]
|
43
|
Yang XH, Yang SF, Wang RM. Comparative proteomic analysis provides insight into 10-hydroxy-2-decenoic acid biosynthesis in honey bee workers. Amino Acids 2017; 49:1177-1192. [DOI: 10.1007/s00726-017-2418-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 04/08/2017] [Indexed: 12/01/2022]
|
44
|
Martines ACMF, van Eunen K, Reijngoud DJ, Bakker BM. The promiscuous enzyme medium-chain 3-keto-acyl-CoA thiolase triggers a vicious cycle in fatty-acid beta-oxidation. PLoS Comput Biol 2017; 13:e1005461. [PMID: 28369071 PMCID: PMC5397069 DOI: 10.1371/journal.pcbi.1005461] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 04/19/2017] [Accepted: 03/16/2017] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial fatty-acid beta-oxidation (mFAO) plays a central role in mammalian energy metabolism. Multiple severe diseases are associated with defects in this pathway. Its kinetic structure is characterized by a complex wiring of which the functional implications have hardly been explored. Repetitive cycles of reversible reactions, each cycle shortening the fatty acid by two carbon atoms, evoke competition between intermediates of different chain lengths for a common set of 'promiscuous' enzymes (enzymes with activity towards multiple substrates). In our validated kinetic model of the pathway, substrate overload causes a steep and detrimental flux decline. Here, we unravel the underlying mechanism and the role of enzyme promiscuity in it. Comparison of alternative model versions elucidated the role of promiscuity of individual enzymes. Promiscuity of the last enzyme of the pathway, medium-chain ketoacyl-CoA thiolase (MCKAT), was both necessary and sufficient to elicit the flux decline. Subsequently, Metabolic Control Analysis revealed that MCKAT had insufficient capacity to cope with high substrate influx. Next, we quantified the internal metabolic regulation, revealing a vicious cycle around MCKAT. Upon substrate overload, MCKAT's ketoacyl-CoA substrates started to accumulate. The unfavourable equilibrium constant of the preceding enzyme, medium/short-chain hydroxyacyl-CoA dehydrogenase, worked as an amplifier, leading to accumulation of upstream CoA esters, including acyl-CoA esters. These acyl-CoA esters are at the same time products of MCKAT and inhibited its already low activity further. Finally, the accumulation of CoA esters led to a sequestration of free CoA. CoA being a cofactor for MCKAT, its sequestration limited the MCKAT activity even further, thus completing the vicious cycle. Since CoA is also a substrate for distant enzymes, it efficiently communicated the 'traffic jam' at MCKAT to the entire pathway. This novel mechanism provides a basis to explore the role of mFAO in disease and elucidate similar principles in other pathways of lipid metabolism.
Collapse
Affiliation(s)
- Anne-Claire M. F. Martines
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, The Netherlands
- Systems Biology Centre for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Karen van Eunen
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, The Netherlands
- Systems Biology Centre for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Dirk-Jan Reijngoud
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, The Netherlands
- Systems Biology Centre for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Barbara M. Bakker
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, The Netherlands
- Systems Biology Centre for Energy Metabolism and Ageing, University of Groningen, University Medical Center Groningen, The Netherlands
| |
Collapse
|
45
|
Ojuka E, Andrew B, Bezuidenhout N, George S, Maarman G, Madlala HP, Mendham A, Osiki PO. Measurement of β-oxidation capacity of biological samples by respirometry: a review of principles and substrates. Am J Physiol Endocrinol Metab 2016; 310:E715-23. [PMID: 26908505 DOI: 10.1152/ajpendo.00475.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 02/22/2016] [Indexed: 02/06/2023]
Abstract
Oxidation of fatty acids is a major source of energy in the heart, liver, and skeletal muscle. It can be measured accurately using respirometry in isolated mitochondria, intact cells, and permeabilized cells or tissues. This technique directly measures the rate of oxygen consumption or flux at various respiratory states when appropriate substrates, uncouplers, and inhibitors are used. Acylcarnitines such as palmitoylcarnitine or octanoylcarnitine are the commonly used substrates. The β-oxidation pathway is prone to feedforward inhibition resulting from accumulation of short-chain acyl-CoA and depletion of CoA, but inclusion of malate or carnitine prevents accumulation of these intermediaries and CoA depletion.
Collapse
Affiliation(s)
- Edward Ojuka
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Brittany Andrew
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Nicole Bezuidenhout
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Siddiqah George
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Gerald Maarman
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Hlengiwe P Madlala
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Amy Mendham
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Prisca Ofure Osiki
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
46
|
Metabolomics-assisted proteomics identifies succinylation and SIRT5 as important regulators of cardiac function. Proc Natl Acad Sci U S A 2016; 113:4320-5. [PMID: 27051063 DOI: 10.1073/pnas.1519858113] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cellular metabolites, such as acyl-CoA, can modify proteins, leading to protein posttranslational modifications (PTMs). One such PTM is lysine succinylation, which is regulated by sirtuin 5 (SIRT5). Although numerous proteins are modified by lysine succinylation, the physiological significance of lysine succinylation and SIRT5 remains elusive. Here, by profiling acyl-CoA molecules in various mouse tissues, we have discovered that different tissues have different acyl-CoA profiles and that succinyl-CoA is the most abundant acyl-CoA molecule in the heart. This interesting observation has prompted us to examine protein lysine succinylation in different mouse tissues in the presence and absence of SIRT5. Protein lysine succinylation predominantly accumulates in the heart whenSirt5is deleted. Using proteomic studies, we have identified many cardiac proteins regulated by SIRT5. Our data suggest that ECHA, a protein involved in fatty acid oxidation, is a major enzyme that is regulated by SIRT5 and affects heart function.Sirt5knockout (KO) mice have lower ECHA activity, increased long-chain acyl-CoAs, and decreased ATP in the heart under fasting conditions.Sirt5KO mice develop hypertrophic cardiomyopathy, as evident from the increased heart weight relative to body weight, as well as reduced shortening and ejection fractions. These findings establish that regulating heart metabolism and function is a major physiological function of lysine succinylation and SIRT5.
Collapse
|
47
|
Sehrawat U, Pokhriyal R, Gupta AK, Hariprasad R, Khan MI, Gupta D, Naru J, Singh SB, Mohanty AK, Vanamail P, Kumar L, Kumar S, Hariprasad G. Comparative Proteomic Analysis of Advanced Ovarian Cancer Tissue to Identify Potential Biomarkers of Responders and Nonresponders to First-Line Chemotherapy of Carboplatin and Paclitaxel. BIOMARKERS IN CANCER 2016; 8:43-56. [PMID: 26997873 PMCID: PMC4795487 DOI: 10.4137/bic.s35775] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 02/08/2016] [Accepted: 02/11/2016] [Indexed: 12/16/2022]
Abstract
Conventional treatment for advanced ovarian cancer is an initial debulking surgery followed by chemotherapy combination of carboplatin and paclitaxel. Despite initial high response, three-fourths of these women experience disease recurrence with a dismal prognosis. Patients with advanced-stage ovarian cancer who underwent cytoreductive surgery were enrolled and tissue samples were collected. Post surgery, these patients were started on chemotherapy and followed up till the end of the cycle. Fluorescence-based differential in-gel expression coupled with mass spectrometric analysis was used for discovery phase of experiments, and real-time polymerase chain reaction, Western blotting, and pathway analysis were performed for expression and functional validation of differentially expressed proteins. While aldehyde reductase, hnRNP, cyclophilin A, heat shock protein-27, and actin are upregulated in responders, prohibitin, enoyl-coA hydratase, peroxiredoxin, and fibrin-β are upregulated in the nonresponders. The expressions of some of these proteins correlated with increased apoptotic activity in responders and decreased apoptotic activity in nonresponders. Therefore, the proteins qualify as potential biomarkers to predict chemotherapy response.
Collapse
Affiliation(s)
- Urmila Sehrawat
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Ruchika Pokhriyal
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Ashish Kumar Gupta
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Roopa Hariprasad
- Department of Medical Oncology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Mohd Imran Khan
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Divya Gupta
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Jasmine Naru
- National Dairy Research Institute, Karnal, India
| | | | | | - Perumal Vanamail
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Lalit Kumar
- Department of Medical Oncology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Sunesh Kumar
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Gururao Hariprasad
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| |
Collapse
|
48
|
Bo R, Hasegawa Y, Yamada K, Kobayashi H, Taketani T, Fukuda S, Yamaguchi S. A fetus with mitochondrial trifunctional protein deficiency: Elevation of 3-OH-acylcarnitines in amniotic fluid functionally assured the genetic diagnosis. Mol Genet Metab Rep 2015; 6:1-4. [PMID: 27014569 PMCID: PMC4789351 DOI: 10.1016/j.ymgmr.2015.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/25/2015] [Accepted: 11/25/2015] [Indexed: 10/27/2022] Open
Abstract
Mitochondrial trifunctional protein (TFP) is a multienzyme complex that catalyzes the last three steps of the β-oxidation cycle of long-chain fatty acids. In the prenatal diagnosis of TFP deficiency, acylcarnitine (AC) analysis has been considered difficult because of limited excretion of long-chain ACs into the fetal urine and hence into the amniotic fluid. Here, we report our experience with prenatally diagnosing TFP deficiency using AC analysis of amniotic fluid. The index case was a boy born at 38 weeks gestation and weighing 2588 g. He suddenly became unconscious and hypoglycemic and died on day 6 of life. Postmortem blood AC analysis and gene sequencing revealed TFP deficiency. Therefore, the parents underwent prenatal diagnoses for their subsequent 2 pregnancies. Mutation analysis suggested that one (Case 1) was affected and the other (Case 2) was not. AC analysis also demonstrated identical results, with significantly elevated 3-hydroxy-AC levels in the amniotic fluid of the affected pregnancy compared with those of heterozygotes and normal controls (n = 2 for heterozygotes and n = 8 for normal controls). Our findings suggest that AC analysis can functionally confirm results even in families with unidentified mutations, without raising issues related to maternal cell contamination. During prenatal diagnosis, misdiagnosis has to be avoided, and combining AC analysis with gene sequencing may result in more accurate prenatal diagnosis of TFP deficiency.
Collapse
Affiliation(s)
- Ryosuke Bo
- Department of Pediatrics, Shimane University Faculty of Medicine, 89-1, Ennya-cho, Izumo, Shimane 6938501, Japan; Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-1, Kusunokicho, Chuo, Kobe, Hyogo 6500017, Japan
| | - Yuki Hasegawa
- Department of Pediatrics, Shimane University Faculty of Medicine, 89-1, Ennya-cho, Izumo, Shimane 6938501, Japan
| | - Kenji Yamada
- Department of Pediatrics, Shimane University Faculty of Medicine, 89-1, Ennya-cho, Izumo, Shimane 6938501, Japan
| | - Hironori Kobayashi
- Department of Pediatrics, Shimane University Faculty of Medicine, 89-1, Ennya-cho, Izumo, Shimane 6938501, Japan
| | - Takeshi Taketani
- Department of Pediatrics, Shimane University Faculty of Medicine, 89-1, Ennya-cho, Izumo, Shimane 6938501, Japan
| | - Seiji Fukuda
- Department of Pediatrics, Shimane University Faculty of Medicine, 89-1, Ennya-cho, Izumo, Shimane 6938501, Japan
| | - Seiji Yamaguchi
- Department of Pediatrics, Shimane University Faculty of Medicine, 89-1, Ennya-cho, Izumo, Shimane 6938501, Japan
| |
Collapse
|
49
|
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
|
50
|
Sakai C, Yamaguchi S, Sasaki M, Miyamoto Y, Matsushima Y, Goto YI. ECHS1 mutations cause combined respiratory chain deficiency resulting in Leigh syndrome. Hum Mutat 2015; 36:232-9. [PMID: 25393721 DOI: 10.1002/humu.22730] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/05/2014] [Indexed: 12/31/2022]
Abstract
The human ECHS1 gene encodes the short-chain enoyl coenzyme A hydratase, the enzyme that catalyzes the second step of β-oxidation of fatty acids in the mitochondrial matrix. We report on a boy with ECHS1 deficiency who was diagnosed with Leigh syndrome at 21 months of age. The patient presented with hypotonia, metabolic acidosis, and developmental delay. A combined respiratory chain deficiency was also observed. Targeted exome sequencing of 776 mitochondria-associated genes encoded by nuclear DNA identified compound heterozygous mutations in ECHS1. ECHS1 protein expression was severely depleted in the patient's skeletal muscle and patient-derived myoblasts; a marked decrease in enzyme activity was also evident in patient-derived myoblasts. Immortalized patient-derived myoblasts that expressed exogenous wild-type ECHS1 exhibited the recovery of the ECHS1 activity, indicating that the gene defect was pathogenic. Mitochondrial respiratory complex activity was also mostly restored in these cells, suggesting that there was an unidentified link between deficiency of ECHS1 and respiratory chain. Here, we describe the patient with ECHS1 deficiency; these findings will advance our understanding not only the pathology of mitochondrial fatty acid β-oxidation disorders, but also the regulation of mitochondrial metabolism.
Collapse
Affiliation(s)
- Chika Sakai
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|