51
|
Malvi P, Janostiak R, Nagarajan A, Zhang X, Wajapeyee N. N-acylsphingosine amidohydrolase 1 promotes melanoma growth and metastasis by suppressing peroxisome biogenesis-induced ROS production. Mol Metab 2021; 48:101217. [PMID: 33766731 PMCID: PMC8081993 DOI: 10.1016/j.molmet.2021.101217] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/02/2021] [Accepted: 03/17/2021] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Metabolic deregulation is a key hallmark of cancer cells and has been shown to drive cancer growth and metastasis. However, not all metabolic drivers of melanoma are known. Based on our finding that N-acylsphingosine amidohydrolase 1 (ASAH1) is overexpressed in melanoma, the objective of these studies was to establish its role in melanoma tumor growth and metastasis, understand its mechanism of action, and evaluate ASAH1 targeting for melanoma therapy. METHODS We used publicly available melanoma datasets and patient-derived samples of melanoma and normal skin tissue and analyzed them for ASAH1 mRNA expression and ASAH1 protein expression using immunohistochemistry. ASAH1 was knocked down using short-hairpin RNAs in multiple melanoma cell lines that were tested in a series of cell culture-based assays and mouse-based melanoma xenograft assays to monitor the effect of ASAH1 knockdown on melanoma tumor growth and metastasis. An unbiased metabolomics analysis was performed to identify the mechanism of ASAH1 action. Based on the metabolomics findings, the role of peroxisome-mediated reactive oxygen species (ROS) production was explored in regard to mediating the effect of ASAH1. The ASAH1 inhibitor was used alone or in combination with a BRAFV600E inhibitor to evaluate the therapeutic value of ASAH1 targeting for melanoma therapy. RESULTS We determined that ASAH1 was overexpressed in a large percentage of melanoma cells and regulated by transcription factor E2F1 in a mitogen-activated protein (MAP) kinase pathway-dependent manner. ASAH1 expression was necessary to maintain melanoma tumor growth and metastatic attributes in cell cultures and mouse models of melanoma tumor growth and metastasis. To identify the mechanism by which ASAH1 facilitates melanoma tumor growth and metastasis, we performed a large-scale and unbiased metabolomics analysis of melanoma cells expressing ASAH1 short-hairpin RNAs (shRNAs). We found that ASAH1 inhibition increased peroxisome biogenesis through ceramide-mediated PPARγ activation. ASAH1 loss increased ceramide and peroxisome-derived ROS, which in turn inhibited melanoma growth. Pharmacological inhibition of ASAH1 also attenuated melanoma growth and enhanced the effectiveness of BRAF kinase inhibitor in the cell cultures and mice. CONCLUSIONS Collectively, these results demonstrate that ASAH1 is a druggable driver of melanoma tumor growth and metastasis that functions by suppressing peroxisome biogenesis, thereby inhibiting peroxisome-derived ROS production. These studies also highlight the therapeutic utility of ASAH1 inhibitors for melanoma therapy.
Collapse
Affiliation(s)
- Parmanand Malvi
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Alabama, 35233, USA
| | - Radoslav Janostiak
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06510, USA; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Arvindhan Nagarajan
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Xuchen Zhang
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Narendra Wajapeyee
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Alabama, 35233, USA.
| |
Collapse
|
52
|
Schühle K, Saft M, Vögeli B, Erb TJ, Heider J. Benzylmalonyl-CoA dehydrogenase, an enzyme involved in bacterial auxin degradation. Arch Microbiol 2021; 203:4149-4159. [PMID: 34059946 PMCID: PMC8360864 DOI: 10.1007/s00203-021-02406-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/21/2021] [Accepted: 05/23/2021] [Indexed: 11/28/2022]
Abstract
A novel acyl-CoA dehydrogenase involved in degradation of the auxin indoleacetate by Aromatoleum aromaticum was identified as a decarboxylating benzylmalonyl-CoA dehydrogenase (IaaF). It is encoded within the iaa operon coding for enzymes of indoleacetate catabolism. Using enzymatically produced benzylmalonyl-CoA, the reaction was characterized as simultaneous oxidation and decarboxylation of benzylmalonyl-CoA to cinnamoyl-CoA and CO2. Oxygen served as electron acceptor and was reduced to H2O2, whereas electron transfer flavoprotein or artificial dyes serving as electron acceptors for other acyl-CoA dehydrogenases were not used. The enzyme is homotetrameric, contains an FAD cofactor and is enantiospecific in benzylmalonyl-CoA turnover. It shows high catalytic efficiency and strong substrate inhibition with benzylmalonyl-CoA, but otherwise accepts only a few medium-chain alkylmalonyl-CoA compounds as alternative substrates with low activities. Its reactivity of oxidizing 2-carboxyacyl-CoA with simultaneous decarboxylation is unprecedented and indicates a modified reaction mechanism for acyl-CoA dehydrogenases, where elimination of the 2-carboxy group replaces proton abstraction from C2.
Collapse
Affiliation(s)
- Karola Schühle
- Laboratory for Microbial Biochemistry, Philipps University of Marburg, 35043, Marburg, Germany
| | - Martin Saft
- Laboratory for Microbial Biochemistry, Philipps University of Marburg, 35043, Marburg, Germany
| | - Bastian Vögeli
- Max Planck Institute for Terrestrial Microbiology, 35043, Marburg, Germany
| | - Tobias J Erb
- Max Planck Institute for Terrestrial Microbiology, 35043, Marburg, Germany.,LOEWE-Center for Synthetic Microbiology, Marburg, Germany
| | - Johann Heider
- Laboratory for Microbial Biochemistry, Philipps University of Marburg, 35043, Marburg, Germany. .,LOEWE-Center for Synthetic Microbiology, Marburg, Germany.
| |
Collapse
|
53
|
Pang B, Liao R, Tang Z, Guo S, Wu Z, Liu W. Caerulomycin and collismycin antibiotics share a trans-acting flavoprotein-dependent assembly line for 2,2'-bipyridine formation. Nat Commun 2021; 12:3124. [PMID: 34035275 PMCID: PMC8149447 DOI: 10.1038/s41467-021-23475-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 04/28/2021] [Indexed: 11/09/2022] Open
Abstract
Linear nonribosomal peptide synthetases (NRPSs) and polyketide synthases (PKSs) template the modular biosynthesis of numerous nonribosomal peptides, polyketides and their hybrids through assembly line chemistry. This chemistry can be complex and highly varied, and thus challenges our understanding in NRPS and PKS-programmed, diverse biosynthetic processes using amino acid and carboxylate building blocks. Here, we report that caerulomycin and collismycin peptide-polyketide hybrid antibiotics share an assembly line that involves unusual NRPS activity to engage a trans-acting flavoprotein in C-C bond formation and heterocyclization during 2,2'-bipyridine formation. Simultaneously, this assembly line provides dethiolated and thiolated 2,2'-bipyridine intermediates through differential treatment of the sulfhydryl group arising from L-cysteine incorporation. Subsequent L-leucine extension, which does not contribute any atoms to either caerulomycins or collismycins, plays a key role in sulfur fate determination by selectively advancing one of the two 2,2'-bipyridine intermediates down a path to the final products with or without sulfur decoration. These findings further the appreciation of assembly line chemistry and will facilitate the development of related molecules using synthetic biology approaches.
Collapse
Affiliation(s)
- Bo Pang
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Shanghai, China
| | - Rijing Liao
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhijun Tang
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Shanghai, China
| | - Shengjie Guo
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhuhua Wu
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Shanghai, China
| | - Wen Liu
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Shanghai, China. .,Huzhou Center of Bio-Synthetic Innovation, Huzhou, China.
| |
Collapse
|
54
|
Jeremias G, Jesus F, Ventura SPM, Gonçalves FJM, Asselman J, Pereira JL. New insights on the effects of ionic liquid structural changes at the gene expression level: Molecular mechanisms of toxicity in Daphnia magna. JOURNAL OF HAZARDOUS MATERIALS 2021; 409:124517. [PMID: 33199138 DOI: 10.1016/j.jhazmat.2020.124517] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/18/2020] [Accepted: 11/05/2020] [Indexed: 06/11/2023]
Abstract
Knowledge on the molecular basis of ionic liquids' (ILs) ecotoxicity is critical for the development of these designer solvents as their structure can be engineered to simultaneously meet functionality performance and environmental safety. The molecular effects of ILs were investigated by using RNA-sequencing following Daphnia magna exposure to imidazolium- and cholinium-based ILs: 1-ethyl-3-methylimidazolium chloride ([C2mim]Cl), 1-dodecyl-3-methylimidazolium chloride ([C12mim]Cl) and cholinium chloride ([Chol]Cl)-; the selection allowing to compare different families and cation alkyl chains. ILs shared mechanisms of toxicity focusing e.g. cellular membrane and cytoskeleton, oxidative stress, energy production, protein biosynthesis, DNA damage, disease initiation. [C2mim]Cl and [C12mim]Cl were the least and the most toxic ILs at the transcriptional level, denoting the role of the alkyl chain as a driver of ILs toxicity. Also, it was reinforced that [Chol]Cl is not devoid of environmental hazardous potential regardless of its argued biological compatibility. Unique gene expression signatures could also be identified for each IL, enlightening specific mechanisms of toxicity.
Collapse
Affiliation(s)
- Guilherme Jeremias
- Department of Biology & CESAM - Centre for Environmental and Marine Studies, University of Aveiro, Portugal
| | - Fátima Jesus
- Department of Biology & CESAM - Centre for Environmental and Marine Studies, University of Aveiro, Portugal
| | - Sónia P M Ventura
- Department of Chemistry & CICECO - Aveiro Institute of Materials, University of Aveiro, Portugal
| | - Fernando J M Gonçalves
- Department of Biology & CESAM - Centre for Environmental and Marine Studies, University of Aveiro, Portugal
| | - Jana Asselman
- Blue Growth Research Lab, Ghent University, Bluebridge Building, Ostend Science Park 1, 8400 Ostend, Belgium
| | - Joana L Pereira
- Department of Biology & CESAM - Centre for Environmental and Marine Studies, University of Aveiro, Portugal.
| |
Collapse
|
55
|
Ünsaldı E, Kurt-Kızıldoğan A, Özcan S, Becher D, Voigt B, Aktaş C, Özcengiz G. Proteomic analysis of a hom-disrupted, cephamycin C overproducing Streptomyces clavuligerus. Protein Pept Lett 2021; 28:205-220. [PMID: 32707026 DOI: 10.2174/0929866527666200723163655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/18/2020] [Accepted: 06/20/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Streptomyces clavuligerus is prolific producer of cephamycin C, a medically important antibiotic. In our former study, cephamycin C titer was 2-fold improved by disrupting homoserine dehydrogenase (hom) gene of aspartate pahway in Streptomyces clavuligerus NRRL 3585. OBJECTIVE In this article, we aimed to provide a comprehensive understanding at the proteome level on potential complex metabolic changes as a consequence of hom disruption in Streptomyces clavuligerus AK39. METHODS A comparative proteomics study was carried out between the wild type and its hom disrupted AK39 strain by 2 Dimensional Electrophoresis-Matrix Assisted Laser Desorption and Ionization Time-Of-Flight Mass Spectrometry (2DE MALDI-TOF/MS) and Nanoscale Liquid Chromatography- Tandem Mass Spectrometry (nanoLC-MS/MS) analyses. Clusters of Orthologous Groups (COG) database was used to determine the functional categories of the proteins. The theoretical pI and Mw values of the proteins were calculated using Expasy pI/Mw tool. RESULTS "Hypothetical/Unknown" and "Secondary Metabolism" were the most prominent categories of the differentially expressed proteins. Upto 8.7-fold increased level of the positive regulator CcaR was a key finding since CcaR was shown to bind to cefF promoter thereby direcly controlling its expression. Consistently, CeaS2, the first enzyme of CA biosynthetic pathway, was 3.3- fold elevated. There were also many underrepresented proteins associated with the biosynthesis of several Non-Ribosomal Peptide Synthases (NRPSs), clavams, hybrid NRPS/Polyketide synthases (PKSs) and tunicamycin. The most conspicuously underrepresented protein of amino acid metabolism was 4-Hydroxyphenylpyruvate dioxygenase (HppD) acting in tyrosine catabolism. The levels of a Two Component System (TCS) response regulator containing a CheY-like receiver domain and an HTH DNA-binding domain as well as DNA-binding protein HU were elevated while a TetR-family transcriptional regulator was underexpressed. CONCLUSION The results obtained herein will aid in finding out new targets for further improvement of cephamycin C production in Streptomyces clavuligerus.
Collapse
Affiliation(s)
- Eser Ünsaldı
- Department of Biological Sciences, Middle East Technical University, Ankara 06800, Turkey
| | | | - Servet Özcan
- Department of Biology, Erciyes University, Kayseri 38280, Turkey
| | - Dörte Becher
- Institute of Microbiology, Ernst- Moritz-Arndt-University of Greifswald, Greifswald D-17487, Germany
| | - Birgit Voigt
- Institute of Microbiology, Ernst- Moritz-Arndt-University of Greifswald, Greifswald D-17487, Germany
| | - Caner Aktaş
- Department of Biological Sciences, Middle East Technical University, Ankara 06800, Turkey
| | - Gülay Özcengiz
- Department of Biological Sciences, Middle East Technical University, Ankara 06800, Turkey
| |
Collapse
|
56
|
Liu XY, Chen XJ, Zhao M, Wang ZQ, Chen HZ, Li HF, Wang CJ, Wu SF, Peng C, Yin Y, Fu HX, Lin MT, Yu L, Xiong ZQ, Wu ZY, Wang N. CHIP control degradation of mutant ETF:QO through ubiquitylation in late-onset multiple acyl-CoA dehydrogenase deficiency. J Inherit Metab Dis 2021; 44:450-468. [PMID: 33438237 DOI: 10.1002/jimd.12361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 12/24/2020] [Accepted: 01/11/2021] [Indexed: 11/12/2022]
Abstract
Late-onset multiple acyl-CoA dehydrogenase deficiency (MADD) is the most common form of lipid storage myopathy. The disease is mainly caused by mutations in electron-transfer flavoprotein dehydrogenase gene (ETFDH), which leads to decreased levels of ETF:QO in skeletal muscle. However, the specific underlying mechanisms triggering such degradation remain unknown. We constructed expression plasmids containing wild type ETF:QO and mutants ETF:QO-A84T, R175H, A215T, Y333C, and cultured patient-derived fibroblasts containing the following mutations in ETFDH: c.250G>A (p.A84T), c.998A>G (p.Y333C), c.770A>G (p.Y257C), c.1254_1257delAACT (p. L418TfsX10), c.524G>A (p.R175H), c.380T>A (p.L127P), and c.892C>T (p.P298S). We used in vitro expression systems and patient-derived fibroblasts to detect stability of ETF:QO mutants then evaluated their interaction with Hsp70 interacting protein CHIP with active/inactive ubiquitin E3 ligase carboxyl terminus using western blot and immunofluorescence staining. This interaction was confirmed in vitro and in vivo by co-immunoprecipitation and immunofluorescence staining. We confirmed the existence two ubiquitination sites in mutant ETF:QO using mass spectrometry (MS) analysis. We found that mutant ETF:QO proteins were unstable and easily degraded in patient fibroblasts and in vitro expression systems by ubiquitin-proteasome pathway, and identified the specific ubiquitin E3 ligase as CHIP, which forms complex to control mutant ETF:QO degradation through poly-ubiquitination. CHIP-dependent degradation of mutant ETF:QO proteins was confirmed by MS and site-directed mutagenesis of ubiquitination sites. Hsp70 is directly involved in this process as molecular chaperone of CHIP. CHIP plays an important role in ubiquitin-proteasome pathway dependent degradation of mutant ETF:QO by working as a chaperone-assisted E3 ligase, which reveals CHIP's potential role in pathological mechanisms of late-onset MADD.
Collapse
Affiliation(s)
- Xin-Yi Liu
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xue-Jiao Chen
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Neurology, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Miao Zhao
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhi-Qiang Wang
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Hai-Zhu Chen
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Hong-Fu Li
- Department of Neurology and Research Center of Neurology in the Second Affiliated Hospital, and the Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chen-Ji Wang
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Shi-Fei Wu
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, China
| | - Yue Yin
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, China
| | - Hong-Xia Fu
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Min-Ting Lin
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Long Yu
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Zhi-Qi Xiong
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology in the Second Affiliated Hospital, and the Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ning Wang
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
57
|
Mereis M, Wanders RJA, Schoonen M, Dercksen M, Smuts I, van der Westhuizen FH. Disorders of flavin adenine dinucleotide metabolism: MADD and related deficiencies. Int J Biochem Cell Biol 2021; 132:105899. [PMID: 33279678 DOI: 10.1016/j.biocel.2020.105899] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022]
Abstract
Multiple acyl-coenzyme A dehydrogenase deficiency (MADD), or glutaric aciduria type II (GAII), is a group of clinically heterogeneous disorders caused by mutations in electron transfer flavoprotein (ETF) and ETF-ubiquinone oxidoreductase (ETFQO) - the two enzymes responsible for the re-oxidation of enzyme-bound flavin adenine dinucleotide (FADH2) via electron transfer to the respiratory chain at the level of coenzyme Q10. Over the past decade, an increasing body of evidence has further coupled mutations in FAD metabolism (including intercellular riboflavin transport, FAD biosynthesis and FAD transport) to MADD-like phenotypes. In this review we provide a detailed description of the overarching and specific metabolic pathways involved in MADD. We examine the eight associated genes (ETFA, ETFB, ETFDH, FLAD1, SLC25A32 and SLC52A1-3) and clinical phenotypes, and report ∼436 causative mutations following a systematic literature review. Finally, we focus attention on the value and shortcomings of current diagnostic approaches, as well as current and future therapeutic options for MADD and its phenotypic disorders.
Collapse
Affiliation(s)
- Michelle Mereis
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Ronald J A Wanders
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Maryke Schoonen
- Human Metabolomics, North-West University, Potchefstroom, South Africa; Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Marli Dercksen
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Izelle Smuts
- Department of Paediatrics, Steve Biko Academic Hospital, University of Pretoria, South Africa
| | | |
Collapse
|
58
|
Silverstein TP. How enzymes harness highly unfavorable proton transfer reactions. Protein Sci 2021; 30:735-744. [PMID: 33554401 DOI: 10.1002/pro.4037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 11/12/2022]
Abstract
Acid-base reactions that are exceedingly unfavorable under standard conditions can be catalytically important at enzyme active sites. For example, in triose phosphate isomerase, a glutamate side chain (nominal pKa ≈ 4 in solution) can in fact deprotonate a CH group that is vicinal to a carbonyl (pKa ≈ 18 in solution). This is true because of three distinct interactions: (a) ground state pKa shifts due to environment polarity and electrostatics; (b) dramatic increases in effective molarity due to optimization of proximity and orientation; and (c) transition state pKa shifts due to binding interactions and the formation of strong low barrier hydrogen bonds. In this report, we review the literature showing that the sum of these three effects supplies more than enough free energy to push forward proton transfer reactions that under standard conditions are exceedingly nonspontaneous and slow.
Collapse
|
59
|
Cai D, Li Y, Zhang K, Zhou B, Guo F, Holm L, Liu HY. Co-option of PPARα in the regulation of lipogenesis and fatty acid oxidation in CLA-induced hepatic steatosis. J Cell Physiol 2020; 236:4387-4402. [PMID: 33184849 DOI: 10.1002/jcp.30157] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/27/2020] [Accepted: 11/02/2020] [Indexed: 11/06/2022]
Abstract
Nonalcoholic-fatty-liver-disease (NAFLD) is the result of imbalances in hepatic lipid partitioning and is linked to dietary factors. We demonstrate that conjugated linoleic acid (CLA) when given to mice as a dietary supplement, induced an enlarged liver, hepatic steatosis, and increased plasma levels of fatty acid (FA), alanine transaminase, and triglycerides. The progression of NAFLD and insulin resistance was reversed by GW6471 a small-molecule antagonist of peroxisome proliferator-activated receptor α (PPARα). Transcriptional profiling of livers revealed that the genes involved in FA oxidation and lipogenesis as two core gene programs controlled by PPARα in response to CLA and GW6471 including Acaca and Acads. Bioinformatic analysis of PPARα ChIP-seq data set and ChIP-qPCR showed that GW6471 blocks PPARα binding to Acaca and Acads and abolishes the PPARα-mediated local histone modifications of H3K27ac and H3K4me1 in CLA-treated hepatocytes. Thus, our findings reveal a dual role of PPARα in the regulation of lipid homeostasis and highlight its druggable nature in NAFLD.
Collapse
Affiliation(s)
- Demin Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yanwei Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Kexin Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Bo Zhou
- Institute of Digestive Disease, Zhengzhou University, Zhengzhou, China
| | - Feilong Guo
- Department of General Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Lena Holm
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Hao-Yu Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China.,Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
60
|
Mezzina MP, Manoli MT, Prieto MA, Nikel PI. Engineering Native and Synthetic Pathways in Pseudomonas putida for the Production of Tailored Polyhydroxyalkanoates. Biotechnol J 2020; 16:e2000165. [PMID: 33085217 DOI: 10.1002/biot.202000165] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/16/2020] [Indexed: 12/16/2022]
Abstract
Growing environmental concern sparks renewed interest in the sustainable production of (bio)materials that can replace oil-derived goods. Polyhydroxyalkanoates (PHAs) are isotactic polymers that play a critical role in the central metabolism of producer bacteria, as they act as dynamic reservoirs of carbon and reducing equivalents. PHAs continue to attract industrial attention as a starting point toward renewable, biodegradable, biocompatible, and versatile thermoplastic and elastomeric materials. Pseudomonas species have been known for long as efficient biopolymer producers, especially for medium-chain-length PHAs. The surge of synthetic biology and metabolic engineering approaches in recent years offers the possibility of exploiting the untapped potential of Pseudomonas cell factories for the production of tailored PHAs. In this article, an overview of the metabolic and regulatory circuits that rule PHA accumulation in Pseudomonas putida is provided, and approaches leading to the biosynthesis of novel polymers (e.g., PHAs including nonbiological chemical elements in their structures) are discussed. The potential of novel PHAs to disrupt existing and future market segments is closer to realization than ever before. The review is concluded by pinpointing challenges that currently hinder the wide adoption of bio-based PHAs, and strategies toward programmable polymer biosynthesis from alternative substrates in engineered P. putida strains are proposed.
Collapse
Affiliation(s)
- Mariela P Mezzina
- Systems Environmental Microbiology Group, The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs Lyngby, 2800, Denmark
| | - María Tsampika Manoli
- Microbial and Plant Biotechnology Department, Centro de Investigaciones Biológicas «Margarita Salas» (CIB-CSIC), Polymer Biotechnology Group, Madrid, 28040, Spain.,Spanish National Research Council (SusPlast-CSIC), Interdisciplinary Platform for Sustainable Plastics Toward a Circular Economy, Madrid, 28040, Spain
| | - M Auxiliadora Prieto
- Microbial and Plant Biotechnology Department, Centro de Investigaciones Biológicas «Margarita Salas» (CIB-CSIC), Polymer Biotechnology Group, Madrid, 28040, Spain.,Spanish National Research Council (SusPlast-CSIC), Interdisciplinary Platform for Sustainable Plastics Toward a Circular Economy, Madrid, 28040, Spain
| | - Pablo I Nikel
- Systems Environmental Microbiology Group, The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs Lyngby, 2800, Denmark
| |
Collapse
|
61
|
Wang Y, Ehsan M, Wang S, Tian X, Yan R, Song X, Xu L, Li X. Modulatory functions of recombinant electron transfer flavoprotein α subunit protein from Haemonchus contortus on goat immune cells in vitro. Vet Parasitol 2020; 288:109300. [PMID: 33152677 DOI: 10.1016/j.vetpar.2020.109300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/25/2020] [Accepted: 10/26/2020] [Indexed: 01/13/2023]
Abstract
Suppression and modulation of the host immune response to parasitic nematodes have been extensively studied. In the present study, we cloned and produced recombinant electron transfer flavoprotein α subunit (ETFα) protein from Haemonchus contortus (rHCETFα), a parasitic nematode of small ruminants, and studied the effect of this protein on modulating the immune response of goat peripheral blood mononuclear cells (PBMCs) and monocytes. Immunohistochemical tests verified that the HCETFα protein was localized mainly in the intestinal wall and on the body surface of worms. Immunoblot analysis revealed that rHCETFα was recognized by the serum of goats artificially infected with H. contortus. Immunofluorescence analysis indicated that rHCETFα bound to the surface of PBMCs. rHCETFα was co-incubated with goat PBMCs to observe the immunomodulatory effects exerted by HCETFα on proliferation, apoptosis, cytokine secretion and nitric oxide (NO) production. The results showed that rHCETFα suppressed the proliferation of goat PBMCs stimulated by concanavalin A and induced apoptosis in goat PBMCs. After rHCETFα exposure, IL-2, IL-4, IL-17A and TNF-α expression was markedly reduced, whereas secretion of TGF-β1 was significantly elevated, in goat PBMCs. Moreover, rHCETFα up-regulated NO production in a dose-dependent manner. FITC-dextran internalization assays showed that rHCETFα inhibited phagocytosis of goat monocytes. These results elucidate the interaction between parasites and hosts at the molecular level, suggest a possible immunomodulatory target and contribute to the search for innovative proteins that may be candidate targets for drugs and vaccines.
Collapse
Affiliation(s)
- Yujian Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; School of Life Science, Huizhou University, Huizhou 516007, PR China.
| | - Muhammad Ehsan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - Shuai Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - Xiaowei Tian
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - RuoFeng Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - XiaoKai Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - LiXin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - XiangRui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
62
|
Boldarine VT, Pedroso AP, Brandão-Teles C, LoTurco EG, Nascimento CMO, Oyama LM, Bueno AA, Martins-de-Souza D, Ribeiro EB. Ovariectomy modifies lipid metabolism of retroperitoneal white fat in rats: a proteomic approach. Am J Physiol Endocrinol Metab 2020; 319:E427-E437. [PMID: 32663100 DOI: 10.1152/ajpendo.00094.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Menopause is often accompanied by visceral obesity. With the aim of exploring the consequences of ovarian failure on visceral fat, we evaluated the effects of ovariectomy and estrogen replacement on the proteome/phosphoproteome and on the fatty acid profile of the retroperitoneal adipose depot (RAT) of rats. Eighteen 3-mo-old female Wistar rats were either ovariectomized or sham operated and fed with standard chow for 3 mo. A subgroup of ovariectomized rats received estradiol replacement. RAT samples were analyzed with data-independent acquisitions LC-MS/MS, and pathway analysis was performed with the differentially expressed/phosphorylated proteins. RAT lipid profile was analyzed by gas chromatography. Ovariectomy induced high adiposity and insulin resistance and promoted alterations in protein expression and phosphorylation. Pathway analysis showed that five pathways were significantly affected by ovariectomy, namely, metabolism of lipids (including fatty acid metabolism and mitochondrial fatty acid β-oxidation), fatty acyl-CoA biosynthesis, innate immune system (including neutrophil degranulation), metabolism of vitamins and cofactors, and integration of energy metabolism (including ChREBP activates metabolic gene expression). Lipid profile analysis showed increased palmitic and palmitoleic acid content. The analysis of the data indicated that ovariectomy favored lipogenesis whereas it impaired fatty acid oxidation and induced a proinflammatory state in the visceral adipose tissue. These effects are consistent with the findings of high adiposity, hyperleptinemia, and impaired insulin sensitivity. The observed alterations were partially attenuated by estradiol replacement. The data point to a role of disrupted lipid metabolism in adipose tissue in the genesis of obesity after menopause.
Collapse
Affiliation(s)
- Valter T Boldarine
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Amanda P Pedroso
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Caroline Brandão-Teles
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Edson G LoTurco
- Divisão de Urologia e Reprodução Humana, Departamento de Cirurgia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Cláudia M O Nascimento
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Lila M Oyama
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Allain A Bueno
- Department of Biological Sciences, College of Health, Life and Environmental Sciences, University of Worcester, Worcester, United Kingdom
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
- Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION) Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, Brazil
- D'Or Institute for Research and Education (IDOR), São Paulo, Brazil
| | - Eliane B Ribeiro
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
63
|
Flavin adenine dinucleotide ameliorates hypertensive vascular remodeling via activating short chain acyl-CoA dehydrogenase. Life Sci 2020; 258:118156. [PMID: 32735886 DOI: 10.1016/j.lfs.2020.118156] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/07/2020] [Accepted: 07/23/2020] [Indexed: 11/23/2022]
Abstract
AIMS Flavin adenine dinucleotide (FAD), participates in fatty acid β oxidation as a cofactor, which has been confirmed to enhance SCAD activity and expression. However, the role of FAD on hypertensive vascular remodeling is unclear. In this study, we investigated the underlying mechanisms of FAD on vascular remodeling and endothelial homeostasis. MAIN METHODS Morphological examination of vascular remodeling were analyzed with hematoxylin and eosin (HE) staining, Verhoeff's Van Gieson (EVG) staing, Dihydroethidium (DHE) staining and Sirius red staining. HUVECs apoptotic rate was detected by flow cytometry and HUVECs reactive oxygen species (ROS) was detected by DHE-probe. Enzymatic reactions were used to detect SCAD enzyme activity. The protein level was detected by Western Blots, the mRNA level was detected by quantitative real-time PCR. KEY FINDINGS In vivo experiments, FAD significantly decreased blood pressure and ameliorated vascular remodeling by increasing SCAD expression, Nitric Oxide (NO) production and reducing ROS production. In vitro experiments, FAD protected against the tBHP induced injury in HUVEC, by increasing the activity of SCAD, increasing the elimination of free fatty acid (FFA), scavenging ROS, reducing apoptotic rate, thereby improving endothelial cell function. SIGNIFICANCE FAD has a new possibility for preventing and treating hypertensive vascular remodeling.
Collapse
|
64
|
Chen H, Liu L, Huang T, Chen J, Chen T. Direct Dehydrogenation for the Synthesis of α,β‐Unsaturated Carbonyl Compounds. Adv Synth Catal 2020. [DOI: 10.1002/adsc.202000454] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Hong Chen
- Haikou Hospital affiliated to Xiangya School of MedicineCentral South University Haikou 570100 People's Republic of China
| | - Long Liu
- Key Laboratory of Ministry of Education for Advanced Materials in Tropical Island ResourcesHainan Provincial Key Lab of Fine ChemHainan University Haikou 570228 People's Republic of China
| | - Tianzeng Huang
- Key Laboratory of Ministry of Education for Advanced Materials in Tropical Island ResourcesHainan Provincial Key Lab of Fine ChemHainan University Haikou 570228 People's Republic of China
| | - Jing Chen
- Haikou Hospital affiliated to Xiangya School of MedicineCentral South University Haikou 570100 People's Republic of China
| | - Tieqiao Chen
- Key Laboratory of Ministry of Education for Advanced Materials in Tropical Island ResourcesHainan Provincial Key Lab of Fine ChemHainan University Haikou 570228 People's Republic of China
| |
Collapse
|
65
|
Gao X, Xue H, Luo J, Ji J, Zhang L, Niu L, Zhu X, Wang L, Zhang S, Cui J. Molecular Evidence that Lysiphlebia japonica Regulates the Development and Physiological Metabolism of Aphis gossypii. Int J Mol Sci 2020; 21:ijms21134610. [PMID: 32610524 PMCID: PMC7370083 DOI: 10.3390/ijms21134610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
Lysiphlebia japonica Ashmead (Hymenoptera, Braconidae) is an endophagous parasitoid and Aphis gossypii Glover (Hemiptera, Aphididae) is a major pest in cotton. The relationship between insect host-parasitoids and their hosts involves complex physiological, biochemical and genetic interactions. This study examines changes in the development and physiological metabolism of A. gossypii regulated by L. japonica. Our results demonstrated that both the body length and width increased compared to non-parasitized aphids. We detected significantly increases in the developmental period as well as severe reproductive castration following parasitization by L. japonica. We then used proteomics to characterize these biological changes, and when combined with transcriptomes, this analysis demonstrated that the differential expression of mRNA (up or downregulation) captured a maximum of 48.7% of the variations of protein expression. We assigned these proteins to functional categories that included immunity, energy metabolism and transport, lipid metabolism, and reproduction. We then verified the contents of glycogen and 6-phosphate glucose, which demonstrated that these important energy sources were significantly altered following parasitization. These results uncover the effects on A. gossypii following parasitization by L. japonica, additional insight into the mechanisms behind insect-insect parasitism, and a better understanding of host-parasite interactions.
Collapse
Affiliation(s)
- Xueke Gao
- Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang 455000, China; (X.G.); (J.L.); (J.J.); (L.Z.); (L.N.); (X.Z.); (L.W.)
- Zhengzhou Reseach Base, State Key Laboratory of Cotton Biology, Zhengzhou University, Zhengzhou 4550001, China
| | - Hui Xue
- College of Plant Science and Technology, Huazhong Agricultural University, Wuhan 430070, China;
| | - Junyu Luo
- Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang 455000, China; (X.G.); (J.L.); (J.J.); (L.Z.); (L.N.); (X.Z.); (L.W.)
- Zhengzhou Reseach Base, State Key Laboratory of Cotton Biology, Zhengzhou University, Zhengzhou 4550001, China
| | - Jichao Ji
- Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang 455000, China; (X.G.); (J.L.); (J.J.); (L.Z.); (L.N.); (X.Z.); (L.W.)
- Zhengzhou Reseach Base, State Key Laboratory of Cotton Biology, Zhengzhou University, Zhengzhou 4550001, China
| | - Lijuan Zhang
- Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang 455000, China; (X.G.); (J.L.); (J.J.); (L.Z.); (L.N.); (X.Z.); (L.W.)
- Zhengzhou Reseach Base, State Key Laboratory of Cotton Biology, Zhengzhou University, Zhengzhou 4550001, China
| | - Lin Niu
- Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang 455000, China; (X.G.); (J.L.); (J.J.); (L.Z.); (L.N.); (X.Z.); (L.W.)
- Zhengzhou Reseach Base, State Key Laboratory of Cotton Biology, Zhengzhou University, Zhengzhou 4550001, China
| | - Xiangzhen Zhu
- Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang 455000, China; (X.G.); (J.L.); (J.J.); (L.Z.); (L.N.); (X.Z.); (L.W.)
- Zhengzhou Reseach Base, State Key Laboratory of Cotton Biology, Zhengzhou University, Zhengzhou 4550001, China
| | - Li Wang
- Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang 455000, China; (X.G.); (J.L.); (J.J.); (L.Z.); (L.N.); (X.Z.); (L.W.)
- Zhengzhou Reseach Base, State Key Laboratory of Cotton Biology, Zhengzhou University, Zhengzhou 4550001, China
| | - Shuai Zhang
- Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang 455000, China; (X.G.); (J.L.); (J.J.); (L.Z.); (L.N.); (X.Z.); (L.W.)
- Zhengzhou Reseach Base, State Key Laboratory of Cotton Biology, Zhengzhou University, Zhengzhou 4550001, China
- Correspondence: (S.Z.); (J.C.)
| | - Jinjie Cui
- Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang 455000, China; (X.G.); (J.L.); (J.J.); (L.Z.); (L.N.); (X.Z.); (L.W.)
- Zhengzhou Reseach Base, State Key Laboratory of Cotton Biology, Zhengzhou University, Zhengzhou 4550001, China
- Correspondence: (S.Z.); (J.C.)
| |
Collapse
|
66
|
Liu J, Yu M, Chatnaparat T, Lee JH, Tian Y, Hu B, Zhao Y. Comparative transcriptomic analysis of global gene expression mediated by (p) ppGpp reveals common regulatory networks in Pseudomonas syringae. BMC Genomics 2020; 21:296. [PMID: 32272893 PMCID: PMC7146990 DOI: 10.1186/s12864-020-6701-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/25/2020] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Pseudomonas syringae is an important plant pathogen, which could adapt many different environmental conditions. Under the nutrient-limited and other stress conditions, P. syringae produces nucleotide signal molecules, i.e., guanosine tetra/pentaphosphate ((p)ppGpp), to globally regulate gene expression. Previous studies showed that (p) ppGpp played an important role in regulating virulence factors in P. syringae pv. tomato DC3000 (PstDC3000) and P. syringae pv. syringae B728a (PssB728a). Here we present a comparative transcriptomic analysis to uncover the overall effects of (p)ppGpp-mediated stringent response in P. syringae. RESULTS In this study, we investigated global gene expression profiles of PstDC3000 and PssB728a and their corresponding (p)ppGpp0 mutants in hrp-inducing minimal medium (HMM) using RNA-seq. A total of 1886 and 1562 differentially expressed genes (DEGs) were uncovered between the (p)ppGpp0 mutants and the wild-type in PstDC3000 and PssB728a, respectively. Comparative transcriptomics identified 1613 common DEGs, as well as 444 and 293 unique DEGs in PstDC3000 and PssB728a, respectively. Functional cluster analysis revealed that (p) ppGpp positively regulated a variety of virulence-associated genes, including type III secretion system (T3SS), type VI secretion system (T6SS), cell motility, cell division, and alginate biosynthesis, while negatively regulated multiple basic physiological processes, including DNA replication, RNA processes, nucleotide biosynthesis, fatty acid metabolism, ribosome protein biosynthesis, and amino acid metabolism in both PstDC3000 and PssB728a. Furthermore, (p) ppGpp had divergent effects on other processes in PstDC3000 and PssB728a, including phytotoxin, nitrogen regulation and general secretion pathway (GSP). CONCLUSION In this study, comparative transcriptomic analysis reveals common regulatory networks in both PstDC3000 and PssB728a mediated by (p) ppGpp in HMM. In both P. syringae systems, (p) ppGpp re-allocate cellular resources by suppressing multiple basic physiological activities and enhancing virulence gene expression, suggesting a balance between growth, survival and virulence. Our research is important in that due to similar global gene expression mediated by (p) ppGpp in both PstDC3000 and PssB728a, it is reasonable to propose that (p) ppGpp could be used as a target to develop novel control measures to fight against important plant bacterial diseases.
Collapse
Affiliation(s)
- Jun Liu
- College of Plant Protection, Key Laboratory of Integrated Management of Crop Diseases and Pests, Nanjing Agricultural University, Nanjing, 210095, P. R. China.,Department of Crop Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA
| | - Menghao Yu
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA
| | - Tiyakhon Chatnaparat
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA
| | - Jae Hoon Lee
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA
| | - Yanli Tian
- College of Plant Protection, Key Laboratory of Integrated Management of Crop Diseases and Pests, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - Baishi Hu
- College of Plant Protection, Key Laboratory of Integrated Management of Crop Diseases and Pests, Nanjing Agricultural University, Nanjing, 210095, P. R. China.
| | - Youfu Zhao
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, IL, 61801, USA.
| |
Collapse
|
67
|
Gadbery JE, Round JW, Yuan T, Wipperman MF, Story KT, Crowe AM, Casabon I, Liu J, Yang X, Eltis LD, Sampson NS. IpdE1-IpdE2 Is a Heterotetrameric Acyl Coenzyme A Dehydrogenase That Is Widely Distributed in Steroid-Degrading Bacteria. Biochemistry 2020; 59:1113-1123. [PMID: 32101684 PMCID: PMC7081610 DOI: 10.1021/acs.biochem.0c00005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Steroid-degrading bacteria, including Mycobacterium tuberculosis (Mtb), utilize an architecturally distinct subfamily of acyl coenzyme A dehydrogenases (ACADs) for steroid catabolism. These ACADs are α2β2 heterotetramers that are usually encoded by adjacent fadE-like genes. In mycobacteria, ipdE1 and ipdE2 (formerly fadE30 and fadE33) occur in divergently transcribed operons associated with the catabolism of 3aα-H-4α(3'-propanoate)-7aβ-methylhexahydro-1,5-indanedione (HIP), a steroid metabolite. In Mycobacterium smegmatis, ΔipdE1 and ΔipdE2 mutants had similar phenotypes, showing impaired growth on cholesterol and accumulating 5-OH HIP in the culture supernatant. Bioinformatic analyses revealed that IpdE1 and IpdE2 share many of the features of the α- and β-subunits, respectively, of heterotetrameric ACADs that are encoded by adjacent genes in many steroid-degrading proteobacteria. When coproduced in a rhodococcal strain, IpdE1 and IpdE2 of Mtb formed a complex that catalyzed the dehydrogenation of 5OH-HIP coenzyme A (5OH-HIP-CoA) to 5OH-3aα-H-4α(3'-prop-1-enoate)-7aβ-methylhexa-hydro-1,5-indanedione coenzyme A ((E)-5OH-HIPE-CoA). This corresponds to the initial step in the pathway that leads to degradation of steroid C and D rings via β-oxidation. Small-angle X-ray scattering revealed that the IpdE1-IpdE2 complex was an α2β2 heterotetramer typical of other ACADs involved in steroid catabolism. These results provide insight into an important class of steroid catabolic enzymes and a potential virulence determinant in Mtb.
Collapse
Affiliation(s)
- John E Gadbery
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - James W Round
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Tianao Yuan
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - Matthew F Wipperman
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States.,Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States.,Clinical & Translational Science Center, Weill Cornell Medicine, New York, New York 10065, United States
| | - Keith T Story
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Adam M Crowe
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Israel Casabon
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Jie Liu
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Xinxin Yang
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - Lindsay D Eltis
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.,Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Nicole S Sampson
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
| |
Collapse
|
68
|
Zhou B, Ma Y, Tian Y, Li J, Zhong H. Quantitative Proteomics Analysis by Sequential Window Acquisition of All Theoretical Mass Spectra-Mass Spectrometry Reveals Inhibition Mechanism of Pigments and Citrinin Production of Monascus Response to High Ammonium Chloride Concentration. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:808-817. [PMID: 31870144 DOI: 10.1021/acs.jafc.9b05852] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Various Monascus bioactive metabolites used as food or food additives in Asia for centuries are subjected to constant physical and chemical changes and different Monascus genus. With the aim to identify enzymes that participate in or indirectly regulate the pigments and citrinin biosynthesis pathways of Monascus purpureus cultured under high ammonium chloride, the changes of the proteome profile were examined using sequential window acquisition of all theoretical mass spectra-mass spectrometry-based quantitative proteomics approach in combination with bioinformatics analysis. A total of 292 proteins were confidently detected and quantified in each sample, including 163 that increased and 129 that decreased (t-tests, p ≤ 0.05). Pathway analysis indicated that high ammonium chloride in the present study accelerates the carbon substrate utilization and promotes the activity of key enzymes in glycolysis and β-oxidation of fatty acid catabolism to generate sufficient acetyl-CoA. However, the synthesis of the monascus pigments and citrinin was not enhanced because of inhibition of the polyketide synthase activity. All results demonstrated that the cause of initiation of pigments and citrinin synthesis is mainly due to the apparent inhibition of acyl and acetyl transfer by some acyltransferase and acetyltransferase, likely malony-CoA:ACP transacylase.
Collapse
Affiliation(s)
- Bo Zhou
- School of Food Science and Engineering , Central South University of Forestry and Technology , Changsha 410004 , P. R China
- Hunan Province Key Laboratory of Edible Forestry Resources Safety and Processing Utilization , Changsha 410004 , P. R China
- Hunan Key Laboratory of Processed Food for Special Medical Purpose , Changsha 410004 , China
| | - Yifan Ma
- School of Food Science and Engineering , Central South University of Forestry and Technology , Changsha 410004 , P. R China
- Hunan Province Key Laboratory of Edible Forestry Resources Safety and Processing Utilization , Changsha 410004 , P. R China
| | - Yuan Tian
- School of Food Science and Engineering , Central South University of Forestry and Technology , Changsha 410004 , P. R China
- Hunan Province Key Laboratory of Edible Forestry Resources Safety and Processing Utilization , Changsha 410004 , P. R China
| | - Jingbo Li
- Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Haiyan Zhong
- School of Food Science and Engineering , Central South University of Forestry and Technology , Changsha 410004 , P. R China
- Hunan Province Key Laboratory of Edible Forestry Resources Safety and Processing Utilization , Changsha 410004 , P. R China
| |
Collapse
|
69
|
Williams AN, Stavrinides J. Pantoea Natural Product 3 is encoded by an eight-gene biosynthetic gene cluster and exhibits antimicrobial activity against multi-drug resistant Acinetobacter baumannii and Pseudomonas aeruginosa. Microbiol Res 2020; 234:126412. [PMID: 32062363 DOI: 10.1016/j.micres.2020.126412] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/22/2019] [Accepted: 01/10/2020] [Indexed: 01/08/2023]
Abstract
Multi-drug resistant Acinetobacter baumannii and Pseudomonas aeruginosa continue to pose a serious health threat worldwide. Two Pantoea agglomerans strains, 3581 and SN01080, produce an antibiotic effective against these pathogens. To identify the antibiotic biosynthetic gene clusters, independent genetic screens were conducted for each strain using a mini-Tn5 transposon, which resulted in the identification of the same conserved eight-gene cluster. We have named this antibiotic Pantoea Natural Product 3 (PNP-3). The PNP-3 biosynthetic cluster is composed of genes encoding two Major Facilitator Superfamily (MFS) transporters, an ArsR family regulator, and five predicted enzymes. The biosynthetic gene cluster is found in only a few Pantoea strains and is not present within the antiSMASH and BAGEL4 databases, suggesting it may be novel. In strain 3581, PNP-3 production is linked to pantocin A production, where loss of pantocin A production results in a larger PNP-3 zone of inhibition. To evaluate the spectrum of activity, PNP-3 producers, including several PNP-3 mutants and pantocin A site-directed mutants, were tested against a collection of clinical, drug-resistant strains of A. baumannii and P. aeruginosa, as well as, Klebsiella, Escherichia coli, Enterobacter, Staphylococcus aureus, and Streptococcus mutans. PNP-3 was found to be effective against all strains except vancomycin-resistant Enterococcus under the tested conditions. Heterologous expression of the four predicted biosynthetic genes in Erwinia amylovora resulted in antibiotic production, providing a means for future overexpression and purification. PNP-3 is a natural product that is effective against drug-resistant A. baumannii, P. aeruginosa, and enteric species for which there are currently few treatment options.
Collapse
Affiliation(s)
- Ashley N Williams
- Department of Biology, University of Regina, 3737 Wascana Parkway, Regina, Saskatchewan, S4S0A2, Canada
| | - John Stavrinides
- Department of Biology, University of Regina, 3737 Wascana Parkway, Regina, Saskatchewan, S4S0A2, Canada.
| |
Collapse
|
70
|
Blake-Hedges JM, Pereira JH, Cruz-Morales P, Thompson MG, Barajas JF, Chen J, Krishna RN, Chan LJG, Nimlos D, Alonso-Martinez C, Baidoo EEK, Chen Y, Gin JW, Katz L, Petzold CJ, Adams PD, Keasling JD. Structural Mechanism of Regioselectivity in an Unusual Bacterial Acyl-CoA Dehydrogenase. J Am Chem Soc 2020; 142:835-846. [PMID: 31793780 DOI: 10.1021/jacs.9b09187] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Terminal alkenes are easily derivatized, making them desirable functional group targets for polyketide synthase (PKS) engineering. However, they are rarely encountered in natural PKS systems. One mechanism for terminal alkene formation in PKSs is through the activity of an acyl-CoA dehydrogenase (ACAD). Herein, we use biochemical and structural analysis to understand the mechanism of terminal alkene formation catalyzed by an γ,δ-ACAD from the biosynthesis of the polyketide natural product FK506, TcsD. While TcsD is homologous to canonical α,β-ACADs, it acts regioselectively at the γ,δ-position and only on α,β-unsaturated substrates. Furthermore, this regioselectivity is controlled by a combination of bulky residues in the active site and a lateral shift in the positioning of the FAD cofactor within the enzyme. Substrate modeling suggests that TcsD utilizes a novel set of hydrogen bond donors for substrate activation and positioning, preventing dehydrogenation at the α,β position of substrates. From the structural and biochemical characterization of TcsD, key residues that contribute to regioselectivity and are unique to the protein family were determined and used to identify other putative γ,δ-ACADs that belong to diverse natural product biosynthetic gene clusters. These predictions are supported by the demonstration that a phylogenetically distant homologue of TcsD also regioselectively oxidizes α,β-unsaturated substrates. This work exemplifies a powerful approach to understand unique enzymatic reactions and will facilitate future enzyme discovery, inform enzyme engineering, and aid natural product characterization efforts.
Collapse
Affiliation(s)
- Jacquelyn M Blake-Hedges
- Department of Chemistry , University of California , Berkeley , California 94720 , United States.,Joint BioEnergy Institute , Emeryville , California 94608 , United States.,Biological Systems and Engineering Division , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States
| | - Jose Henrique Pereira
- Joint BioEnergy Institute , Emeryville , California 94608 , United States.,Molecular Biophysics and Integrated Bioimaging , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States
| | - Pablo Cruz-Morales
- Joint BioEnergy Institute , Emeryville , California 94608 , United States.,Biological Systems and Engineering Division , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States
| | - Mitchell G Thompson
- Joint BioEnergy Institute , Emeryville , California 94608 , United States.,Biological Systems and Engineering Division , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States.,Department of Plant and Microbial Biology , University of California-Berkeley , Berkeley , California 94720 , United States
| | - Jesus F Barajas
- Joint BioEnergy Institute , Emeryville , California 94608 , United States.,Biological Systems and Engineering Division , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States.,Department of Plant and Microbial Biology , University of California-Berkeley , Berkeley , California 94720 , United States
| | - Jeffrey Chen
- Department of Chemistry , University of California , Berkeley , California 94720 , United States
| | - Rohith N Krishna
- Department of Chemistry , University of California , Berkeley , California 94720 , United States
| | - Leanne Jade G Chan
- Joint BioEnergy Institute , Emeryville , California 94608 , United States.,Biological Systems and Engineering Division , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States
| | - Danika Nimlos
- Department of Chemistry , University of California , Berkeley , California 94720 , United States
| | - Catalina Alonso-Martinez
- Department of Chemistry , University of California , Berkeley , California 94720 , United States
| | - Edward E K Baidoo
- Joint BioEnergy Institute , Emeryville , California 94608 , United States.,Biological Systems and Engineering Division , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States
| | - Yan Chen
- Joint BioEnergy Institute , Emeryville , California 94608 , United States.,Biological Systems and Engineering Division , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States.,Department of Energy Agile BioFoundry , Emeryville , California 94608 , United States
| | - Jennifer W Gin
- Joint BioEnergy Institute , Emeryville , California 94608 , United States.,Biological Systems and Engineering Division , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States.,Department of Energy Agile BioFoundry , Emeryville , California 94608 , United States
| | - Leonard Katz
- Joint BioEnergy Institute , Emeryville , California 94608 , United States.,QB3 Institute , University of California-Berkeley , Emeryville , California 94608 , United States
| | - Christopher J Petzold
- Joint BioEnergy Institute , Emeryville , California 94608 , United States.,Biological Systems and Engineering Division , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States.,Department of Energy Agile BioFoundry , Emeryville , California 94608 , United States
| | - Paul D Adams
- Joint BioEnergy Institute , Emeryville , California 94608 , United States.,Biological Systems and Engineering Division , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States.,Molecular Biophysics and Integrated Bioimaging , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States
| | - Jay D Keasling
- Joint BioEnergy Institute , Emeryville , California 94608 , United States.,Biological Systems and Engineering Division , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States.,QB3 Institute , University of California-Berkeley , Emeryville , California 94608 , United States.,Department of Chemical & Biomolecular Engineering, Department of Bioengineering , University of California-Berkeley , Berkeley , California 94720 , United States.,Novo Nordisk Foundation Center for Biosustainability , Technical University Denmark , DK2970 Horsholm , Denmark.,Center for Synthetic Biochemistry , Shenzhen Institutes for Advanced Technologies , Shenzhen 518055 , P. R. China
| |
Collapse
|
71
|
Lepore R, Kryshtafovych A, Alahuhta M, Veraszto HA, Bomble YJ, Bufton JC, Bullock AN, Caba C, Cao H, Davies OR, Desfosses A, Dunne M, Fidelis K, Goulding CW, Gurusaran M, Gutsche I, Harding CJ, Hartmann MD, Hayes CS, Joachimiak A, Leiman PG, Loppnau P, Lovering AL, Lunin VV, Michalska K, Mir-Sanchis I, Mitra AK, Moult J, Phillips GN, Pinkas DM, Rice PA, Tong Y, Topf M, Walton JD, Schwede T. Target highlights in CASP13: Experimental target structures through the eyes of their authors. Proteins 2019; 87:1037-1057. [PMID: 31442339 PMCID: PMC6851490 DOI: 10.1002/prot.25805] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/09/2019] [Accepted: 08/19/2019] [Indexed: 01/10/2023]
Abstract
The functional and biological significance of selected CASP13 targets are described by the authors of the structures. The structural biologists discuss the most interesting structural features of the target proteins and assess whether these features were correctly reproduced in the predictions submitted to the CASP13 experiment.
Collapse
Affiliation(s)
- Rosalba Lepore
- BSC-CNS Barcelona Supercomputing Center, Barcelona, Spain
| | | | - Markus Alahuhta
- Biosciences Center, National Renewable Energy Laboratory, Golden, Colorado
| | - Harshul A Veraszto
- Department of Protein Evolution, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Yannick J Bomble
- Biosciences Center, National Renewable Energy Laboratory, Golden, Colorado
| | - Joshua C Bufton
- Nuffield Department of Medicine; Structural Genomics Consortium, University of Oxford, Oxford, UK.,School of Biochemistry, University of Bristol, Bristol, UK
| | - Alex N Bullock
- Nuffield Department of Medicine; Structural Genomics Consortium, University of Oxford, Oxford, UK
| | - Cody Caba
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Hongnan Cao
- Department of BioSciences, Rice University, Houston, Texas.,Great Lakes Bioenergy Research Center, University of Wisconsin, Madison, Wisconsin
| | - Owen R Davies
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Ambroise Desfosses
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Matthew Dunne
- Institute of Food, Nutrition and Health, Zurich, Switzerland
| | | | - Celia W Goulding
- Department of Molecular Biology and Biochemistry; Pharmaceutical Sciences, University of California Irvine, Irvine, California
| | - Manickam Gurusaran
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Irina Gutsche
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS, Grenoble, France
| | | | - Marcus D Hartmann
- Department of Protein Evolution, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Christopher S Hayes
- Department of Molecular, Cellular and Developmental Biology, Biomolecular Science and Engineering Program, University of California, Santa Barbara, California
| | - Andrzej Joachimiak
- Structural Biology Center, Biosciences Division, Midwest Center for Structural Genomics, Argonne.,Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois
| | - Petr G Leiman
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas
| | - Peter Loppnau
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada
| | | | - Vladimir V Lunin
- Biosciences Center, National Renewable Energy Laboratory, Golden, Colorado
| | - Karolina Michalska
- Structural Biology Center, Biosciences Division, Midwest Center for Structural Genomics, Argonne
| | - Ignacio Mir-Sanchis
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois
| | - A K Mitra
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - John Moult
- Institute for Bioscience and Biotechnology Research, Department of Cell Biology and Molecular genetics, University of Maryland, Rockville, Maryland, USA
| | - George N Phillips
- Department of BioSciences, Rice University, Houston, Texas.,Great Lakes Bioenergy Research Center, University of Wisconsin, Madison, Wisconsin
| | - Daniel M Pinkas
- Nuffield Department of Medicine; Structural Genomics Consortium, University of Oxford, Oxford, UK
| | - Phoebe A Rice
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois
| | - Yufeng Tong
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada.,Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada
| | - Maya Topf
- Institute of Structural and Molecular Biology, Birkbeck, University College London, London, UK
| | - Jonathan D Walton
- Great Lakes Bioenergy Research Center and Department of Plant Biology, Michigan State University, East Lansing, Michigan
| | - Torsten Schwede
- Biozentrum University of Basel, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Biozentrum University of Basel, Basel, Switzerland
| |
Collapse
|
72
|
Mohamed-Raseek N, Duan HD, Hildebrandt P, Mroginski MA, Miller AF. Spectroscopic, thermodynamic and computational evidence of the locations of the FADs in the nitrogen fixation-associated electron transfer flavoprotein. Chem Sci 2019; 10:7762-7772. [PMID: 31588324 PMCID: PMC6764259 DOI: 10.1039/c9sc00942f] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 06/24/2019] [Indexed: 01/15/2023] Open
Abstract
Flavin-based electron bifurcation allows enzymes to redistribute energy among electrons by coupling endergonic and exergonic electron transfer reactions. Diverse bifurcating enzymes employ a two-flavin electron transfer flavoprotein (ETF) that accepts hydride from NADH at a flavin (the so-called bifurcating FAD, Bf-FAD). The Bf-FAD passes one electron exergonically to a second flavin thereby assuming a reactive semiquinone state able to reduce ferredoxin or flavodoxin semiquinone. The flavin that accepts one electron and passes it on via exergonic electron transfer is known as the electron transfer FAD (ET-FAD) and is believed to correspond to the single FAD present in canonical ETFs, in domain II. The Bf-FAD is believed to be the one that is unique to bifurcating ETFs, bound between domains I and III. This very reasonable model has yet to be challenged experimentally. Herein we used site-directed mutagenesis to disrupt FAD binding to the presumed Bf site between domains I and III, in the Bf-ETF from Rhodopseudomonas palustris (RpaETF). The resulting protein contained only 0.80 ± 0.05 FAD, plus 1.21 ± 0.04 bound AMP as in canonical ETFs. The flavin was not subject to reduction by NADH, confirming absence of Bf-FAD. The retained FAD displayed visible circular dichroism (CD) similar to that of the ET-FAD of RpaETF. Likewise, the mutant underwent two sequential one-electron reductions forming and then consuming anionic semiquinone, reproducing the reactivity of the ET-FAD. These data confirm that the retained FAD in domain II corresponds the ET-FAD. Quantum chemical calculations of the absorbance and CD spectra of each of WT RpaETF's two flavins reproduced the observed differences between their CD and absorbance signatures. The calculations for the flavin bound in domain II agreed better with the spectra of the ET-flavin, and those calculated based on the flavin between domains I and III agreed better with spectra of the Bf-flavin. Thus calculations independently confirm the locations of each flavin. We conclude that the site in domain II harbours the ET-FAD whereas the mutated site between domains I and III is the Bf-FAD site, confirming the accepted model by two different tests.
Collapse
Affiliation(s)
- Nishya Mohamed-Raseek
- Dept. Chemistry , University of Kentucky , 505 Rose Street , Lexington , KY 40506-0055 , USA .
| | - H Diessel Duan
- Dept. Chemistry , University of Kentucky , 505 Rose Street , Lexington , KY 40506-0055 , USA .
| | - Peter Hildebrandt
- Max Volmer Laboratorum für Biophysikalische Chemie , Technische Universität - Berlin , Sekr. PC 14, 135 Straße des 17. Juni , 10623 Berlin , Germany
| | - Maria Andrea Mroginski
- Max Volmer Laboratorum für Biophysikalische Chemie , Technische Universität - Berlin , Sekr. PC 14, 135 Straße des 17. Juni , 10623 Berlin , Germany
| | - Anne-Frances Miller
- Dept. Chemistry , University of Kentucky , 505 Rose Street , Lexington , KY 40506-0055 , USA .
- Max Volmer Laboratorum für Biophysikalische Chemie , Technische Universität - Berlin , Sekr. PC 14, 135 Straße des 17. Juni , 10623 Berlin , Germany
| |
Collapse
|
73
|
Toplak M, Brunner J, Tabib CR, Macheroux P. Closing the gap: yeast electron-transferring flavoprotein links the oxidation of d-lactate and d-α-hydroxyglutarate to energy production via the respiratory chain. FEBS J 2019; 286:3611-3628. [PMID: 31081204 PMCID: PMC6771786 DOI: 10.1111/febs.14924] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/19/2019] [Accepted: 05/10/2019] [Indexed: 01/07/2023]
Abstract
Electron-transferring flavoproteins (ETFs) have been found in all kingdoms of life, mostly assisting in shuttling electrons to the respiratory chain for ATP production. While the human (h) ETF has been studied in great detail, very little is known about the biochemical properties of the homologous protein in the model organism Saccharomyces cerevisiae (yETF). In view of the absence of client dehydrogenases, for example, the acyl-CoA dehydrogenases involved in the β-oxidation of fatty acids, d-lactate dehydrogenase 2 (Dld2) appeared to be the only relevant enzyme that is serviced by yETF for electron transfer to the mitochondrial electron transport chain. However, this hypothesis was never tested experimentally. Here, we report the biochemical properties of yETF and Dld2 as well as the electron transfer reaction between the two proteins. Our study revealed that Dld2 oxidizes d-α-hydroxyglutarate more efficiently than d-lactate exhibiting kcatapp /KMapp values of 1200 ± 300 m-1 ·s-1 and 11 ± 2 m-1 ·s-1 , respectively. As expected, substrate-reduced Dld2 very slowly reacted with oxygen or the artificial electron acceptor 2,6-dichlorophenol indophenol. However, photoreduced Dld2 was rapidly reoxidized by oxygen, suggesting that the reaction products, that is, α-ketoglutarate and pyruvate, 'lock' the reduced enzyme in an unreactive state. Interestingly, however, we could demonstrate that substrate-reduced Dld2 rapidly transfers electrons to yETF. Therefore, we conclude that the formation of a product-reduced Dld2 complex suppresses electron transfer to dioxygen but favors the rapid reduction in yETF, thus preventing the loss of electrons and the generation of reactive oxygen species.
Collapse
Affiliation(s)
- Marina Toplak
- Institute of BiochemistryGraz University of TechnologyAustria
| | - Julia Brunner
- Institute of BiochemistryGraz University of TechnologyAustria
| | | | - Peter Macheroux
- Institute of BiochemistryGraz University of TechnologyAustria
| |
Collapse
|
74
|
Toplak M, Brunner J, Schmidt J, Macheroux P. Biochemical characterization of human D-2-hydroxyglutarate dehydrogenase and two disease related variants reveals the molecular cause of D-2-hydroxyglutaric aciduria. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2019; 1867:140255. [PMID: 31349060 DOI: 10.1016/j.bbapap.2019.07.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/12/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023]
Abstract
D-2-hydroxyglutaric aciduria is a neurometabolic disorder, characterized by the accumulation of D-2-hydroxyglutarate (D-2HG) in human mitochondria. Increased levels of D-2HG are detected in humans exhibiting point mutations in the genes encoding isocitrate dehydrogenase, citrate carrier, the electron transferring flavoprotein (ETF) and its downstream electron acceptor ETF-ubiquinone oxidoreductase or D-2-hydroxyglutarate dehydrogenase (hD2HGDH). However, while the pathogenicity of several amino acid replacements in the former four proteins has been studied extensively, not much is known about the effect of certain point mutations on the biochemical properties of hD2HGDH. Therefore, we recombinantly produced wild type hD2HGDH as well as two recently identified disease-related variants (hD2HGDH-I147S and -V444A) and performed their detailed biochemical characterization. We could show that hD2HGDH is a FAD dependent protein, which is able to catalyze the oxidation of D-2HG and D-lactate to α-ketoglutarate and pyruvate, respectively. The two variants were obtained as apo-proteins and were thus catalytically inactive. The addition of FAD failed to restore enzymatic activity of the variants, indicating that the cofactor binding site is compromised by the single amino acid replacements. Further analyses revealed that both variants form aggregates that are apparently unable to bind the FAD cofactor. Since, D-2-hydroxyglutaric aciduria may also result from a loss of function of either the ETF or its downstream electron acceptor ETF-ubiquinone oxidoreductase, ETF may serve as the cognate electron acceptor of reduced hD2HGDH. Here, we show that hD2HGDH directly reduces recombinant human ETF, thus establishing a metabolic link between the oxidation of D-2-hydroxyglutarate and the mitochondrial electron transport chain.
Collapse
Affiliation(s)
- Marina Toplak
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/2, A-8010 Graz, Austria
| | - Julia Brunner
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/2, A-8010 Graz, Austria
| | - Julia Schmidt
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/2, A-8010 Graz, Austria
| | - Peter Macheroux
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/2, A-8010 Graz, Austria.
| |
Collapse
|
75
|
Rohman A, Dijkstra BW. The role and mechanism of microbial 3-ketosteroid Δ 1-dehydrogenases in steroid breakdown. J Steroid Biochem Mol Biol 2019; 191:105366. [PMID: 30991094 DOI: 10.1016/j.jsbmb.2019.04.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/26/2019] [Accepted: 04/12/2019] [Indexed: 02/08/2023]
Abstract
3-Ketosteroid Δ1-dehydrogenases are FAD-dependent enzymes that catalyze the introduction of a double bond between the C1 and C2 atoms of the A-ring of 3-ketosteroid substrates. These enzymes are found in a large variety of microorganisms, especially in bacteria belonging to the phylum Actinobacteria. They play a critical role in the early steps of the degradation of the steroid core. 3-Ketosteroid Δ1-dehydrogenases are of particular interest for the etiology of some infectious diseases, for the production of starting materials for the pharmaceutical industry, and for environmental bioremediation applications. Here we summarize and discuss the biochemical and enzymological properties of these enzymes, their microbial sources, and their natural diversity. The three-dimensional structure of a 3-ketosteroid Δ1-dehydrogenase in connection with the enzyme mechanism is highlighted.
Collapse
Affiliation(s)
- Ali Rohman
- Department of Chemistry, Faculty of Science and Technology, Universitas Airlangga, Surabaya 60115, Indonesia; The Laboratory of Proteomics, Institute of Tropical Disease, Universitas Airlangga, Surabaya 60115, Indonesia; The Laboratory of Biophysical Chemistry, University of Groningen, 9747 AG Groningen, the Netherlands
| | - Bauke W Dijkstra
- The Laboratory of Biophysical Chemistry, University of Groningen, 9747 AG Groningen, the Netherlands.
| |
Collapse
|
76
|
Zhang Y, Bharathi SS, Beck ME, Goetzman ES. The fatty acid oxidation enzyme long-chain acyl-CoA dehydrogenase can be a source of mitochondrial hydrogen peroxide. Redox Biol 2019; 26:101253. [PMID: 31234015 PMCID: PMC6597861 DOI: 10.1016/j.redox.2019.101253] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 05/31/2019] [Accepted: 06/10/2019] [Indexed: 11/29/2022] Open
Abstract
Fatty acid oxidation (FAO)-driven H2O2 has been shown to be a major source of oxidative stress in several tissues and disease states. Here, we established that the mitochondrial flavoprotein long-chain acyl-CoA dehydrogenase (LCAD), which catalyzes a key step in mitochondrial FAO, directly produces H2O2in vitro by leaking electrons to oxygen. Kinetic analysis of recombinant human LCAD showed that it produces H2O2 15-fold faster than the related mitochondrial enzyme very long-chain acyl-CoA dehydrogenase (VLCAD), but 50-fold slower than a bona fide peroxisomal acyl-CoA oxidase. The rate of H2O2 formation by human LCAD is slow compared to its activity as a dehydrogenase (about 1%). However, expression of hLCAD in HepG2 cells is sufficient to significantly increase H2O2 in the presence of fatty acids. Liver mitochondria from LCAD−/− mice, but not VLCAD−/− mice, produce significantly less H2O2 during incubation with fatty acids. Finally, we observe highest LCAD expression in human liver, followed by kidney, lung, and pancreas. Based on our data, we propose that the presence of LCAD drives H2O2 formation in response to fatty acids in these tissues.
Collapse
Affiliation(s)
- Yuxun Zhang
- 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
| | - Sivakama S Bharathi
- 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
| | - Megan E Beck
- 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
| | - Eric S Goetzman
- 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
|
77
|
An acyl-CoA dehydrogenase microplate activity assay using recombinant porcine electron transfer flavoprotein. Anal Biochem 2019; 581:113332. [PMID: 31194945 DOI: 10.1016/j.ab.2019.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 12/12/2022]
Abstract
Acyl-CoA dehydrogenases (ACADs) play key roles in the mitochondrial catabolism of fatty acids and branched-chain amino acids. All nine characterized ACAD enzymes use electron transfer flavoprotein (ETF) as their redox partner. The gold standard for measuring ACAD activity is the anaerobic ETF fluorescence reduction assay, which follows the decrease of pig ETF fluorescence as it accepts electrons from an ACAD in vitro. Although first described 35 years ago, the assay has not been widely used due to the need to maintain an anaerobic assay environment and to purify ETF from pig liver mitochondria. Here, we present a method for expressing recombinant pig ETF in E coli and purifying it to homogeneity. The recombinant protein is virtually pure after one chromatography step, bears higher intrinsic fluorescence than the native enzyme, and provides enhanced activity in the ETF fluorescence reduction assay. Finally, we present a simplified protocol for removing molecular oxygen that allows adaption of the assay to a 96-well plate format. The availability of recombinant pig ETF and the microplate version of the ACAD activity assay will allow wide application of the assay for both basic research and clinical diagnostics.
Collapse
|
78
|
Matejka K, Stückler F, Salomon M, Ensenauer R, Reischl E, Hoerburger L, Grallert H, Kastenmüller G, Peters A, Daniel H, Krumsiek J, Theis FJ, Hauner H, Laumen H. Dynamic modelling of an ACADS genotype in fatty acid oxidation - Application of cellular models for the analysis of common genetic variants. PLoS One 2019; 14:e0216110. [PMID: 31120904 PMCID: PMC6532850 DOI: 10.1371/journal.pone.0216110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 04/15/2019] [Indexed: 11/19/2022] Open
Abstract
Background Genome-wide association studies of common diseases or metabolite quantitative traits often identify common variants of small effect size, which may contribute to phenotypes by modulation of gene expression. Thus, there is growing demand for cellular models enabling to assess the impact of gene regulatory variants with moderate effects on gene expression. Mitochondrial fatty acid oxidation is an important energy metabolism pathway. Common noncoding acyl-CoA dehydrogenase short chain (ACADS) gene variants are associated with plasma C4-acylcarnitine levels and allele-specific modulation of ACADS expression may contribute to the observed phenotype. Methods and findings We assessed ACADS expression and intracellular acylcarnitine levels in human lymphoblastoid cell lines (LCL) genotyped for a common ACADS variant associated with plasma C4-acylcarnitine and found a significant genotype-dependent decrease of ACADS mRNA and protein. Next, we modelled gradual decrease of ACADS expression using a tetracycline-regulated shRNA-knockdown of ACADS in Huh7 hepatocytes, a cell line with high fatty acid oxidation-(FAO)-capacity. Assessing acylcarnitine flux in both models, we found increased C4-acylcarnitine levels with decreased ACADS expression levels. Moreover, assessing time-dependent changes of acylcarnitine levels in shRNA-hepatocytes with altered ACADS expression levels revealed an unexpected effect on long- and medium-chain fatty acid intermediates. Conclusions Both, genotyped LCL and regulated shRNA-knockdown are valuable tools to model moderate, gradual gene-regulatory effects of common variants on cellular phenotypes. Decreasing ACADS expression levels modulate short and surprisingly also long/medium chain acylcarnitines, and may contribute to increased plasma acylcarnitine levels.
Collapse
Affiliation(s)
- Kerstin Matejka
- Chair of Nutritional Medicine, Else Kröner-Fresenius-Center for Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
- ZIEL-Research Center for Nutrition and Food Sciences, TUM School of Life Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Clinical Cooperation Group Nutrigenomics and Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany
- Clinical Cooperation Group Nutrigenomics and Type 2 Diabetes, Technische Universität München, Freising-Weihenstephan, Germany
| | - Ferdinand Stückler
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | | | - Regina Ensenauer
- Research Center, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-Universität München, München, Germany
- Experimental Pediatrics and Metabolism, Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Child Nutrition, Max Rubner-Institut, Karlsruhe, Germany
| | - Eva Reischl
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lena Hoerburger
- Paediatric Nutritional Medicine, Else Kröner-Fresenius-Centre for Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
| | - Harald Grallert
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Clinical Cooperation Group Nutrigenomics and Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany
- Clinical Cooperation Group Nutrigenomics and Type 2 Diabetes, Technische Universität München, Freising-Weihenstephan, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Annette Peters
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- German Research Center for Cardiovascular Disease (DZHK-Munich partner site), Neuherberg, Germany
| | - Hannelore Daniel
- ZIEL-Research Center for Nutrition and Food Sciences, TUM School of Life Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
- Chair of Physiology of Human Nutrition, TUM School of Life Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
| | - Jan Krumsiek
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, United States of America
| | - Fabian J. Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Mathematical Science, Technische Universität München, Garching, Germany
- * E-mail: (FJT); (HL)
| | - Hans Hauner
- Chair of Nutritional Medicine, Else Kröner-Fresenius-Center for Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
- ZIEL-Research Center for Nutrition and Food Sciences, TUM School of Life Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Clinical Cooperation Group Nutrigenomics and Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany
- Clinical Cooperation Group Nutrigenomics and Type 2 Diabetes, Technische Universität München, Freising-Weihenstephan, Germany
- Else Kröner-Fresenius-Center for Nutritional Medicine, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Helmut Laumen
- Chair of Nutritional Medicine, Else Kröner-Fresenius-Center for Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
- ZIEL-Research Center for Nutrition and Food Sciences, TUM School of Life Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Clinical Cooperation Group Nutrigenomics and Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany
- Clinical Cooperation Group Nutrigenomics and Type 2 Diabetes, Technische Universität München, Freising-Weihenstephan, Germany
- Paediatric Nutritional Medicine, Else Kröner-Fresenius-Centre for Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Research Unit Protein Science, Helmholtz Zentrum München, Neuherberg, Germany
- * E-mail: (FJT); (HL)
| |
Collapse
|
79
|
Peters V, Morath M, Mack M, Liesert M, Buckel W, Hoffmann GF, Vockley J, Ghisla S, Zschocke J. Formation of 3-hydroxyglutaric acid in glutaric aciduria type I: in vitro participation of medium chain acyl-CoA dehydrogenase. JIMD Rep 2019; 47:30-34. [PMID: 31240164 PMCID: PMC6498835 DOI: 10.1002/jmd2.12026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/28/2019] [Indexed: 11/13/2022] Open
Abstract
3-Hydroxyglutaric acid (3-OH-GA) in urine has been identified as the most reliable diagnostic marker for glutaric aciduria type I (GA I). We showed that hydratation of glutaconyl-CoA to 3-hydroxyglutaryl-CoA, which is subsequently hydrolyzed to 3-OH-GA, is efficiently catalyzed by 3-methylglutaconyl-CoA hydratase (3-MGH). We have now investigated whether mitochondrial acyl-CoA-dehydrogenases can convert glutaryl-CoA to glutaconyl-CoA. Short-chain acyl-CoA dehydrogenase (SCAD), medium-chain acyl-CoA dehydrogenase (MCAD), and long-chain acyl-CoA dehydrogenase (LCAD) accepted glutaryl-CoA as a substrate. The highest k cat of glutaryl-CoA was found for MCAD (0.12 ± 0.01 second-1) and was about 26-fold and 52-fold higher than those of LCAD and SCAD, respectively. The turnover of MCAD for glutaryl-CoA was about 1.5% of that of its natural substrate octanoyl-CoA. Despite high K m (above 600 μM) and low turnover rate, the oxidation of glutaryl-CoA by MCAD in combination with 3-MGH could explain the urinary concentration of 3-OH-GA in GA I patients.
Collapse
Affiliation(s)
- Verena Peters
- Center für Paediatric and Adolescent MedicineUniversity of HeidelbergGermany
| | - Marina Morath
- Center für Paediatric and Adolescent MedicineUniversity of HeidelbergGermany
| | - Matthias Mack
- Institut für Technische MikrobiologieHochschule MannheimGermany
| | - Michael Liesert
- Laboratorium für Mikrobiologie, Fachbereich BiologiePhilipps‐Universität MarburgGermany
| | - Wolfgang Buckel
- Laboratorium für Mikrobiologie, Fachbereich BiologiePhilipps‐Universität MarburgGermany
| | - Georg F. Hoffmann
- Center für Paediatric and Adolescent MedicineUniversity of HeidelbergGermany
| | - Jerry Vockley
- Department of Pediatric and Human GeneticsChildren's Hospital of Pittsburgh and University of PittsburghPittsburghPennsylvania
| | - Sandro Ghisla
- Department of Biology, Section of Natural SciencesUniversität KonstanzGermany
| | | |
Collapse
|
80
|
Buckel W. Enzymatic Reactions Involving Ketyls: From a Chemical Curiosity to a General Biochemical Mechanism. Biochemistry 2019; 58:5221-5233. [PMID: 30995029 DOI: 10.1021/acs.biochem.9b00171] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ketyls are radical anions with nucleophilic properties. Ketyls obtained by enzymatic one-electron reduction of thioesters were proposed as intermediates for the dehydration of (R)-2-hydroxyacyl-CoA to (E)-2-enoyl-CoA. This concept was extended to the Birch-like reduction of benzoyl-CoA to 1,5-cyclohexadienecarboxyl-CoA. Nature uses two methods to achieve the therefore required low reduction potentials of less than -600 mV, either by an ATP-driven electron transfer similar to that catalyzed by the iron protein of nitrogenase or by electron bifurcation. Ketyls formed by thiyl radical-initiated oxidation of alcohols followed by deprotonation are involved in coenzyme B12-independent diol dehydratases, other glycyl radical enzymes mediating key reactions in the degradations of choline, taurine, and 4-hydroxyproline, and all three classes of ribonucleotide reductases. A special case is the dehydration of 4-hydroxybutyryl-CoA to crotonyl-CoA, which most likely proceeds via an oxidation to an allylic ketyl but requires neither a strong reductant nor an external radical generator.
Collapse
Affiliation(s)
- Wolfgang Buckel
- Fachbereich Biologie , Philipps-Universität , 35032 Marburg , Germany
| |
Collapse
|
81
|
Evaluation of Metabolic Defects in Fatty Acid Oxidation Using Peripheral Blood Mononuclear Cells Loaded with Deuterium-Labeled Fatty Acids. DISEASE MARKERS 2019; 2019:2984747. [PMID: 30881520 PMCID: PMC6383405 DOI: 10.1155/2019/2984747] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/13/2018] [Accepted: 12/03/2018] [Indexed: 11/17/2022]
Abstract
Because tandem mass spectrometry- (MS/MS-) based newborn screening identifies many suspicious cases of fatty acid oxidation and carnitine cycle disorders, a simple, noninvasive test is required to confirm the diagnosis. We have developed a novel method to evaluate the metabolic defects in peripheral blood mononuclear cells loaded with deuterium-labeled fatty acids directly using the ratios of acylcarnitines determined by flow injection MS/MS. We have identified diagnostic indices for the disorders as follows: decreased ratios of d27-C14-acylcarnitine/d31-C16-acylcarnitine and d23-C12-acylcarnitine/d31-C16-acylcarnitine for carnitine palmitoyltransferase-II (CPT-II) deficiency, decreased ratios of d23-C12-acylcarnitine/d27-C14-acylcarnitine for very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency, and increased ratios of d29-C16-OH-acylcarnitine/d31-C16-acylcarnitine for trifunctional protein (TFP) deficiency, together with increased ratios of d7-C4-acylcarnitine/d31-C16-acylcarnitine for carnitine palmitoyltransferase-I deficiency. The decreased ratios of d1-acetylcarnitine/d31-C16-acylcarnitine could be indicative of β-oxidation ability in patients with CPT-II, VLCAD, and TFP deficiencies. Overall, our data showed that the present method was valuable for establishing a rapid diagnosis of fatty acid oxidation disorders and carnitine cycle disorders and for complementing gene analysis because our diagnostic indices may overcome the weaknesses of conventional enzyme activity measurements using fibroblasts or mononuclear cells with assumedly uncertain viability.
Collapse
|
82
|
Tang J, Hu J, Xue M, Guo Z, Xie M, Zhang B, Zhou Z, Huang W, Hou S. Maternal diet deficient in riboflavin induces embryonic death associated with alterations in the hepatic proteome of duck embryos. Nutr Metab (Lond) 2019; 16:19. [PMID: 30918526 PMCID: PMC6419344 DOI: 10.1186/s12986-019-0345-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 03/05/2019] [Indexed: 12/14/2022] Open
Abstract
Background Maternal riboflavin deficiency (RD) induces embryonic death in poultry. The underlying mechanisms, however, remain to be established and an overview of molecular alterations at the protein level is still lacking. We investigated embryonic hepatic proteome changes induced by maternal RD to explain embryonic death. Methods A total of 80 45-week-old breeding female ducks were divided into two groups of 40 birds each, and all birds were raised individually for 8 weeks. All the female ducks received either a RD or a riboflavin adequate (control, CON) diet, which supplemented the basal diet with 0 or 10 mg riboflavin /kg of diet respectively. Results The riboflavin concentrations of maternal plasma and egg yolk, as well as egg hatchability declined markedly in the RD group compared to those in the CON group after 2 weeks, and declined further over time. The hepatic proteome of E13 viable embryos from 8-week fertile eggs showed that 223 proteins were upregulated and 366 proteins were downregulated (> 1.5-fold change) in the RD group compared to those in the CON group. Pathway analysis showed that differentially expressed proteins were mainly enriched in the fatty acid beta-oxidation, electron transport chain (ETC), and tricarboxylic acid (TCA) cycle. Specifically, all the proteins involved in the fatty acid beta-oxidation and ETC, as well as six out of seven proteins involved in the TCA cycle, were diminished in the RD group, indicating that these processes could be impaired by RD. Conclusion Maternal RD leads to embryonic death of offspring and is associated with impaired energy generation processes, indicated by a number of downregulated proteins involved in the fatty acid beta-oxidation, ETC, and TCA cycle in the hepatic of duck embryos. These findings contribute to our understanding of the mechanisms of liver metabolic disorders due to maternal RD. Electronic supplementary material The online version of this article (10.1186/s12986-019-0345-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jing Tang
- 1State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Jian Hu
- 1State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Ming Xue
- 2National Animal Husbandry Service, Beijing, 100125 China
| | - Zhanbao Guo
- 1State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Ming Xie
- 1State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Bo Zhang
- 1State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Zhengkui Zhou
- 1State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Wei Huang
- 1State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Shuisheng Hou
- 1State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| |
Collapse
|
83
|
Li Y, Zheng X, Zhu M, Chen M, Zhang S, He F, Chen X, Lv J, Pei M, Zhang Y, Zhang Y, Wang W, Zhang J, Wang M, Wang Z, Li G, Lu G. MoIVD-Mediated Leucine Catabolism Is Required for Vegetative Growth, Conidiation and Full Virulence of the Rice Blast Fungus Magnaporthe oryzae. Front Microbiol 2019; 10:444. [PMID: 30923517 PMCID: PMC6426774 DOI: 10.3389/fmicb.2019.00444] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/20/2019] [Indexed: 01/03/2023] Open
Abstract
Isovaleryl-CoA dehydrogenase (IVD), a member of the acyl-CoA dehydrogenase (ACAD) family, is a key enzyme catalyzing the conversion of isovaleryl-CoA to β-methylcrotonyl-CoA in the third reaction of the leucine catabolism pathway and simultaneously transfers electrons to the electron-transferring flavoprotein (ETF) for ATP synthesis. We previously identified the ETF ortholog in rice blast fungus Magnaporthe oryzae (MoETF) and showed that MoETF was essential for fungal growth, conidiation and pathogenicity. To further investigate the biological function of electron-transferring proteins and clarify the role of leucine catabolism in growth and pathogenesis, we characterized MoIVD (M. oryzaeisovaleryl-CoA dehydrogenase). MoIvd is highly conserved in fungi and its expression was highly induced by leucine. The Δmoivd mutants showed reduced growth, decreased conidiation and compromised pathogenicity, while the conidial germination and appressorial formation appeared normal. Consistent with a block in leucine degradation, the Δmoivd mutants accumulated isovaleric acid, grew more slowly, fully lacked pigmentation and completely failed to produce conidia on leucine-rich medium. These defects were largely rescued by raising the extracellular pH, suggesting that the accumulation of isovaleric acid contributes to the growth and conidiation defects. However, the reduced virulence of the mutants was probably due to their inability to overcome oxidative stress, since a large amount of ROS (reactive oxygen species) accumulated in infected host cell. In addition, MoIvd is localized to mitochondria and interacted with its electron receptor MoEtfb, the β subunit of MoEtf. Taken together, our results suggest that MoIVD functions in leucine catabolism and is required for the vegetative growth, conidiation and full virulence of M. oryzae, providing the first evidence for IVD-mediated leucine catabolism in the development and pathogenesis of plant fungal pathogens.
Collapse
Affiliation(s)
- Ya Li
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiuxia Zheng
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Minghui Zhu
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Mengting Chen
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Shengnan Zhang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Fangyuan He
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiaomin Chen
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jiarui Lv
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Mengtian Pei
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Ye Zhang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yunhui Zhang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Wenzong Wang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jing Zhang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Mo Wang
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, Plant Immunity Center, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Zonghua Wang
- Institute of Oceanography, Minjiang University, Fuzhou, China
| | - Guangpu Li
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Guodong Lu
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
84
|
Shao X, Cao HY, Zhao F, Peng M, Wang P, Li CY, Shi WL, Wei TD, Yuan Z, Zhang XH, Chen XL, Todd JD, Zhang YZ. Mechanistic insight into 3-methylmercaptopropionate metabolism and kinetical regulation of demethylation pathway in marine dimethylsulfoniopropionate-catabolizing bacteria. Mol Microbiol 2019; 111:1057-1073. [PMID: 30677184 PMCID: PMC6850173 DOI: 10.1111/mmi.14211] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2019] [Indexed: 01/25/2023]
Abstract
The vast majority of oceanic dimethylsulfoniopropionate (DMSP) is thought to be catabolized by bacteria via the DMSP demethylation pathway. This pathway contains four enzymes termed DmdA, DmdB, DmdC and DmdD/AcuH, which together catabolize DMSP to acetylaldehyde and methanethiol as carbon and sulfur sources respectively. While molecular mechanisms for DmdA and DmdD have been proposed, little is known of the catalytic mechanisms of DmdB and DmdC, which are central to this pathway. Here, we undertake physiological, structural and biochemical analyses to elucidate the catalytic mechanisms of DmdB and DmdC. DmdB, a 3-methylmercaptopropionate (MMPA)-coenzyme A (CoA) ligase, undergoes two sequential conformational changes to catalyze the ligation of MMPA and CoA. DmdC, a MMPA-CoA dehydrogenase, catalyzes the dehydrogenation of MMPA-CoA to generate MTA-CoA with Glu435 as the catalytic base. Sequence alignment suggests that the proposed catalytic mechanisms of DmdB and DmdC are likely widely adopted by bacteria using the DMSP demethylation pathway. Analysis of the substrate affinities of involved enzymes indicates that Roseobacters kinetically regulate the DMSP demethylation pathway to ensure DMSP functioning and catabolism in their cells. Altogether, this study sheds novel lights on the catalytic and regulative mechanisms of bacterial DMSP demethylation, leading to a better understanding of bacterial DMSP catabolism.
Collapse
Affiliation(s)
- Xuan Shao
- State Key Laboratory of Microbial Technology, Marine Biotechnology Research Center, Shandong University, Qingdao, 266237, China
| | - Hai-Yan Cao
- State Key Laboratory of Microbial Technology, Marine Biotechnology Research Center, Shandong University, Qingdao, 266237, China
| | - Fang Zhao
- State Key Laboratory of Microbial Technology, Marine Biotechnology Research Center, Shandong University, Qingdao, 266237, China
| | - Ming Peng
- State Key Laboratory of Microbial Technology, Marine Biotechnology Research Center, Shandong University, Qingdao, 266237, China
| | - Peng Wang
- State Key Laboratory of Microbial Technology, Marine Biotechnology Research Center, Shandong University, Qingdao, 266237, China
| | - Chun-Yang Li
- State Key Laboratory of Microbial Technology, Marine Biotechnology Research Center, Shandong University, Qingdao, 266237, China.,College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.,Suzhou Institute of Shandong University, Suzhou, 215123, China
| | - Wei-Ling Shi
- State Key Laboratory of Microbial Technology, Marine Biotechnology Research Center, Shandong University, Qingdao, 266237, China
| | - Tian-Di Wei
- State Key Laboratory of Microbial Technology, Marine Biotechnology Research Center, Shandong University, Qingdao, 266237, China
| | - Zenglin Yuan
- State Key Laboratory of Microbial Technology, Marine Biotechnology Research Center, Shandong University, Qingdao, 266237, China
| | - Xiao-Hua Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Xiu-Lan Chen
- State Key Laboratory of Microbial Technology, Marine Biotechnology Research Center, Shandong University, Qingdao, 266237, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Jonathan D Todd
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Yu-Zhong Zhang
- State Key Laboratory of Microbial Technology, Marine Biotechnology Research Center, Shandong University, Qingdao, 266237, China.,College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| |
Collapse
|
85
|
Sinha T, Al-Ubaidi MR, Naash MI. Flavin Imbalance as an Important Player in Diabetic Retinopathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1185:575-579. [PMID: 31884673 DOI: 10.1007/978-3-030-27378-1_94] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The retina and RPE together constitute the most metabolically active ecosystem in the body, harboring high levels of flavins. Although diabetic patients have been reported to suffer from riboflavin deficiency and use of flavins as nutritional interventions to combat diabetic insult on other tissues have been investigated, such attempts have never been tested for the retina to avoid diabetic retinopathy. Furthermore, the role of flavins in pathophysiology of the retina and RPE has mostly been overlooked. Herein, we review the impact of flavins on various clinical manifestations of diabetic retinopathy and discuss possible ways to address them.
Collapse
Affiliation(s)
- Tirthankar Sinha
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA.
| | - Muayyad R Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Muna I Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| |
Collapse
|
86
|
|
87
|
Wu GQ, Wang JL, Feng RJ, Li SJ, Wang CM. iTRAQ-Based Comparative Proteomic Analysis Provides Insights into Molecular Mechanisms of Salt Tolerance in Sugar Beet ( Beta vulgaris L.). Int J Mol Sci 2018; 19:ijms19123866. [PMID: 30518064 PMCID: PMC6321137 DOI: 10.3390/ijms19123866] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/31/2018] [Accepted: 10/31/2018] [Indexed: 01/24/2023] Open
Abstract
Salinity is one of the major abiotic stress factors that limit plant growth and crop yield worldwide. To understand the molecular mechanisms and screen the key proteins in response of sugar beet (Beta vulgaris L.) to salt, in the present study, the proteomics of roots and shoots in three-week-old sugar beet plants exposed to 50 mM NaCl for 72 h was investigated by isobaric Tags for Relative and Absolute Quantitation (iTRAQ) technology. The results showed that 105 and 30 differentially expressed proteins (DEPs) were identified in roots and shoots of salt-treated plants compared with untreated plants, respectively. There were 46 proteins up-regulated and 59 proteins down-regulated in roots; and 13 up-regulated proteins and 17 down-regulated proteins found in shoots, respectively. These DEPs were mainly involved in carbohydrate metabolism, energy metabolism, lipid metabolism, biosynthesis of secondary metabolites, transcription, translation, protein folding, sorting, and degradation as well as transport. It is worth emphasizing that some novel salt-responsive proteins were identified, such as PFK5, MDH, KAT2, ACAD10, CYP51, F3H, TAL, SRPR, ZOG, V-H+-ATPase, V-H+-PPase, PIPs, TIPs, and tubulin α-2/β-1 chain. qRT-PCR analysis showed that six of the selected proteins, including BvPIP1-4, BvVP and BvVAP in root and BvTAL, BvURO-D1, and BvZOG in shoot, displayed good correlation between the expression levels of protein and mRNA. These novel proteins provide a good starting point for further research into their functions using genetic or other approaches. These findings should significantly improve the understanding of the molecular mechanisms involved in salt tolerance of sugar beet plants.
Collapse
Affiliation(s)
- Guo-Qiang Wu
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China.
| | - Jin-Long Wang
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China.
| | - Rui-Jun Feng
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China.
| | - Shan-Jia Li
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China.
| | - Chun-Mei Wang
- Lanzhou Institute of Husbandry and Pharmaceutical Science, CAAS, Lanzhou 730050, China.
| |
Collapse
|
88
|
Expansive microbial metabolic versatility and biodiversity in dynamic Guaymas Basin hydrothermal sediments. Nat Commun 2018; 9:4999. [PMID: 30479325 PMCID: PMC6258724 DOI: 10.1038/s41467-018-07418-0] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 10/31/2018] [Indexed: 12/16/2022] Open
Abstract
Microbes in Guaymas Basin (Gulf of California) hydrothermal sediments thrive on hydrocarbons and sulfur and experience steep, fluctuating temperature and chemical gradients. The functional capacities of communities inhabiting this dynamic habitat are largely unknown. Here, we reconstructed 551 genomes from hydrothermally influenced, and nearby cold sediments belonging to 56 phyla (40 uncultured). These genomes comprise 22 unique lineages, including five new candidate phyla. In contrast to findings from cold hydrocarbon seeps, hydrothermal-associated communities are more diverse and archaea dominate over bacteria. Genome-based metabolic inferences provide first insights into the ecological niches of these uncultured microbes, including methane cycling in new Crenarchaeota and alkane utilization in ANME-1. These communities are shaped by a high biodiversity, partitioning among nitrogen and sulfur pathways and redundancy in core carbon-processing pathways. The dynamic sediments select for distinctive microbial communities that stand out by expansive biodiversity, and open up new physiological perspectives into hydrothermal ecosystem function. The diversity and function of microbes inhabiting hydrothermal areas is a topic of active interest in marine microbiology. Here, the authors assemble genomes from Guaymas Basin hydrothermal sediments and describe the metabolic roles of the bacterial community, which includes five new bacterial candidate phyla
Collapse
|
89
|
Xu J, Li D, Lv J, Xu X, Wen B, Lin P, Liu F, Ji K, Shan J, Li H, Li W, Zhao Y, Zhao D, Pok JY, Yan C. ETFDH Mutations and Flavin Adenine Dinucleotide Homeostasis Disturbance Are Essential for Developing Riboflavin-Responsive Multiple Acyl-Coenzyme A Dehydrogenation Deficiency. Ann Neurol 2018; 84:659-673. [PMID: 30232818 DOI: 10.1002/ana.25338] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 08/26/2018] [Accepted: 09/09/2018] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Riboflavin-responsive multiple acyl-coenzyme A dehydrogenation deficiency (RR-MADD) is an inherited fatty acid metabolism disorder mainly caused by genetic defects in electron transfer flavoprotein-ubiquinone oxidoreductase (ETF:QO). The variant ETF:QO protein folding deficiency, which can be corrected by therapeutic dosage of riboflavin supplement, has been identified in HEK-293 cells and is believed to be the molecular mechanism of this disease. To verify this hypothesis in vivo, we generated Etfdh (h)A84T knockin (KI) mice. METHODS Tissues from these mice as well as muscle biopsies and fibroblasts from 7 RR-MADD patients were used to examine the flavin adenine dinucleotide (FAD) concentration and ETF:QO protein amount. RESULTS All of the homozygous KI mice (Etfdh (h)A84T/(h)A84T , KI/KI) were initially normal. After being given a high-fat and vitamin B2 -deficient (HF-B2 D) diet for 5 weeks, they developed weight loss, movement ability defects, lipid storage in muscle and liver, and elevated serum acyl-carnitine levels, which are clinically and biochemically similar to RR-MADD patients. Both ETF:QO protein and FAD concentrations were significantly decreased in tissues of HF-B2 D-KI/KI mice and in cultured fibroblasts from RR-MADD patients. After riboflavin treatment, ETF:QO protein increased in proportion to elevated FAD concentrations, but not related to mRNA levels. These results were further confirmed in cultured fibroblasts from RR-MADD patients. INTERPRETATION For the first time, we successfully developed a RR-MADD mice model and confirmed that FAD homeostasis disturbances played a crucial role on the pathomechanism of RR-MADD in this mouse model and culture cells from patients. Supplementation of riboflavin may stabilize variant ETF:QO protein by rebuilding FAD homeostasis. Ann Neurol 2018;84:667-681.
Collapse
Affiliation(s)
- Jingwen Xu
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, Jinan, China
| | - Duoling Li
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, Jinan, China
| | - Jingwei Lv
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, Jinan, China
| | - Xuebi Xu
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, Jinan, China
| | - Bing Wen
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, Jinan, China
| | - Pengfei Lin
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, Jinan, China
| | - Fuchen Liu
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, Jinan, China.,Department of Neurobiology, Yale University School of Medicine, New Haven, CT
| | - Kunqian Ji
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, Jinan, China
| | - Jingli Shan
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, Jinan, China
| | - Honghao Li
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, Jinan, China
| | - Wei Li
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, Jinan, China
| | - Yuying Zhao
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, Jinan, China
| | - Dandan Zhao
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, Jinan, China
| | - Joo Y Pok
- Department of Neurology, Yale University, New Haven, CT
| | - Chuanzhu Yan
- Research Institute of Neuromuscular and Neurodegenerative Disease, Department of Neurology, Qilu Hospital, Shandong University, Jinan, China.,Brain Science Research Institute, Shandong University, Jinan, China.,Laboratory of Mitochondrial Medicine, Qilu Hospital (Qingdao), Qingdao, China
| |
Collapse
|
90
|
Lee CC, Ko TP, Chen CT, Chan YT, Lo SY, Chang JY, Chen YW, Chung TF, Hsieh HJ, Hsiao CD, Wang AHJ. Crystal Structure of PigA: A Prolyl Thioester-Oxidizing Enzyme in Prodigiosin Biosynthesis. Chembiochem 2018; 20:193-202. [DOI: 10.1002/cbic.201800409] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Cheng-Chung Lee
- Institute of Biological Chemistry; Academia Sinica; 128 Academia Road Section 2 Taipei 11529 Taiwan
| | - Tzu-Ping Ko
- Institute of Biological Chemistry; Academia Sinica; 128 Academia Road Section 2 Taipei 11529 Taiwan
| | - Chun-Ting Chen
- Institute of Biological Chemistry; Academia Sinica; 128 Academia Road Section 2 Taipei 11529 Taiwan
| | - Yueh-Te Chan
- Institute of Biological Chemistry; Academia Sinica; 128 Academia Road Section 2 Taipei 11529 Taiwan
| | - Shin-Yi Lo
- Institute of Biological Chemistry; Academia Sinica; 128 Academia Road Section 2 Taipei 11529 Taiwan
| | - Jen-Yu Chang
- Institute of Biological Chemistry; Academia Sinica; 128 Academia Road Section 2 Taipei 11529 Taiwan
| | - Ya-Wen Chen
- Institute of Biological Chemistry; Academia Sinica; 128 Academia Road Section 2 Taipei 11529 Taiwan
| | - Ting-Fang Chung
- Institute of Biological Chemistry; Academia Sinica; 128 Academia Road Section 2 Taipei 11529 Taiwan
| | - Hsin-Ju Hsieh
- Industrial Technology Research Institute; 195 Chung Hsing Road Section 4, Chutung Hsinchu 31057 Taiwan
| | - Chwan-Deng Hsiao
- Institute of Molecular Biology; Academia Sinica; 128 Academia Road Section 2 Taipei 11529 Taiwan
| | - Andrew H.-J. Wang
- Institute of Biological Chemistry; Academia Sinica; 128 Academia Road Section 2 Taipei 11529 Taiwan
| |
Collapse
|
91
|
Talà A, Damiano F, Gallo G, Pinatel E, Calcagnile M, Testini M, Fico D, Rizzo D, Sutera A, Renzone G, Scaloni A, De Bellis G, Siculella L, De Benedetto GE, Puglia AM, Peano C, Alifano P. Pirin: A novel redox-sensitive modulator of primary and secondary metabolism in Streptomyces. Metab Eng 2018; 48:254-268. [PMID: 29944936 DOI: 10.1016/j.ymben.2018.06.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 10/28/2022]
Abstract
Pirins are evolutionarily conserved iron-containing proteins that are found in all kingdoms of life, and have been implicated in diverse molecular processes, mostly associated with cellular stress. In the present study, we started from the evidence that the insertional inactivation of pirin-like gene SAM23877_RS18305 (pirA) by ΦC31 Att/Int system-based vectors in spiramycin-producing strain Streptomyces ambofaciens ATCC 23877 resulted in marked effects on central carbon and energy metabolism gene expression, high sensitivity to oxidative injury and repression of polyketide antibiotic production. By using integrated transcriptomic, proteomic and metabolite profiling, together with genetic complementation, we here show that most of these effects could be traced to the inability of the pirA-defective strain to modulate beta-oxidation pathway, leading to an unbalanced supply of precursor monomers for polyketide biosynthesis. Indeed, in silico protein-protein interaction modeling and in vitro experimental validation allowed us to demonstrate that PirA is a novel redox-sensitive negative modulator of very long-chain acyl-CoA dehydrogenase, which catalyzes the first committed step of the beta-oxidation pathway.
Collapse
Affiliation(s)
- Adelfia Talà
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Fabrizio Damiano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Giuseppe Gallo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy; Advanced Technologies Network (ATeN) Center, University of Palermo, Palermo, Italy
| | - Eva Pinatel
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Matteo Calcagnile
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Mariangela Testini
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Daniela Fico
- Laboratory of Analytical and Isotopic Mass Spectrometry, Department of Cultural Heritage, University of Salento, Lecce, Italy
| | - Daniela Rizzo
- Laboratory of Analytical and Isotopic Mass Spectrometry, Department of Cultural Heritage, University of Salento, Lecce, Italy
| | - Alberto Sutera
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy; Advanced Technologies Network (ATeN) Center, University of Palermo, Palermo, Italy
| | - Giovanni Renzone
- Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, Naples, Italy
| | - Andrea Scaloni
- Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, Naples, Italy
| | - Gianluca De Bellis
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Luisa Siculella
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Giuseppe Egidio De Benedetto
- Laboratory of Analytical and Isotopic Mass Spectrometry, Department of Cultural Heritage, University of Salento, Lecce, Italy
| | - Anna Maria Puglia
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Clelia Peano
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy; Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, Rozzano, Milan, Italy; Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Pietro Alifano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy.
| |
Collapse
|
92
|
Yang SC, Yu GE, Ha J, Kwon S, Hwang JH, Park DH, Kang DG, Kim TW, Park HC, An SM, Kim CW. Association between an electron transfer flavoprotein alpha subunit polymorphism (rs321948383) and the meat quality of Berkshire pigs. JOURNAL OF APPLIED ANIMAL RESEARCH 2018. [DOI: 10.1080/09712119.2018.1481856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Seung Chang Yang
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | - Go Eun Yu
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | - Jeongim Ha
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | - Seulgi Kwon
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | - Jung Hye Hwang
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | - Da Hye Park
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | - Deok Gyeong Kang
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | - Tae Wan Kim
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | | | - Sang Mi An
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| | - Chul Wook Kim
- Swine Science and Technology Center, Gyeongnam National University of Science & Technology, Jinju, South Korea
| |
Collapse
|
93
|
Li ZQ, Yu H, Huang GH. Changes in lipid, protein and carbohydrate metabolism in Spodoptera exigua larvae associated with infection by Heliothis virescens ascovirus 3h. J Invertebr Pathol 2018; 155:55-63. [PMID: 29778741 DOI: 10.1016/j.jip.2018.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 05/11/2018] [Accepted: 05/16/2018] [Indexed: 12/30/2022]
Abstract
Spodoptera exigua (Lepidoptera: Noctuidae) larvae were used to analyze the regulation of lipid, protein and carbohydrate metabolism in host larvae infected with the Heliothis virescens ascovirus 3h (HvAV-3h). Using histological sections, significant pathological changes were found in the fat bodies of infected larvae from 24 h to 72 h post-infection (hpi). The lipid and protein contents of the infected larvae were significantly higher than those of the uninfected larvae, while the carbohydrate content of the infected larvae was significantly lower than that of the mock-infected larvae. The selected primary metabolite metabolism-associated genes showed different expression patterns. Further co-relationship analysis of the gene expression level and content changes of primary metabolites indicated the following: the acyl-CoA dehydrogenase and fatty acid synthase genes were closely associated with lipid metabolism, and the hexokinase and the glycogen synthase gene expression levels were related to carbohydrate metabolism, while the aminopeptidase N and the protein disulfide isomerase gene expression levels were not correlated with protein metabolism. These results indicate that the HvAV-3h virus stimulates host larval lipid and protein syntheses and inhibits carbohydrate synthesis.
Collapse
Affiliation(s)
- Zi-Qi Li
- Hunan Provincial Key Laboratory for Biology and Control of Plant Diseases and Insect Pests, Hunan Agricultural University, Changsha 410128, China
| | - Huan Yu
- Hunan Provincial Key Laboratory for Biology and Control of Plant Diseases and Insect Pests, Hunan Agricultural University, Changsha 410128, China
| | - Guo-Hua Huang
- Hunan Provincial Key Laboratory for Biology and Control of Plant Diseases and Insect Pests, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
94
|
Herrou J, Czyż DM, Fiebig A, Willett JW, Kim Y, Wu R, Babnigg G, Crosson S. Molecular control of gene expression by Brucella BaaR, an IclR-type transcriptional repressor. J Biol Chem 2018; 293:7437-7456. [PMID: 29567835 PMCID: PMC5949995 DOI: 10.1074/jbc.ra118.002045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/19/2018] [Indexed: 01/06/2023] Open
Abstract
The general stress response sigma factor σE1 directly and indirectly regulates the transcription of dozens of genes that influence stress survival and host infection in the zoonotic pathogen Brucella abortus Characterizing the functions of σE1-regulated genes therefore would contribute to our understanding of B. abortus physiology and infection biology. σE1 indirectly activates transcription of the IclR family regulator Bab2_0215, but the function of this regulator remains undefined. Here, we present a structural and functional characterization of Bab2_0215, which we have named B rucella adipic acid-activated regulator (BaaR). We found that BaaR adopts a classic IclR-family fold and directly represses the transcription of two operons with predicted roles in carboxylic acid oxidation. BaaR binds two sites on chromosome II between baaR and a divergently transcribed hydratase/dehydrogenase (acaD2), and it represses transcription of both genes. We identified three carboxylic acids (adipic acid, tetradecanedioic acid, and ϵ-aminocaproic acid) and a lactone (ϵ-caprolactone) that enhance transcription from the baaR and acaD2 promoters. However, neither the activating acids nor caprolactone enhanced transcription by binding directly to BaaR. Induction of baaR transcription by adipic acid required the gene bab2_0213, which encodes a major facilitator superfamily transporter, suggesting that Bab2_0213 transports adipic acid across the inner membrane. We conclude that a suite of structurally related organic molecules activate transcription of genes repressed by BaaR. Our study provides molecular-level understanding of a gene expression program in B. abortus that is downstream of σE1.
Collapse
Affiliation(s)
- Julien Herrou
- Departments of Biochemistry and Molecular Biology, Chicago, Illinois 60637; Howard Taylor Ricketts Laboratory, University of Chicago, Argonne, Illinois 60439
| | - Daniel M Czyż
- Departments of Biochemistry and Molecular Biology, Chicago, Illinois 60637; Howard Taylor Ricketts Laboratory, University of Chicago, Argonne, Illinois 60439
| | - Aretha Fiebig
- Howard Taylor Ricketts Laboratory, University of Chicago, Argonne, Illinois 60439
| | - Jonathan W Willett
- Departments of Biochemistry and Molecular Biology, Chicago, Illinois 60637; Howard Taylor Ricketts Laboratory, University of Chicago, Argonne, Illinois 60439
| | | | - Ruiying Wu
- Argonne National Laboratory, Argonne, Illinois 60439
| | | | - Sean Crosson
- Departments of Biochemistry and Molecular Biology, Chicago, Illinois 60637; Howard Taylor Ricketts Laboratory, University of Chicago, Argonne, Illinois 60439; Microbiology, University of Chicago, Chicago, Illinois 60637.
| |
Collapse
|
95
|
Huang Y, Zhang X, Zhao C, Zhuang X, Zhu L, Guo C, Song Y. Improvement of Spinosad Production upon Utilization of Oils and Manipulation of β-Oxidation in a High-Producing Saccharopolyspora spinosa Strain. J Mol Microbiol Biotechnol 2018; 28:53-64. [PMID: 29730661 DOI: 10.1159/000487854] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/20/2018] [Indexed: 11/19/2022] Open
Abstract
Spinosad, a member of polyketide-derived macrolides produced in the actinomycete Saccharopolyspora spinosa, has been developed as a broad-spectrum and effective insecticide. The β-oxidation pathway could be an important source of building blocks for the biosynthesis of spinosad, thus the effect of vegetable oils on the production of spinosad in a high-yield strain was investigated. The spinosad production increased significantly with the addition of strawberry seed oil (511.64 mg/L) and camellia oil (520.07 mg/L) compared to the control group without oil (285.76 mg/L) and soybean oil group (398.11 mg/L). It also revealed that the addition of oils would affect the expression of genes involved in fatty acid metabolism, precursor supply, and oxidative stress. The genetically engineered strain, in which fadD1 and fadE genes of Streptomyces coelicolor were inserted, produced spinosad up to 784.72 mg/L in the medium containing camellia oil, while a higher spinosad production level (843.40 mg/L) was detected with the addition of 0.01 mM of thiourea.
Collapse
Affiliation(s)
- Ying Huang
- Key Laboratory of Soil Microbiology, Ministry of Agriculture, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiaolin Zhang
- Academy of State Administration of Grain, Beijing, China
| | - Chen Zhao
- Academy of State Administration of Grain, Beijing, China
| | - Xuhui Zhuang
- Academy of State Administration of Grain, Beijing, China
| | - Lin Zhu
- Academy of State Administration of Grain, Beijing, China
| | - Chao Guo
- Academy of State Administration of Grain, Beijing, China
| | - Yuan Song
- Key Laboratory of Soil Microbiology, Ministry of Agriculture, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
96
|
The Uptake and Metabolism of Amino Acids, and Their Unique Role in the Biology of Pathogenic Trypanosomatids. Pathogens 2018; 7:pathogens7020036. [PMID: 29614775 PMCID: PMC6027508 DOI: 10.3390/pathogens7020036] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/28/2018] [Accepted: 03/29/2018] [Indexed: 01/24/2023] Open
Abstract
Trypanosoma brucei, as well as Trypanosoma cruzi and more than 20 species of the genus Leishmania, form a group of flagellated protists that threaten human health. These organisms are transmitted by insects that, together with mammals, are their natural hosts. This implies that during their life cycles each of them faces environments with different physical, chemical, biochemical, and biological characteristics. In this work we review how amino acids are obtained from such environments, how they are metabolized, and how they and some of their intermediate metabolites are used as a survival toolbox to cope with the different conditions in which these parasites should establish the infections in the insects and mammalian hosts.
Collapse
|
97
|
Augustin P, Toplak M, Fuchs K, Gerstmann EC, Prassl R, Winkler A, Macheroux P. Oxidation of the FAD cofactor to the 8-formyl-derivative in human electron-transferring flavoprotein. J Biol Chem 2018; 293:2829-2840. [PMID: 29301933 PMCID: PMC5827430 DOI: 10.1074/jbc.ra117.000846] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/14/2017] [Indexed: 11/06/2022] Open
Abstract
The heterodimeric human (h) electron-transferring flavoprotein (ETF) transfers electrons from at least 13 different flavin dehydrogenases to the mitochondrial respiratory chain through a non-covalently bound FAD cofactor. Here, we describe the discovery of an irreversible and pH-dependent oxidation of the 8α-methyl group to 8-formyl-FAD (8f-FAD), which represents a unique chemical modification of a flavin cofactor in the human flavoproteome. Furthermore, a set of hETF variants revealed that several conserved amino acid residues in the FAD-binding pocket of electron-transferring flavoproteins are required for the conversion to the formyl group. Two of the variants generated in our study, namely αR249C and αT266M, cause glutaric aciduria type II, a severe inherited disease. Both of the variants showed impaired formation of 8f-FAD shedding new light on the potential molecular cause of disease development. Interestingly, the conversion of FAD to 8f-FAD yields a very stable flavin semiquinone that exhibited slightly lower rates of electron transfer in an artificial assay system than hETF containing FAD. In contrast, the formation of 8f-FAD enhanced the affinity to human dimethylglycine dehydrogenase 5-fold, indicating that formation of 8f-FAD modulates the interaction of hETF with client enzymes in the mitochondrial matrix. Thus, we hypothesize that the FAD cofactor bound to hETF is subject to oxidation in the alkaline (pH 8) environment of the mitochondrial matrix, which may modulate electron transport between client dehydrogenases and the respiratory chain. This discovery challenges the current concepts of electron transfer processes in mitochondria.
Collapse
Affiliation(s)
- Peter Augustin
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/II
| | - Marina Toplak
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/II
| | - Katharina Fuchs
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/II
| | | | - Ruth Prassl
- Institute of Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6/IV, 8010 Graz, Austria
| | - Andreas Winkler
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/II
| | - Peter Macheroux
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/II.
| |
Collapse
|
98
|
Romero E, Gómez Castellanos JR, Gadda G, Fraaije MW, Mattevi A. Same Substrate, Many Reactions: Oxygen Activation in Flavoenzymes. Chem Rev 2018; 118:1742-1769. [DOI: 10.1021/acs.chemrev.7b00650] [Citation(s) in RCA: 216] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Elvira Romero
- Molecular Enzymology Group, University of Groningen, Nijenborgh 4, 9747AG Groningen, The Netherlands
| | - J. Rubén Gómez Castellanos
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy
| | - Giovanni Gadda
- Departments of Chemistry and Biology, Center for Diagnostics and Therapeutics, and Center for Biotechnology and Drug Design, Georgia State University, Atlanta, Georgia 30302-3965, United States
| | - Marco W. Fraaije
- Molecular Enzymology Group, University of Groningen, Nijenborgh 4, 9747AG Groningen, The Netherlands
| | - Andrea Mattevi
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy
| |
Collapse
|
99
|
Molecular Basis for Converting (2S)-Methylsuccinyl-CoA Dehydrogenase into an Oxidase. Molecules 2017; 23:molecules23010068. [PMID: 29283425 PMCID: PMC6017585 DOI: 10.3390/molecules23010068] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/18/2017] [Accepted: 12/21/2017] [Indexed: 11/17/2022] Open
Abstract
Although flavoenzymes have been studied in detail, the molecular basis of their dioxygen reactivity is only partially understood. The members of the flavin adenosine dinucleotide (FAD)-dependent acyl-CoA dehydrogenase and acyl-CoA oxidase families catalyze similar reactions and share common structural features. However, both enzyme families feature opposing reaction specificities in respect to dioxygen. Dehydrogenases react with electron transfer flavoproteins as terminal electron acceptors and do not show a considerable reactivity with dioxygen, whereas dioxygen serves as a bona fide substrate for oxidases. We recently engineered (2S)-methylsuccinyl-CoA dehydrogenase towards oxidase activity by rational mutagenesis. Here we characterized the (2S)-methylsuccinyl-CoA dehydrogenase wild-type, as well as the engineered (2S)-methylsuccinyl-CoA oxidase, in detail. Using stopped-flow UV-spectroscopy and liquid chromatography-mass spectrometry (LC-MS) based assays, we explain the molecular base for dioxygen reactivity in the engineered oxidase and show that the increased oxidase function of the engineered enzyme comes at a decreased dehydrogenase activity. Our findings add to the common notion that an increased activity for a specific substrate is achieved at the expense of reaction promiscuity and provide guidelines for rational engineering efforts of acyl-CoA dehydrogenases and oxidases.
Collapse
|
100
|
Homoacetogenesis in Deep-Sea Chloroflexi, as Inferred by Single-Cell Genomics, Provides a Link to Reductive Dehalogenation in Terrestrial Dehalococcoidetes. mBio 2017; 8:mBio.02022-17. [PMID: 29259088 PMCID: PMC5736913 DOI: 10.1128/mbio.02022-17] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The deep marine subsurface is one of the largest unexplored biospheres on Earth and is widely inhabited by members of the phylum Chloroflexi. In this report, we investigated genomes of single cells obtained from deep-sea sediments of the Peruvian Margin, which are enriched in such Chloroflexi. 16S rRNA gene sequence analysis placed two of these single-cell-derived genomes (DscP3 and Dsc4) in a clade of subphylum I Chloroflexi which were previously recovered from deep-sea sediment in the Okinawa Trough and a third (DscP2-2) as a member of the previously reported DscP2 population from Peruvian Margin site 1230. The presence of genes encoding enzymes of a complete Wood-Ljungdahl pathway, glycolysis/gluconeogenesis, a Rhodobacter nitrogen fixation (Rnf) complex, glyosyltransferases, and formate dehydrogenases in the single-cell genomes of DscP3 and Dsc4 and the presence of an NADH-dependent reduced ferredoxin:NADP oxidoreductase (Nfn) and Rnf in the genome of DscP2-2 imply a homoacetogenic lifestyle of these abundant marine Chloroflexi. We also report here the first complete pathway for anaerobic benzoate oxidation to acetyl coenzyme A (CoA) in the phylum Chloroflexi (DscP3 and Dsc4), including a class I benzoyl-CoA reductase. Of remarkable evolutionary significance, we discovered a gene encoding a formate dehydrogenase (FdnI) with reciprocal closest identity to the formate dehydrogenase-like protein (complex iron-sulfur molybdoenzyme [CISM], DET0187) of terrestrial Dehalococcoides/Dehalogenimonas spp. This formate dehydrogenase-like protein has been shown to lack formate dehydrogenase activity in Dehalococcoides/Dehalogenimonas spp. and is instead hypothesized to couple HupL hydrogenase to a reductive dehalogenase in the catabolic reductive dehalogenation pathway. This finding of a close functional homologue provides an important missing link for understanding the origin and the metabolic core of terrestrial Dehalococcoides/Dehalogenimonas spp. and of reductive dehalogenation, as well as the biology of abundant deep-sea Chloroflexi. The deep marine subsurface is one of the largest unexplored biospheres on Earth and is widely inhabited by members of the phylum Chloroflexi. In this report, we investigated genomes of single cells obtained from deep-sea sediments and provide evidence for a homacetogenic lifestyle of these abundant marine Chloroflexi. Moreover, genome signature and key metabolic genes indicate an evolutionary relationship between these deep-sea sediment microbes and terrestrial, reductively dehalogenating Dehalococcoides.
Collapse
|