1
|
Brett C, Gout I. The two faces of coenzyme A in cellular biology. Free Radic Biol Med 2025; 233:162-173. [PMID: 40107571 DOI: 10.1016/j.freeradbiomed.2025.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
Coenzyme A (CoA) is an essential cofactor present in all living cells, which plays critical roles in diverse biochemical processes, including cellular metabolism, signal transduction, regulation of gene expression, and the antioxidant response. This review summarizes current knowledge on the role of CoA and its metabolically active thioesters in promoting cellular growth and proliferation (pro-growth) and discusses emerging research on CoA's antioxidant properties that enhance cell survival (pro-survival).
Collapse
Affiliation(s)
- Charlie Brett
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, UK
| | - Ivan Gout
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
2
|
Garg R, Zhu Z, Hernandez FG, Wang Y, David MS, Bruno VM, Culotta VC. A response to iron involving carbon metabolism in the opportunistic fungal pathogen Candida albicans. mSphere 2025; 10:e0004025. [PMID: 40183578 PMCID: PMC12039268 DOI: 10.1128/msphere.00040-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/09/2025] [Indexed: 04/05/2025] Open
Abstract
Iron (Fe) is an essential micronutrient, and during infection, the host attempts to starve pathogens of this vital element through a process known as nutritional immunity. Successful pathogens have evolved means to evade this attack, an example being Candida albicans, the most prevalent human fungal pathogen. When Fe-starved, C. albicans induces multiple pathways for Fe uptake using the SEF1 trans-regulator, and we now describe a previously unrecognized effect of Fe on C. albicans metabolism that occurs independent of SEF1. Specifically, Fe limitation leads to inhibition of pyruvate dehydrogenase (PDH) connecting glycolysis to mitochondrial respiration. PDH inactivation involves loss of the LAT1 catalytic subunit harboring a lipoic acid co-factor. Protein lipoylation is a Fe-S dependent process, and lipoylated alpha-ketoglutarate dehydrogenase is also inhibited in Fe-starved C. albicans. SEF1 does not protect against PDH inactivation, and despite SEF1 induction of Fe import genes, cellular Fe levels drop dramatically during chronic Fe starvation. Such loss of LAT1 and lipoylation is also seen in Fe-starved bakers' yeast Saccharomyces cerevisiae. In both yeast species, glucose is diverted toward the pentose phosphate pathway (PPP) and PPP production of NADPH is increased in response to low Fe and PDH loss. Additionally, glucose consumption is lowered in Fe-starved C. albicans, and non-PDH alternatives to producing Ac-CoA are induced, including pyruvate bypass and fatty acid oxidation pathways. C. albicans can adapt well to the effects of micronutrient loss on cell metabolism. IMPORTANCE We describe a new response to Fe-starvation in a fungal pathogen involving carbon metabolism. Pyruvate dehydrogenase (PDH) that is central to glucose metabolism is inactivated at the post-translational level in Fe-starved cells. Nevertheless, the fungal pathogen can thrive by activating backup systems for metabolizing glucose. Methods that inhibit these compensatory pathways for carbon metabolism may prove beneficial in future anti-fungal strategies.
Collapse
Affiliation(s)
- Ritu Garg
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Zhengkai Zhu
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Francisco G. Hernandez
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Yiran Wang
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Marika S. David
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Vincent M. Bruno
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Valeria C. Culotta
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
4
|
Daems M, Ponomarev LC, Simoes-Faria R, Nobis M, Scheele CLGJ, Luttun A, Ghesquière B, Zwijsen A, Jones EAV. Smad1/5 is acetylated in the dorsal aortae of the mouse embryo before the onset of blood flow, driving early arterial gene expression. Cardiovasc Res 2024; 120:2078-2091. [PMID: 39253943 DOI: 10.1093/cvr/cvae201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 06/22/2024] [Indexed: 09/11/2024] Open
Abstract
AIMS During embryonic development, arteriovenous (AV) differentiation ensures proper blood vessel formation and maturation. Defects in arterial or venous identity cause inappropriate fusion of vessels, resulting in atypical shunts, so-called AV malformations (AVMs). Currently, the mechanism behind AVM formation remains unclear, and treatment options are fairly limited. Mammalian AV differentiation is initiated before the onset of blood flow in the embryo; however, this pre-flow mechanism is poorly understood. Here, we aimed to unravel the role of Smad1/5 signalling in pre-flow arterial identity and, in the process, uncovered an unexpected control mechanism of Smad1/5 signalling. METHODS AND RESULTS We establish that despite Notch1 being expressed in the pre-flow mouse embryo, it is not activated, nor is it necessary for the expression of the earliest arterial genes in the dorsal aortae (i.e. Hey1 and Gja4). Furthermore, interrupting blood flow by using the Ncx1 KO model completely prevents the activation of Notch1 signalling, suggesting a strong role of shear stress in maintaining arterial identity. We demonstrate that early expression of Hey1 and Gja4 requires SMAD1/5 signalling. Using embryo cultures, we show that Smad1/5 signalling is activated through the Alk1/Alk5/transforming growth factor (TGF)βR2 receptor complex, with TGFβ1 as a necessary ligand. Furthermore, our findings demonstrate that early arterial gene expression requires the acetylation of Smad1/5 proteins, rendering them more sensitive to TGFβ1 stimulation. Blocking acetyl-CoA production prevents pre-flow arterial expression of Hey1 and Gja4, while stabilizing acetylation rescues their expression. CONCLUSION Our findings highlight the importance of the acetyl-CoA production in the cell and provide a novel control mechanism of Smad1/5 signalling involving protein acetylation. As disturbed canonical Smad1/5 signalling is involved in several vascular conditions, our results offer new insights in treatment options for circumventing canonical Smad1/5 signalling.
Collapse
MESH Headings
- Animals
- Smad5 Protein/metabolism
- Smad5 Protein/genetics
- Smad1 Protein/metabolism
- Smad1 Protein/genetics
- Acetylation
- Gene Expression Regulation, Developmental
- Receptor, Notch1/metabolism
- Receptor, Notch1/genetics
- Mice, Knockout
- Regional Blood Flow
- Signal Transduction
- Aorta/metabolism
- Aorta/physiopathology
- Aorta/embryology
- Receptor, Transforming Growth Factor-beta Type I/metabolism
- Receptor, Transforming Growth Factor-beta Type I/genetics
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Protein Processing, Post-Translational
- Transforming Growth Factor beta1/metabolism
- Transforming Growth Factor beta1/genetics
- Mice, Inbred C57BL
- Receptors, Transforming Growth Factor beta/metabolism
- Receptors, Transforming Growth Factor beta/genetics
- Mice
- Sodium-Calcium Exchanger
Collapse
Affiliation(s)
- Margo Daems
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
| | - Ljuba C Ponomarev
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
| | - Rita Simoes-Faria
- Metabolomics Expertise Centre, VIB Centre for Cancer Biology, 3000 Leuven, Belgium
- Department of Cellular and Molecular Medicine, Laboratory of Applied Mass Spectrometry, KU Leuven, 3000 Leuven, Belgium
| | - Max Nobis
- Intravital Imaging Expertise Centre, VIB Centre for Cancer Biology, KU Leuven, 3000 Leuven, Belgium
| | - Colinda L G J Scheele
- Department of Oncology, Laboratory for Intravital Imaging and Dynamics of Tumour Progression, VIB Centre for Cancer Biology, KU Leuven, 3000 Leuven, Belgium
| | - Aernout Luttun
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
| | - Bart Ghesquière
- Metabolomics Expertise Centre, VIB Centre for Cancer Biology, 3000 Leuven, Belgium
- Department of Cellular and Molecular Medicine, Laboratory of Applied Mass Spectrometry, KU Leuven, 3000 Leuven, Belgium
| | - An Zwijsen
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
| | - Elizabeth A V Jones
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
5
|
Lu F, Zhang S, Dong S, Wang M, Pang K, Zhao Y, Huang J, Kang J, Liu N, Zhang X, Zhao D, Lu F, Zhang W. Exogenous hydrogen sulfide enhances myogenic differentiation of C2C12 myoblasts under high palmitate stress. Heliyon 2024; 10:e38661. [PMID: 39416846 PMCID: PMC11481675 DOI: 10.1016/j.heliyon.2024.e38661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024] Open
Abstract
Skeletal muscle atrophy was one of main complications of type 2 diabetes mellitus. Hydrogen sulfide (H2S) is involved in various physiological functions, such as anti-hypertension and anti-oxidant. Skeletal muscle atrophy caused by type 2 diabetes could lead to the regeneration of muscle fibers. Wnt signaling pathway plays a crucial important role in this process. H2S maybe regulate the Wnt signaling pathway to alleviate skeletal muscle atrophy, however, this role has not been clarified. The aim of this study is to investigate the potential regulatory role of H2S in the Wnt signaling pathway. C2C12 myoblasts treated with 500 μmol palmitate as an in vitro model. Western blot was used to detect the levels of CSE, PKM1, β-catenin, MuRF1, MYOG, MYF6 and MYOD1. In addition, MuRF1 was mutated at Cys44 and MuRF1 S-sulfhydration was detected by biotin switch assay. The interaction between PKM1 and MuRF1 was assessed via Co-immunoprecipitation. Differentiation of C2C12 myoblasts was evaluated using LAMININ staining. These data showed the levels of CSE, β-catenin, PKM1, MYOG, MYF6 and MYOD1 were decreased in pal group, compared with control and pal + NaHS groups. MuRF1 Cys44 mutants increased the protein levels of β-catenin, MYOG, MYF6 and MYOD1 in pal group. Our results suggest that H2S regulates the S-sulfhydration levels of MuRF1 at Cys44, influencing the ubiquitination levels of PKM1 and ultimately promoting myoblast differentiation.
Collapse
Affiliation(s)
- Fangping Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
- Department of Pathophysiology, Mudanjiang Medical University, Mudanjiang, China
| | - Shiwu Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shiyun Dong
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Mengyi Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Kemiao Pang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yajun Zhao
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Jiayi Huang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Jiaxin Kang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Ning Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Xueya Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Dechao Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fanghao Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| |
Collapse
|
6
|
Malanchuk O, Khoruzhenko A, Kosach V, Bdzhola A, Bidiuk D, Brett C, Gout I, Filonenko V. Immunofluorescent detection of protein CoAlation in mammalian cells under oxidative stress. Biol Open 2024; 13:bio061685. [PMID: 39344817 PMCID: PMC11463958 DOI: 10.1242/bio.061685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024] Open
Abstract
Previously, we reported the generation and characterisation of highly specific anti-CoA monoclonal antibodies capable of recognizing CoA in various immunological assays. Utilizing these antibodies in conjunction with mass spectrometry, we identified a wide array of cellular proteins modified by CoA in bacteria and mammalian cells. Furthermore, our findings demonstrated that such modifications could be induced by oxidative or metabolic stress. This study advances the utility of anti-CoA monoclonal antibodies in analysing protein CoAlation, highlighting their effectiveness in immunofluorescent assay. Our data corroborates a significant increase in cellular protein CoAlation induced by oxidative agents. Additionally, we observed that hydrogen-peroxide induced protein CoAlation is predominantly associated with mitochondrial proteins.
Collapse
Affiliation(s)
- Oksana Malanchuk
- Department of Cell Signalling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03143, Ukraine
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Antonina Khoruzhenko
- Department of Cell Signalling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03143, Ukraine
| | - Viktoriia Kosach
- Department of Cell Signalling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03143, Ukraine
- Department of Cell Screening, Enamine Ltd., Kyiv 02094, Ukraine
| | - Anna Bdzhola
- Department of Cell Signalling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03143, Ukraine
| | - Dariy Bidiuk
- Department of General Surgery, Danylo Halytsky Lviv National Medical University, Lviv 79000, Ukraine
| | - Charlie Brett
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Ivan Gout
- Department of Cell Signalling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03143, Ukraine
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Valeriy Filonenko
- Department of Cell Signalling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03143, Ukraine
| |
Collapse
|
7
|
Xie J, Yu Z, Zhu Y, Zheng M, Zhu Y. Functions of Coenzyme A and Acyl-CoA in Post-Translational Modification and Human Disease. FRONT BIOSCI-LANDMRK 2024; 29:331. [PMID: 39344325 DOI: 10.31083/j.fbl2909331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 05/24/2024] [Accepted: 07/17/2024] [Indexed: 10/01/2024]
Abstract
Coenzyme A (CoA) is synthesized from pantothenate, L-cysteine and adenosine triphosphate (ATP), and plays a vital role in diverse physiological processes. Protein acylation is a common post-translational modification (PTM) that modifies protein structure, function and interactions. It occurs via the transfer of acyl groups from acyl-CoAs to various amino acids by acyltransferase. The characteristics and effects of acylation vary according to the origin, structure, and location of the acyl group. Acetyl-CoA, formyl-CoA, lactoyl-CoA, and malonyl-CoA are typical acyl group donors. The major acyl donor, acyl-CoA, enables modifications that impart distinct biological functions to both histone and non-histone proteins. These modifications are crucial for regulating gene expression, organizing chromatin, managing metabolism, and modulating the immune response. Moreover, CoA and acyl-CoA play significant roles in the development and progression of neurodegenerative diseases, cancer, cardiovascular diseases, and other health conditions. The goal of this review was to systematically describe the types of commonly utilized acyl-CoAs, their functions in protein PTM, and their roles in the progression of human diseases.
Collapse
Affiliation(s)
- Jumin Xie
- Hubei Key Laboratory of Renal Disease Occurrence and Intervention, Medical School, Hubei Polytechnic University, 435003 Huangshi, Hubei, China
| | - Zhang Yu
- Hubei Key Laboratory of Renal Disease Occurrence and Intervention, Medical School, Hubei Polytechnic University, 435003 Huangshi, Hubei, China
| | - Ying Zhu
- Hubei Key Laboratory of Renal Disease Occurrence and Intervention, Medical School, Hubei Polytechnic University, 435003 Huangshi, Hubei, China
| | - Mei Zheng
- Hubei Key Laboratory of Renal Disease Occurrence and Intervention, Medical School, Hubei Polytechnic University, 435003 Huangshi, Hubei, China
| | - Yanfang Zhu
- Department of Critical Care Medicine, Huangshi Hospital of TCM (Infectious Disease Hospital), 435003 Huangshi, Hubei, China
| |
Collapse
|
8
|
Yoon KN, Yang J, Yeom SJ, Kim SS, Park JH, Song BS, Eun JB, Park SH, Lee JH, Kim HB, Lee JH, Kim JK. Lactiplantibacillus argentoratensis AGMB00912 protects weaning mice from ETEC infection and enhances gut health. Front Microbiol 2024; 15:1440134. [PMID: 39318427 PMCID: PMC11420142 DOI: 10.3389/fmicb.2024.1440134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Maintaining a healthy intestinal environment, optimal epithelial barrier integrity, and balanced gut microbiota composition are essential for the growth performance of weaning pigs. We identified Lactiplantibacillus argentoratensis AGMB00912 (LA) in healthy porcine feces as having antimicrobial activity against pathogens and enhanced short-chain fatty acid (SCFA) production. Herein, we assess the protective role of LA using a weaning mouse model with enterotoxigenic Escherichia coli (ETEC) infection. LA treatment improves feed intake and weight gain and alleviates colon shortening. Furthermore, LA inhibits intestinal damage, increases the small intestine villus height compared with the ETEC group, and enhances SCFA production. Using the Kyoto Encyclopedia of Genes and Genomes and other bioinformatic tools, including InterProScan and COGNIZER, we validated the presence of SCFA-producing pathways of LA and Lactiplantibacillus after whole genome sequencing. LA mitigates ETEC-induced shifts in the gut microbiota, decreasing the proportion of Escherichia and Enterococcus and increasing SCFA-producing bacteria, including Kineothrix, Lachnoclostridium, Roseuburia, Lacrimispora, Jutongia, and Blautia. Metabolic functional prediction analysis revealed enhanced functions linked to carbohydrate, amino acid, and vitamin biosynthesis, along with decreased functions associated with infectious bacterial diseases compared to the ETEC group. LA mitigates the adverse effects of ETEC infection in weaning mice, enhances growth performance and intestinal integrity, rebalances gut microbiota, and promotes beneficial metabolic functions. These findings validate the functionality of LA in a small animal model, supporting its potential application in improving the health and growth performance of weaning pigs.
Collapse
Affiliation(s)
- Ki-Nam Yoon
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
- Department of Food Science and Technology, Graduate School of Chonnam National University, Gwangju, Republic of Korea
| | - Jihye Yang
- Departments of Food and Animal Biotechnology and Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Seo-Joon Yeom
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Sang-Su Kim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Jong-Heum Park
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Beom-Seok Song
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Jong-Bang Eun
- Department of Food Science and Technology, Graduate School of Chonnam National University, Gwangju, Republic of Korea
| | - Seung-Hwan Park
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - Ju Huck Lee
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - Hyeun Bum Kim
- Department of Animal Resources Science, Dankook University, Cheonan, Republic of Korea
| | - Ju-Hoon Lee
- Departments of Food and Animal Biotechnology and Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Jae-Kyung Kim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| |
Collapse
|
9
|
Edwards A, Jiang Z, Nepogodiev S, Rejzek M, Martin C, Emmrich PMF. Reply to: LsBOS utilizes oxalyl-CoA produced by LsAAE3 to synthesize β-ODAP in grass pea. Nat Commun 2024; 15:6714. [PMID: 39117668 PMCID: PMC11310462 DOI: 10.1038/s41467-024-50705-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 07/18/2024] [Indexed: 08/10/2024] Open
Affiliation(s)
- Anne Edwards
- John Innes Centre, Norwich Research Park, Colney Lane, Norwich, NR4 7UH, UK
| | - Zhouqian Jiang
- John Innes Centre, Norwich Research Park, Colney Lane, Norwich, NR4 7UH, UK
| | - Sergey Nepogodiev
- John Innes Centre, Norwich Research Park, Colney Lane, Norwich, NR4 7UH, UK
| | - Martin Rejzek
- John Innes Centre, Norwich Research Park, Colney Lane, Norwich, NR4 7UH, UK
| | - Cathie Martin
- John Innes Centre, Norwich Research Park, Colney Lane, Norwich, NR4 7UH, UK
| | - Peter M F Emmrich
- John Innes Centre, Norwich Research Park, Colney Lane, Norwich, NR4 7UH, UK.
- Norwich Institute for Sustainable Development, School of Global Development, University of East Anglia, Norwich, NR4 7TJ, UK.
| |
Collapse
|
10
|
Wang WY, Bi JF, Hu JX, Li X. Metabolomics comparison of four varieties apple with different browning characters in response to pretreatment during pulp processing. Food Res Int 2024; 190:114600. [PMID: 38945570 DOI: 10.1016/j.foodres.2024.114600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024]
Abstract
Browning commonly appeared in apple processing, which varied in different apple varieties. Present work investigated the metabolomics of four varieties apple of Yataka, Gala, Sansa, and Fuji, which possessed different browning characteristics and related enzymes. Sansa as browning insensitive apple variety, exhibited the least chroma change with the lowest PPO activity and the highest SOD activity among the four apple varieties. Browning inhibition pretreatment increased the activity of SOD and PAL and decreased PPO and POD activity. In addition, metabolomic variances among the four apple varieties (FC), their browning pulp (BR) and browning inhibition pulp (CM) were compared. And the key metabolites were in-depth analyzed to match the relevant KEGG pathways and speculated metabolic networks. There were 487, 644, and 494 significant differential metabolites detected in FC, BR and CM, which were consisted of lipids, benzenoids, phenylpropanoids, organheterocyclic compounds, organic acids, nucleosides, accounting for 23 %, 11 %, 15 %, 16 %, 11 % of the total metabolites. The differential metabolites were matched with 39, 49, and 36 KEGG pathways in FC, BR, and CM, respectively, in which other secondary metabolites biosynthesis metabolism was the most significant in FC, lipid metabolism was the most significant in BR and CM, and energy metabolism was markedly annotated in CM. Notably, Sansa displayed the highest number of differential metabolites in both its BR (484) and CM (342). The BR of Sansa was characterized by flavonoid biosynthesis, while the other three apple varieties were associated with α-linolenic acid metabolism. Furthermore, in browning sensitive apple varieties, the flavonoid and phenylpropanoid biosynthesis pathway was significantly activated by browning inhibition pretreatment. Phenolic compounds, lipids, sugars, organic acids, nucleotides, and adenosine were regulated differently in the four apple varieties, potentially serving as key regulatory sites. Overall, this work provides novel insight for browning prevention in different apple varieties.
Collapse
Affiliation(s)
- Wen-Yue Wang
- Institute of Food Science and Technology, CAAS, Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Beijing 100193, China
| | - Jin-Feng Bi
- Institute of Food Science and Technology, CAAS, Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Beijing 100193, China.
| | - Jia-Xing Hu
- Institute of Food Science and Technology, CAAS, Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Beijing 100193, China
| | - Xuan Li
- Institute of Food Science and Technology, CAAS, Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Beijing 100193, China.
| |
Collapse
|
11
|
Yoon KN, Lee HG, Yeom SJ, Kim SS, Park JH, Song BS, Yi SW, Do YJ, Oh B, Oh SI, Eun JB, Park SH, Lee JH, Kim HB, Lee JH, Hur TY, Kim JK. Lactiplantibacillus argentoratensis AGMB00912 alleviates salmonellosis and modulates gut microbiota in weaned piglets: a pilot study. Sci Rep 2024; 14:15466. [PMID: 38965336 PMCID: PMC11224356 DOI: 10.1038/s41598-024-66092-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 06/27/2024] [Indexed: 07/06/2024] Open
Abstract
This study aimed to evaluate the efficacy of Lactiplantibacillus argentoratensis AGMB00912 (LA) in reducing Salmonella Typhimurium infection in weaned piglets. The investigation focused on the influence of LA on the gut microbiota composition, growth performance, and Salmonella fecal shedding. The results indicated that LA supplementation significantly improved average daily gain and reduced the prevalence and severity of diarrhea. Fecal analysis revealed reduced Salmonella shedding in the LA-supplemented group. Furthermore, LA notably altered the composition of the gut microbiota, increasing the levels of beneficial Bacillus and decreasing those of harmful Proteobacteria and Spirochaetes. Histopathological examination showed less intestinal damage in LA-treated piglets than in the controls. The study also observed that LA affected metabolic functions related to carbohydrate, amino acid, and fatty acid metabolism, thereby enhancing gut health and resilience against infection. Short-chain fatty acid concentrations in the feces were higher in the LA group, suggesting improved gut microbial activity. LA supplementation enriched the population of beneficial bacteria, including Streptococcus, Clostridium, and Bifidobacterium, while reducing the number of harmful bacteria, such as Escherichia and Campylobacter. These findings indicate the potential of LA as a probiotic alternative for swine nutrition, offering protective effects to the gut microbiota against Salmonella infection.
Collapse
Affiliation(s)
- Ki-Nam Yoon
- Research Division for Biotechnology, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, 29 Geumgu-gil, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
- Department of Food Science and Technology, Graduate School of Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Han Gyu Lee
- Division of Animal Diseases and Health, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, 55365, Republic of Korea
| | - Seo-Joon Yeom
- Research Division for Biotechnology, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, 29 Geumgu-gil, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
| | - Sang-Su Kim
- Research Division for Biotechnology, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, 29 Geumgu-gil, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
| | - Jong-Heum Park
- Research Division for Biotechnology, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, 29 Geumgu-gil, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
| | - Beom-Seok Song
- Research Division for Biotechnology, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, 29 Geumgu-gil, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
| | - Seung-Won Yi
- Division of Animal Diseases and Health, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, 55365, Republic of Korea
| | - Yoon Jung Do
- Division of Animal Diseases and Health, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, 55365, Republic of Korea
| | - Byungkwan Oh
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeollabuk-do, 54596, Republic of Korea
| | - Sang-Ik Oh
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeollabuk-do, 54596, Republic of Korea
| | - Jong-Bang Eun
- Department of Food Science and Technology, Graduate School of Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Seung-Hwan Park
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, 56212, Republic of Korea
| | - Ju Huck Lee
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, 56212, Republic of Korea
| | - Hyeun Bum Kim
- Department of Animal Resources Science, Dankook University, Cheonan, 31116, Republic of Korea
| | - Ju-Hoon Lee
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea
| | - Tai-Young Hur
- Division of Animal Diseases and Health, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, 55365, Republic of Korea.
| | - Jae-Kyung Kim
- Research Division for Biotechnology, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, 29 Geumgu-gil, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea.
| |
Collapse
|
12
|
Yuan H, Xun H, Wang J, Wang J, Yao X, Tang F. Integrated Metabolomic and Transcriptomic Analysis Reveals the Underlying Antibacterial Mechanisms of the Phytonutrient Quercetin-Induced Fatty Acids Alteration in Staphylococcus aureus ATCC 27217. Molecules 2024; 29:2266. [PMID: 38792126 PMCID: PMC11123838 DOI: 10.3390/molecules29102266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
The utilization of natural products in food preservation represents a promising strategy for the dual benefits of controlling foodborne pathogens and enhancing the nutritional properties of foods. Among the phytonutrients, flavonoids have been shown to exert antibacterial effects by disrupting bacterial cell membrane functionality; however, the underlying molecular mechanisms remain elusive. In this study, we investigated the effect of quercetin on the cell membrane permeability of Staphylococcus aureus ATCC 27217. A combined metabolomic and transcriptomic approach was adopted to examine the regulatory mechanism of quercetin with respect to the fatty acid composition and associated genes. Kinetic analysis and molecular docking simulations were conducted to assess quercetin's inhibition of β-ketoacyl-acyl carrier protein reductase (FabG), a potential target in the bacterial fatty acid biosynthesis pathway. Metabolomic and transcriptomic results showed that quercetin increased the ratio of unsaturated to saturated fatty acids and the levels of membrane phospholipids. The bacteria reacted to quercetin-induced stress by attempting to enhance fatty acid biosynthesis; however, quercetin directly inhibited FabG activity, thereby disrupting bacterial fatty acid biosynthesis. These findings provide new insights into the mechanism of quercetin's effects on bacterial cell membranes and suggest potential applications for quercetin in bacterial inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | - Feng Tang
- Key Laboratory of National Forestry and Grassland Administration/Beijing for Bamboo & Rattan Science and Technology, International Centre for Bamboo and Rattan, Beijing 100102, China; (H.Y.); (H.X.); (J.W.); (J.W.); (X.Y.)
| |
Collapse
|
13
|
Zheng Y, Liufu T, Wen B, Zhou C, Liu L, Qiu Y, Zou W, Zhang W, Li Y, Pei J, Zeng Y, Chen W, Zhang C, Yuan Y, Wang G, Yan C, Lu X, Deng J, Wang Z, Hong D. COASY variant as a new genetic cause of riboflavin-responsive lipid storage myopathy. Cell Discov 2024; 10:25. [PMID: 38413569 PMCID: PMC10899607 DOI: 10.1038/s41421-023-00641-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/22/2023] [Indexed: 02/29/2024] Open
Affiliation(s)
- Yilei Zheng
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Tongling Liufu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Bing Wen
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Chao Zhou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Lingchun Liu
- Department of Neurology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yusen Qiu
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Wenquan Zou
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Wei Zhang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yu Li
- Intelligence Pharma, Beijing, China
| | - Jianfeng Pei
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yiheng Zeng
- Department of Neurology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Wanjin Chen
- Department of Neurology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Chunhua Zhang
- Department of Research & Development of MILS International, Yokohama, Japan
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Guochun Wang
- Department of Rheumatology, China-Japan Friendship Hospital, Beijing, China
| | - Chuanzhu Yan
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xin Lu
- Department of Rheumatology, China-Japan Friendship Hospital, Beijing, China.
| | - Jianwen Deng
- Department of Neurology, Peking University First Hospital, Beijing, China.
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China.
| | - Daojun Hong
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
14
|
Scholefield M, Church SJ, Xu J, Patassini S, Cooper GJ. Localized Pantothenic Acid (Vitamin B5) Reductions Present Throughout the Dementia with Lewy Bodies Brain. JOURNAL OF PARKINSON'S DISEASE 2024; 14:965-976. [PMID: 38820022 PMCID: PMC11307062 DOI: 10.3233/jpd-240075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/10/2024] [Indexed: 06/02/2024]
Abstract
Background Localized pantothenic acid deficiencies have been observed in several neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease dementia (PDD), and Huntington's disease (HD), indicating downstream energetic pathway perturbations. However, no studies have yet been performed to see whether such deficiencies occur across the dementia with Lewy bodies (DLB) brain, or what the pattern of such dysregulation may be. Objective Firstly, this study aimed to quantify pantothenic acid levels across ten regions of the brain in order to determine the localization of any pantothenic acid dysregulation in DLB. Secondly, the localization of pantothenic acid alterations was compared to that previously in AD, PDD, and HD brains. Methods Pantothenic acid levels were determined in 20 individuals with DLB and 19 controls by ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) across ten brain regions. Case-control differences were determined by nonparametric Mann-Whitney U test, with the calculation of S-values, risk ratios, E-values, and effect sizes. The results were compared with those previously obtained in DLB, AD, and HD. Results Pantothenic acid levels were significantly decreased in six of the ten investigated brain regions: the pons, substantia nigra, motor cortex, middle temporal gyrus, primary visual cortex, and hippocampus. This level of pantothenic acid dysregulation is most similar to that of the AD brain, in which pantothenic acid is also decreased in the motor cortex, middle temporal gyrus, primary visual cortex, and hippocampus. DLB appears to differ from other neurodegenerative diseases in being the only of the four to not show pantothenic acid dysregulation in the cerebellum. Conclusions Pantothenic acid deficiency appears to be a shared mechanism of several neurodegenerative diseases, although differences in the localization of this dysregulation may contribute to the differing clinical pathways observed in these conditions.
Collapse
Affiliation(s)
- Melissa Scholefield
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Stephanie J. Church
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Jingshu Xu
- School of Biological Sciences, Faculty of Science, University of Auckland, Private Bag, Auckland, New Zealand
| | - Stefano Patassini
- School of Biological Sciences, Faculty of Science, University of Auckland, Private Bag, Auckland, New Zealand
| | - Garth J.S. Cooper
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- School of Biological Sciences, Faculty of Science, University of Auckland, Private Bag, Auckland, New Zealand
| |
Collapse
|
15
|
Yoshimoto H, Bogaki T. Mechanisms of production and control of acetate esters in yeasts. J Biosci Bioeng 2023; 136:261-269. [PMID: 37607842 DOI: 10.1016/j.jbiosc.2023.06.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 08/24/2023]
Abstract
Acetate esters, such as isoamyl acetate and ethyl acetate, are major aroma components of alcoholic beverages. They are produced through synthesis from acetyl CoA and the corresponding alcohol by alcohol acetyltransferase (AATase) with specific control of reaction factors, including unsaturated fatty acids and precursors, the percentage of nitrogen, and oxygen. However, the mechanisms by which these specific reaction factors affect acetate ester production remain largely unknown. The cellular mechanisms underlying the effects of these factors on acetate ester production were examined by purifying AATase from yeast, characterizing it, and cloning the ATF gene encoding AATase from sake yeast and bottom-fermenting yeast. Genetic and biochemical studies suggested that the decrease in acetate production with the addition of oxygen and unsaturated fatty acids was due to a decrease in enzyme synthesis resulting from transcriptional repression of the ATF1 gene, which is responsible for most of the AATase activity. Furthermore, these results suggest that expression of the ATF1 gene is intricately regulated by a number of transcriptional regulatory genes such as ROX1 and RAP1. Based on these results, the mechanism of ester regulation by oxygen, unsaturated fatty acids and precursors, and ratio of nitrogen source are becoming clearer from a molecular biological point of view. The physiological significance of ester production by yeast is then discussed. In this review, we summarize the studies on AATase, ATF gene, regulation of ester production, and physiological significance of acetate ester.
Collapse
Affiliation(s)
- Hiroyuki Yoshimoto
- Institute for Future Beverages, Research & Development Division, Kirin Holdings Company Limited, Technovilleage Center 3F, 1-17-1 Namamugi, Tsurumi-ku, Yokohama, Kanagawa 236-8628, Japan.
| | - Takayuki Bogaki
- General Research Laboratory, Ozeki Corporation, 4-9 Imazu Dezaike-cho, Nishinomiya, Hyogo 663-8227, Japan
| |
Collapse
|
16
|
Hansen AW, Venkatachalam KV. Sulfur-Element containing metabolic pathways in human health and crosstalk with the microbiome. Biochem Biophys Rep 2023; 35:101529. [PMID: 37601447 PMCID: PMC10439400 DOI: 10.1016/j.bbrep.2023.101529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/22/2023] Open
Abstract
In humans, methionine derived from dietary proteins is necessary for cellular homeostasis and regeneration of sulfur containing pathways, which produce inorganic sulfur species (ISS) along with essential organic sulfur compounds (OSC). In recent years, inorganic sulfur species have gained attention as key players in the crosstalk of human health and the gut microbiome. Endogenously, ISS includes hydrogen sulfide (H2S), sulfite (SO32-), thiosulfate (S2O32-), and sulfate (SO42-), which are produced by enzymes in the transsulfuration and sulfur oxidation pathways. Additionally, sulfate-reducing bacteria (SRB) in the gut lumen are notable H2S producers which can contribute to the ISS pools of the human host. In this review, we will focus on the systemic effects of sulfur in biological pathways, describe the contrasting mechanisms of sulfurylation versus phosphorylation on the hydroxyl of serine/threonine and tyrosine residues of proteins in post-translational modifications, and the role of the gut microbiome in human sulfur metabolism.
Collapse
Affiliation(s)
- Austin W. Hansen
- College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, 33328, USA
| | | |
Collapse
|
17
|
Carrico C, Cruz A, Walter M, Meyer J, Wehrfritz C, Shah S, Wei L, Schilling B, Verdin E. Coenzyme A binding sites induce proximal acylation across protein families. Sci Rep 2023; 13:5029. [PMID: 36977698 PMCID: PMC10050154 DOI: 10.1038/s41598-023-31900-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Lysine Nɛ-acylations, such as acetylation or succinylation, are post-translational modifications that regulate protein function. In mitochondria, lysine acylation is predominantly non-enzymatic, and only a specific subset of the proteome is acylated. Coenzyme A (CoA) can act as an acyl group carrier via a thioester bond, but what controls the acylation of mitochondrial lysines remains poorly understood. Using published datasets, here we found that proteins with a CoA-binding site are more likely to be acetylated, succinylated, and glutarylated. Using computational modeling, we show that lysine residues near the CoA-binding pocket are highly acylated compared to those farther away. We hypothesized that acyl-CoA binding enhances acylation of nearby lysine residues. To test this hypothesis, we co-incubated enoyl-CoA hydratase short chain 1 (ECHS1), a CoA-binding mitochondrial protein, with succinyl-CoA and CoA. Using mass spectrometry, we found that succinyl-CoA induced widespread lysine succinylation and that CoA competitively inhibited ECHS1 succinylation. CoA-induced inhibition at a particular lysine site correlated inversely with the distance between that lysine and the CoA-binding pocket. Our study indicated that CoA acts as a competitive inhibitor of ECHS1 succinylation by binding to the CoA-binding pocket. Together, this suggests that proximal acylation at CoA-binding sites is a primary mechanism for lysine acylation in the mitochondria.
Collapse
Affiliation(s)
- Chris Carrico
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Andrew Cruz
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Marius Walter
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Jesse Meyer
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | | | - Samah Shah
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Lei Wei
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, 94945, USA.
| |
Collapse
|
18
|
Xu M, Pan L, Wang B, Zou X, Zhang A, Zhou Z, Han Y. Simulated Digestion and Fecal Fermentation Behaviors of Levan and Its Impacts on the Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:1531-1546. [PMID: 36622938 DOI: 10.1021/acs.jafc.2c06897] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Levan is a microbial fructan widely explored in various fields owing to its excellent physical and biochemical properties. However, little is known about its digestion and fermentation characteristics in vitro. This study evaluated the potential prebiotic properties of levan obtained by enzymatic synthesis. Scanning electron microscopy, Fourier transform infrared spectroscopy, and nuclear magnetic resonance spectroscopy showed that the primary structures of levan remained stable after saliva-gastrointestinal digestion. The microtopography, molecular weight, and functional group of levan were seriously damaged during fecal fermentation. Moreover, the total short-chain fatty acid levels increased significantly, especially for propionic acid, butyric acid, and valeric acid. The 16S rDNA sequencing showed that levan mainly increased the abundance of Firmicutes; in genus levels, certain beneficial bacteria such as Megasphaera and Megamonas genera were remarkably promoted, and the proliferation of harmful genera was inhibited (such as Cedecea and Klebsiella). Overall, this study provided new insights into the potential probiotic mechanism of levan.
Collapse
Affiliation(s)
- Min Xu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, Puerto Rico 300350, United States
| | - Lei Pan
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, Puerto Rico 300350, United States
| | - Binbin Wang
- School of Life Science, Shanxi Normal University, Taiyuan 030000, China
| | - Xuan Zou
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, Puerto Rico 300350, United States
| | - Aihua Zhang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, Puerto Rico 300350, United States
| | - Zhijiang Zhou
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, Puerto Rico 300350, United States
| | - Ye Han
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, Puerto Rico 300350, United States
| |
Collapse
|
19
|
Filonenko V, Gout I. Discovery and functional characterisation of protein CoAlation and the antioxidant function of coenzyme A. BBA ADVANCES 2023; 3:100075. [PMID: 37082257 PMCID: PMC10074942 DOI: 10.1016/j.bbadva.2023.100075] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Coenzyme A (CoA) is an essential cofactor in all living cells which plays critical role in cellular metabolism, the regulation of gene expression and the biosynthesis of major cellular constituents. Recently, CoA was found to function as a major antioxidant in both prokaryotic and eukaryotic cells. This unconventional function of CoA is mediated by a novel post-translational modification, termed protein CoAlation. This review will highlight the history of this discovery, current knowledge, and future directions on studying molecular mechanisms of protein CoAlation and whether the antioxidant function of CoA is associated with pathologies, such as neurodegeneration and cancer.
Collapse
Affiliation(s)
- Valeriy Filonenko
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03680, Ukraine
- Corresponding authors.
| | - Ivan Gout
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03680, Ukraine
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, United Kingdom
- Corresponding authors.
| |
Collapse
|
20
|
Matye D, Gunewardena S, Chen J, Wang H, Wang Y, Hasan MN, Gu L, Clayton YD, Du Y, Chen C, Friedman JE, Lu SC, Ding WX, Li T. TFEB regulates sulfur amino acid and coenzyme A metabolism to support hepatic metabolic adaptation and redox homeostasis. Nat Commun 2022; 13:5696. [PMID: 36171419 PMCID: PMC9519740 DOI: 10.1038/s41467-022-33465-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 09/15/2022] [Indexed: 11/15/2022] Open
Abstract
Fatty liver is a highly heterogenous condition driven by various pathogenic factors in addition to the severity of steatosis. Protein insufficiency has been causally linked to fatty liver with incompletely defined mechanisms. Here we report that fatty liver is a sulfur amino acid insufficient state that promotes metabolic inflexibility via limiting coenzyme A availability. We demonstrate that the nutrient-sensing transcriptional factor EB synergistically stimulates lysosome proteolysis and methionine adenosyltransferase to increase cysteine pool that drives the production of coenzyme A and glutathione, which support metabolic adaptation and antioxidant defense during increased lipid influx. Intriguingly, mice consuming an isocaloric protein-deficient Western diet exhibit selective hepatic cysteine, coenzyme A and glutathione deficiency and acylcarnitine accumulation, which are reversed by cystine supplementation without normalizing dietary protein intake. These findings support a pathogenic link of dysregulated sulfur amino acid metabolism to metabolic inflexibility that underlies both overnutrition and protein malnutrition-associated fatty liver development.
Collapse
Affiliation(s)
- David Matye
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Sumedha Gunewardena
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jianglei Chen
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Huaiwen Wang
- Laboratory for Molecular Biology and Cytometry Research, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yifeng Wang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Mohammad Nazmul Hasan
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Lijie Gu
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yung Dai Clayton
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yanhong Du
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Cheng Chen
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Jacob E Friedman
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Shelly C Lu
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Tiangang Li
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
21
|
Generation and Validation of an Anti-Human PANK3 Mouse Monoclonal Antibody. Biomolecules 2022; 12:biom12091323. [PMID: 36139163 PMCID: PMC9496473 DOI: 10.3390/biom12091323] [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: 08/19/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/19/2022] Open
Abstract
Coenzyme A (CoA) is an essential co-factor at the intersection of diverse metabolic pathways. Cellular CoA biosynthesis is regulated at the first committed step—phosphorylation of pantothenic acid—catalyzed by pantothenate kinases (PANK1,2,3 in humans, PANK3 being the most highly expressed). Despite the critical importance of CoA in metabolism, the differential roles of PANK isoforms remain poorly understood. Our investigations of PANK proteins as potential precision oncology collateral lethality targets (PANK1 is co-deleted as part of the PTEN locus in some highly aggressive cancers) were severely hindered by a dearth of commercial antibodies that can reliably detect endogenous PANK3 protein. While we successfully validated commercial antibodies for PANK1 and PANK2 using CRISPR knockout cell lines, we found no commercial antibody that could detect endogenous PANK3. We therefore set out to generate a mouse monoclonal antibody against human PANK3 protein. We demonstrate that a clone (Clone MDA-299-62A) can reliably detect endogenous PANK3 protein in cancer cell lines, with band-specificity confirmed by CRISPR PANK3 knockout and knockdown cell lines. Sub-cellular fractionation shows that PANK3 is overwhelmingly cytosolic and expressed broadly across cancer cell lines. PANK3 monoclonal antibody MDA-299-62A should prove a valuable tool for researchers investigating this understudied family of metabolic enzymes in health and disease.
Collapse
|
22
|
Han Y, Quan X, Chuang Y, Liang Q, Li Y, Yuan Z, Bian Y, Wei L, Wang J, Zhao Y. A multi-omics analysis for the prediction of neurocognitive disorders risk among the elderly in Macao. Clin Transl Med 2022; 12:e909. [PMID: 35696554 PMCID: PMC9191869 DOI: 10.1002/ctm2.909] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Due to the increasing ageing population, neurocognitive disorders (NCDs) have been a global public health issue, and its prevention and early diagnosis are crucial. Our previous study demonstrated that there is a significant correlation between specific populations and NCDs, but the biological characteristics of the vulnerable group predispose to NCDs are unclear. The purpose of this study is to investigate the predictors for the vulnerable group by a multi-omics analysis. METHODS Multi-omics approaches, including metagenomics, metabolomic and proteomic, were used to detect gut microbiota, faecal metabolites and urine exosome of 8 normal controls and 13 vulnerable elders after a rigorous screening of 400 elders in Macao. The multi-omics data were analysed using R and Bioconductor. The two-sided Wilcoxon's rank-sum test, Kruskal-Wallis rank sum test and the linear discriminant analysis effective size were applied to investigate characterized features. Moreover, a 2-year follow-up was conducted to evaluate cognitive function change of the elderly. RESULTS Compared with the control elders, the metagenomics of gut microbiota showed that Ruminococcus gnavus, Lachnospira eligens, Escherichia coli and Desulfovibrio piger were increased significantly in the vulnerable group. Carboxylates, like alpha-ketoglutaric acid and d-saccharic acid, and levels of vitamins had obvious differences in the faecal metabolites. There was a distinct decrease in the expression of eukaryotic translation initiation factor 2 subunit 1 (eIF2α) and amine oxidase A (MAO-A) according to the proteomic results of the urine exosomes. Moreover, the compound annual growth rate of neurocognitive scores was notably decreased in vulnerable elders. CONCLUSIONS The multi-omics characteristics of disturbed glyoxylate and dicarboxylate metabolism (bacteria), vitamin digestion and absorption and tricarboxylic acid cycle in vulnerable elders can serve as predictors of NCDs risk among the elderly of Macao. Intervention with them may be effective therapeutic approaches for NCDs, and the underlying mechanisms merit further exploration.
Collapse
Affiliation(s)
- Yan Han
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauTaipaMacao SARChina
| | - Xingping Quan
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauTaipaMacao SARChina
| | | | - Qiaoxing Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Yang Li
- Department of Gastrointestinal SurgerySecond Clinical Medical College of Jinan University, Shenzhen People's HospitalShenzhenChina
| | - Zhen Yuan
- Centre for Cognitive and Brain SciencesUniversity of MacauTaipaMacao SARChina
| | - Ying Bian
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauTaipaMacao SARChina
| | - Lai Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Ji Wang
- School of Traditional Chinese MedicineBeijing University of Chinese MedicineBeijingChina
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauTaipaMacao SARChina
| |
Collapse
|
23
|
Zhou S, Alper HS, Zhou J, Deng Y. Intracellular biosensor-based dynamic regulation to manipulate gene expression at the spatiotemporal level. Crit Rev Biotechnol 2022; 43:646-663. [PMID: 35450502 DOI: 10.1080/07388551.2022.2040415] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The use of intracellular, biosensor-based dynamic regulation strategies to regulate and improve the production of useful compounds have progressed significantly over previous decades. By employing such an approach, it is possible to simultaneously realize high productivity and optimum growth states. However, industrial fermentation conditions contain a mixture of high- and low-performance non-genetic variants, as well as young and aged cells at all growth phases. Such significant individual variations would hinder the precise controlling of metabolic flux at the single-cell level to achieve high productivity at the macroscopic population level. Intracellular biosensors, as the regulatory centers of metabolic networks, can real-time sense intra- and extracellular conditions and, thus, could be synthetically adapted to balance the biomass formation and overproduction of compounds by individual cells. Herein, we highlight advances in the designing and engineering approaches to intracellular biosensors. Then, the spatiotemporal properties of biosensors associated with the distribution of inducers are compared. Also discussed is the use of such biosensors to dynamically control the cellular metabolic flux. Such biosensors could achieve single-cell regulation or collective regulation goals, depending on whether or not the inducer distribution is only intracellular.
Collapse
Affiliation(s)
- Shenghu Zhou
- National Engineering Laboratory for Cereal Fermentation Technology (NELCF), Jiangnan University, Wuxi, Jiangsu, China.,Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Hal S Alper
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA.,McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Jingwen Zhou
- National Engineering Laboratory for Cereal Fermentation Technology (NELCF), Jiangnan University, Wuxi, Jiangsu, China.,Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Yu Deng
- National Engineering Laboratory for Cereal Fermentation Technology (NELCF), Jiangnan University, Wuxi, Jiangsu, China.,Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
24
|
Dobrzyn P. CoA in Health and Disease. Int J Mol Sci 2022; 23:ijms23084371. [PMID: 35457189 PMCID: PMC9026968 DOI: 10.3390/ijms23084371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/10/2022] [Indexed: 12/10/2022] Open
Abstract
Coenzyme A (CoA) and its thioester derivatives are crucial components of numerous biosynthetic and degradative pathways of the cellular metabolism (including fatty acid synthesis and oxidation, the Krebs cycle, ketogenesis, cholesterol and acetylcholine biosynthesis, amino acid degradation, and neurotransmitter biosynthesis), post-translational modifications of proteins, and the regulation of gene expression [...].
Collapse
Affiliation(s)
- Pawel Dobrzyn
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| |
Collapse
|
25
|
Mizerska-Wasiak M, Płatos E, Cichoń-Kawa K, Demkow U, Pańczyk-Tomaszewska M. The Usefulness of Vanin-1 and Periostin as Markers of an Active Autoimmune Process or Renal Fibrosis in Children with IgA Nephropathy and IgA Vasculitis with Nephritis-A Pilot Study. J Clin Med 2022; 11:jcm11051265. [PMID: 35268356 PMCID: PMC8911128 DOI: 10.3390/jcm11051265] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 01/25/2023] Open
Abstract
This study aimed to evaluate the usefulness of vanin-1 and periostin in urine as markers of the autoimmune process in kidneys and renal fibrosis in IgA nephropathy (IgAN) and IgA vasculitis with nephritis (IgAVN). From a group of 194 patients from the Department of Pediatrics and Nephrology, who were included in the Polish Pediatric Registry of IgAN and IgAVN, we qualified 51 patients (20 with IgAN and 31 with IgAVN) between the ages of 3 and 17, diagnosed based on kidney biopsy, for inclusion in the study. All of the patients received glucocorticosteroids, immunosuppressive drugs, or renoprotective therapy. The control group consisted of 18 healthy individuals. The concentration of vanin was significantly higher in the IgAN and IgAVN groups than in the control group. The concentration of vanin/creatinine correlates positively with the level of IgA and negatively with the serum level of C3 at the end of the observation. Urinary vanin-1 concentration may be useful as a marker of the active autoimmune process in IgAN and IgAVN in children, but the study needs confirmation on a larger group of children, along with evaluation of the dynamics of this marker. Urinary periostin is not a good marker for children with IgAN and IgAVN, especially in stage 1 and 2 CKD.
Collapse
Affiliation(s)
- Małgorzata Mizerska-Wasiak
- Department of Pediatrics and Nephrology, Medical University of Warsaw, 02-091 Warsaw, Poland; (K.C.-K.); (M.P.-T.)
- Correspondence:
| | - Emilia Płatos
- Science Students’ Association at the Department of Pediatrics and Nephrology, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Karolina Cichoń-Kawa
- Department of Pediatrics and Nephrology, Medical University of Warsaw, 02-091 Warsaw, Poland; (K.C.-K.); (M.P.-T.)
| | - Urszula Demkow
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | | |
Collapse
|
26
|
Hala D, Petersen LH, Huggett DB, Puchowicz MA, Brunengraber H, Zhang GF. Overcompensation of CoA Trapping by Di(2-ethylhexyl) Phthalate (DEHP) Metabolites in Livers of Wistar Rats. Int J Mol Sci 2021; 22:ijms222413489. [PMID: 34948286 PMCID: PMC8709406 DOI: 10.3390/ijms222413489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022] Open
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is commonly used as a plasticizer in various industrial and household plastic products, ensuring widespread human exposures. Its routine detection in human bio-fluids and the propensity of its monoester metabolite to activate peroxisome proliferator activated receptor-α (PPARα) and perturb lipid metabolism implicate it as a metabolic disrupter. In this study we evaluated the effects of DEHP exposure on hepatic levels of free CoA and various CoA esters, while also confirming the metabolic activation to CoA esters and partial β-oxidation of a DEHP metabolite (2-ethyhexanol). Male Wistar rats were exposed via diet to 2% (w/w) DEHP for fourteen-days, following which hepatic levels of free CoA and various CoA esters were identified using liquid chromatography-mass spectrometry. DEHP exposed rats showed significantly elevated free CoA and increased levels of physiological, DEHP-derived and unidentified CoA esters. The physiological CoA ester of malonyl-CoA and DEHP-derived CoA ester of 3-keto-2-ethylhexanoyl-CoA were the most highly elevated, at eighteen- and ninety eight-times respectively. We also detected sixteen unidentified CoA esters which may be derivative of DEHP metabolism or induction of other intermediary metabolism metabolites. Our results demonstrate that DEHP is a metabolic disrupter which affects production and sequestration of CoA, an essential cofactor of oxidative and biosynthetic reactions.
Collapse
Affiliation(s)
- David Hala
- Department of Biology, University of North Texas, Denton, TX 76203, USA; (L.H.P.); (D.B.H.)
- Department of Marine Biology, Texas A&M at Galveston, Galveston, TX 77554, USA
- Correspondence: ; Tel.: +1-409-740-4535
| | - Lene H. Petersen
- Department of Biology, University of North Texas, Denton, TX 76203, USA; (L.H.P.); (D.B.H.)
- Department of Marine Biology, Texas A&M at Galveston, Galveston, TX 77554, USA
| | - Duane B. Huggett
- Department of Biology, University of North Texas, Denton, TX 76203, USA; (L.H.P.); (D.B.H.)
- Boehringer Ingelheim Animal Health, Athens, GA 30601, USA
| | - Michelle A. Puchowicz
- Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA; (M.A.P.); (H.B.)
- Department of Pediatrics, The University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Henri Brunengraber
- Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA; (M.A.P.); (H.B.)
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27705, USA;
- Department of Medicine, Division of Endocrinology, Metabolism Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
27
|
Lashley T, Tossounian MA, Costello Heaven N, Wallworth S, Peak-Chew S, Bradshaw A, Cooper JM, de Silva R, Srai SK, Malanchuk O, Filonenko V, Koopman MB, Rüdiger SGD, Skehel M, Gout I. Extensive Anti-CoA Immunostaining in Alzheimer's Disease and Covalent Modification of Tau by a Key Cellular Metabolite Coenzyme A. Front Cell Neurosci 2021; 15:739425. [PMID: 34720880 PMCID: PMC8554225 DOI: 10.3389/fncel.2021.739425] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/17/2021] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, accounting for at least two-thirds of dementia cases. A combination of genetic, epigenetic and environmental triggers is widely accepted to be responsible for the onset and development of AD. Accumulating evidence shows that oxidative stress and dysregulation of energy metabolism play an important role in AD pathogenesis, leading to neuronal dysfunction and death. Redox-induced protein modifications have been reported in the brain of AD patients, indicating excessive oxidative damage. Coenzyme A (CoA) is essential for diverse metabolic pathways, regulation of gene expression and biosynthesis of neurotransmitters. Dysregulation of CoA biosynthesis in animal models and inborn mutations in human genes involved in the CoA biosynthetic pathway have been associated with neurodegeneration. Recent studies have uncovered the antioxidant function of CoA, involving covalent protein modification by this cofactor (CoAlation) in cellular response to oxidative or metabolic stress. Protein CoAlation has been shown to both modulate the activity of modified proteins and protect cysteine residues from irreversible overoxidation. In this study, immunohistochemistry analysis with highly specific anti-CoA monoclonal antibody was used to reveal protein CoAlation across numerous neurodegenerative diseases, which appeared particularly frequent in AD. Furthermore, protein CoAlation consistently co-localized with tau-positive neurofibrillary tangles, underpinning one of the key pathological hallmarks of AD. Double immunihistochemical staining with tau and CoA antibodies in AD brain tissue revealed co-localization of the two immunoreactive signals. Further, recombinant 2N3R and 2N4R tau isoforms were found to be CoAlated in vitro and the site of CoAlation mapped by mass spectrometry to conserved cysteine 322, located in the microtubule binding region. We also report the reversible H2O2-induced dimerization of recombinant 2N3R, which is inhibited by CoAlation. Moreover, CoAlation of transiently expressed 2N4R tau was observed in diamide-treated HEK293/Pank1β cells. Taken together, this study demonstrates for the first time extensive anti-CoA immunoreactivity in AD brain samples, which occurs in structures resembling neurofibrillary tangles and neuropil threads. Covalent modification of recombinant tau at cysteine 322 suggests that CoAlation may play an important role in protecting redox-sensitive tau cysteine from irreversible overoxidation and may modulate its acetyltransferase activity and functional interactions.
Collapse
Affiliation(s)
- Tammaryn Lashley
- Queen Square Brain Bank, UCL Queen Square Institute of Neurology, London, United Kingdom
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Maria-Armineh Tossounian
- Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Neve Costello Heaven
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom
- Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Samantha Wallworth
- Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Sew Peak-Chew
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Aaron Bradshaw
- Department of Molecular Neuroscience, Faculty of Brain Sciences, Royal Free Campus, London, United Kingdom
| | - J. Mark Cooper
- Department of Molecular Neuroscience, Faculty of Brain Sciences, Royal Free Campus, London, United Kingdom
| | - Rohan de Silva
- Reta Lila Weston Institute of Neurological Studies, University College London, London, United Kingdom
| | - Surjit Kaila Srai
- Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Oksana Malanchuk
- Department of Cell Signaling, Institute of Molecular Biology and Genetics, Kyiv, Ukraine
| | - Valeriy Filonenko
- Department of Cell Signaling, Institute of Molecular Biology and Genetics, Kyiv, Ukraine
| | - Margreet B. Koopman
- Cellular Protein Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
- Science for Life, Utrecht University, Utrecht, Netherlands
| | - Stefan G. D. Rüdiger
- Cellular Protein Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
- Science for Life, Utrecht University, Utrecht, Netherlands
| | - Mark Skehel
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Ivan Gout
- Department of Structural and Molecular Biology, University College London, London, United Kingdom
- Department of Cell Signaling, Institute of Molecular Biology and Genetics, Kyiv, Ukraine
| |
Collapse
|
28
|
Audam TN, Howard CM, Garrett LF, Zheng YW, Bradley JA, Brittian KR, Frank MW, Fulghum KL, Pólos M, Herczeg S, Merkely B, Radovits T, Uchida S, Hill BG, Dassanayaka S, Jackowski S, Jones SP. Cardiac PANK1 deletion exacerbates ventricular dysfunction during pressure overload. Am J Physiol Heart Circ Physiol 2021; 321:H784-H797. [PMID: 34533403 PMCID: PMC8794231 DOI: 10.1152/ajpheart.00411.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/03/2021] [Accepted: 09/03/2021] [Indexed: 12/13/2022]
Abstract
Coenzyme A (CoA) is an essential cofactor required for intermediary metabolism. Perturbations in homeostasis of CoA have been implicated in various pathologies; however, whether CoA homeostasis is changed and the extent to which CoA levels contribute to ventricular function and remodeling during pressure overload has not been explored. In this study, we sought to assess changes in CoA biosynthetic pathway during pressure overload and determine the impact of limiting CoA on cardiac function. We limited cardiac CoA levels by deleting the rate-limiting enzyme in CoA biosynthesis, pantothenate kinase 1 (Pank1). We found that constitutive, cardiomyocyte-specific Pank1 deletion (cmPank1-/-) significantly reduced PANK1 mRNA, PANK1 protein, and CoA levels compared with Pank1-sufficient littermates (cmPank1+/+) but exerted no obvious deleterious impact on the mice at baseline. We then subjected both groups of mice to pressure overload-induced heart failure. Interestingly, there was more ventricular dilation in cmPank1-/- during the pressure overload. To explore potential mechanisms contributing to this phenotype, we performed transcriptomic profiling, which suggested a role for Pank1 in regulating fibrotic and metabolic processes during the pressure overload. Indeed, Pank1 deletion exacerbated cardiac fibrosis following pressure overload. Because we were interested in the possibility of early metabolic impacts in response to pressure overload, we performed untargeted metabolomics, which indicated significant changes to metabolites involved in fatty acid and ketone metabolism, among other pathways. Collectively, our study underscores the role of elevated CoA levels in supporting fatty acid and ketone body oxidation, which may be more important than CoA-driven, enzyme-independent acetylation in the failing heart.NEW & NOTEWORTHY Changes in CoA homeostasis have been implicated in a variety of metabolic diseases; however, the extent to which changes in CoA homeostasis impacts remodeling has not been explored. We show that limiting cardiac CoA levels via PANK deletion exacerbated ventricular remodeling during pressure overload. Our results suggest that metabolic alterations, rather than structural alterations, associated with Pank1 deletion may underlie the exacerbated cardiac phenotype during pressure overload.
Collapse
Affiliation(s)
- Timothy N Audam
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Caitlin M Howard
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Lauren F Garrett
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Yi Wei Zheng
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - James A Bradley
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Kenneth R Brittian
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Matthew W Frank
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Kyle L Fulghum
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Miklós Pólos
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Szilvia Herczeg
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Shizuka Uchida
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Bradford G Hill
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Sujith Dassanayaka
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Suzanne Jackowski
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Steven P Jones
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
29
|
Olzhausen J, Grigat M, Seifert L, Ulbricht T, Schüller HJ. Increased biosynthesis of acetyl-CoA in the yeast Saccharomyces cerevisiae by overexpression of a deregulated pantothenate kinase gene and engineering of the coenzyme A biosynthetic pathway. Appl Microbiol Biotechnol 2021; 105:7321-7337. [PMID: 34491400 PMCID: PMC8494682 DOI: 10.1007/s00253-021-11523-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/20/2021] [Accepted: 08/06/2021] [Indexed: 12/04/2022]
Abstract
Coenzyme A (CoA) and its derivatives such as acetyl-CoA are essential metabolites for several biosynthetic reactions. In the yeast S. cerevisiae, five enzymes (encoded by essential genes CAB1-CAB5; coenzyme A biosynthesis) are required to perform CoA biosynthesis from pantothenate, cysteine, and ATP. Similar to enzymes from other eukaryotes, yeast pantothenate kinase (PanK, encoded by CAB1) turned out to be inhibited by acetyl-CoA. By genetic selection of intragenic suppressors of a temperature-sensitive cab1 mutant combined with rationale mutagenesis of the presumed acetyl-CoA binding site within PanK, we were able to identify the variant CAB1 W331R, encoding a hyperactive PanK completely insensitive to inhibition by acetyl-CoA. Using a versatile gene integration cassette containing the TPI1 promoter, we constructed strains overexpressing CAB1 W331R in combination with additional genes of CoA biosynthesis (CAB2, CAB3, HAL3, CAB4, and CAB5). In these strains, the level of CoA nucleotides was 15-fold increased, compared to a reference strain without additional CAB genes. Overexpression of wild-type CAB1 instead of CAB1 W331R turned out as substantially less effective (fourfold increase of CoA nucleotides). Supplementation of overproducing strains with additional pantothenate could further elevate the level of CoA (2.3-fold). Minor increases were observed after overexpression of FEN2 (encoding a pantothenate permease) and deletion of PCD1 (CoA-specific phosphatase). We conclude that the strategy described in this work may improve the efficiency of biotechnological applications depending on acetyl-CoA. Key points • A gene encoding a hyperactive yeast pantothenate kinase (PanK) was constructed. • Overexpression of CoA biosynthetic genes elevated CoA nucleotides 15-fold. • Supplementation with pantothenate further increased the level of CoA nucleotides.
Collapse
Affiliation(s)
- Judith Olzhausen
- Center for Functional Genomics of Microbes, Abteilung Molekulare Genetik und Infektionsbiologie, Universität Greifswald, Felix-Hausdorff-Strasse 8, 17487, Greifswald, Germany
- Cendres+Métaux SA, CH-2501, Biel/Bienne, Switzerland
| | - Mathias Grigat
- Center for Functional Genomics of Microbes, Abteilung Molekulare Genetik und Infektionsbiologie, Universität Greifswald, Felix-Hausdorff-Strasse 8, 17487, Greifswald, Germany
| | - Larissa Seifert
- Center for Functional Genomics of Microbes, Abteilung Molekulare Genetik und Infektionsbiologie, Universität Greifswald, Felix-Hausdorff-Strasse 8, 17487, Greifswald, Germany
- Universitätsklinikum Hamburg-Eppendorf, Medizinische Klinik, Nephrologie, Hamburg, Germany
| | - Tom Ulbricht
- Center for Functional Genomics of Microbes, Abteilung Molekulare Genetik und Infektionsbiologie, Universität Greifswald, Felix-Hausdorff-Strasse 8, 17487, Greifswald, Germany
| | - Hans-Joachim Schüller
- Center for Functional Genomics of Microbes, Abteilung Molekulare Genetik und Infektionsbiologie, Universität Greifswald, Felix-Hausdorff-Strasse 8, 17487, Greifswald, Germany.
| |
Collapse
|
30
|
Coenzyme a Biochemistry: From Neurodevelopment to Neurodegeneration. Brain Sci 2021; 11:brainsci11081031. [PMID: 34439650 PMCID: PMC8392065 DOI: 10.3390/brainsci11081031] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 12/21/2022] Open
Abstract
Coenzyme A (CoA) is an essential cofactor in all living organisms. It is involved in a large number of biochemical processes functioning either as an activator of molecules with carbonyl groups or as a carrier of acyl moieties. Together with its thioester derivatives, it plays a central role in cell metabolism, post-translational modification, and gene expression. Furthermore, recent studies revealed a role for CoA in the redox regulation by the S-thiolation of cysteine residues in cellular proteins. The intracellular concentration and distribution in different cellular compartments of CoA and its derivatives are controlled by several extracellular stimuli such as nutrients, hormones, metabolites, and cellular stresses. Perturbations of the biosynthesis and homeostasis of CoA and/or acyl-CoA are connected with several pathological conditions, including cancer, myopathies, and cardiomyopathies. In the most recent years, defects in genes involved in CoA production and distribution have been found in patients affected by rare forms of neurodegenerative and neurodevelopmental disorders. In this review, we will summarize the most relevant aspects of CoA cellular metabolism, their role in the pathogenesis of selected neurodevelopmental and neurodegenerative disorders, and recent advancements in the search for therapeutic approaches for such diseases.
Collapse
|
31
|
Baković J, Yu BYK, Silva D, Baczynska M, Peak-Chew SY, Switzer A, Burchell L, Wigneshweraraj S, Vandanashree M, Gopal B, Filonenko V, Skehel M, Gout I. Redox Regulation of the Quorum-sensing Transcription Factor AgrA by Coenzyme A. Antioxidants (Basel) 2021; 10:antiox10060841. [PMID: 34070323 PMCID: PMC8228455 DOI: 10.3390/antiox10060841] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus aureus (S. aureus) is an aggressive opportunistic pathogen of prominent virulence and antibiotic resistance. These characteristics are due in part to the accessory gene regulator (agr) quorum-sensing system, which allows for the rapid adaptation of S. aureus to environmental changes and thus promotes virulence and the development of pathogenesis. AgrA is the agr system response regulator that binds to the P2 and P3 promoters and upregulates agr expression. In this study, we reveal that S. aureus AgrA is modified by covalent binding of CoA (CoAlation) in response to oxidative or metabolic stress. The sites of CoAlation were mapped by liquid chromatography tandem mass spectrometry (LC-MS/MS) and revealed that oxidation-sensing Cys199 is modified by CoA. Surface plasmon resonance (SPR) analysis showed an inhibitory effect of CoAlation on the DNA-binding activity, as CoAlated AgrA had significantly lower affinity towards the P2 and P3 promoters than non-CoAlated AgrA. Overall, this study provides novel insights into the mode of transcriptional regulation in S. aureus and further elucidates the link between the quorum-sensing and oxidation-sensing roles of the agr system.
Collapse
Affiliation(s)
- Jovana Baković
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (J.B.); (B.Y.K.Y.); (D.S.); (M.B.)
| | - Bess Yi Kun Yu
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (J.B.); (B.Y.K.Y.); (D.S.); (M.B.)
| | - Daniel Silva
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (J.B.); (B.Y.K.Y.); (D.S.); (M.B.)
| | - Maria Baczynska
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (J.B.); (B.Y.K.Y.); (D.S.); (M.B.)
| | - Sew Yeu Peak-Chew
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK; (S.Y.P.-C.); (M.S.)
| | - Amy Switzer
- Section of Microbiology, Faculty of Medicine and MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, UK; (A.S.); (L.B.); (S.W.)
| | - Lynn Burchell
- Section of Microbiology, Faculty of Medicine and MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, UK; (A.S.); (L.B.); (S.W.)
| | - Sivaramesh Wigneshweraraj
- Section of Microbiology, Faculty of Medicine and MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, UK; (A.S.); (L.B.); (S.W.)
| | | | - Balasubramanian Gopal
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India; (M.V.); (B.G.)
| | - Valeriy Filonenko
- Department of Cell Signaling, Institute of Molecular Biology and Genetics, 143 Kyiv, Ukraine;
| | - Mark Skehel
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK; (S.Y.P.-C.); (M.S.)
| | - Ivan Gout
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (J.B.); (B.Y.K.Y.); (D.S.); (M.B.)
- Department of Cell Signaling, Institute of Molecular Biology and Genetics, 143 Kyiv, Ukraine;
- Correspondence: ; Tel.: +0044-2076794482; Fax: +0044-2076797193
| |
Collapse
|
32
|
Yu BYK, Tossounian MA, Hristov SD, Lawrence R, Arora P, Tsuchiya Y, Peak-Chew SY, Filonenko V, Oxenford S, Angell R, Gouge J, Skehel M, Gout I. Regulation of metastasis suppressor NME1 by a key metabolic cofactor coenzyme A. Redox Biol 2021; 44:101978. [PMID: 33903070 PMCID: PMC8212152 DOI: 10.1016/j.redox.2021.101978] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/28/2021] [Accepted: 04/13/2021] [Indexed: 02/08/2023] Open
Abstract
The metastasis suppressor protein NME1 is an evolutionarily conserved and multifunctional enzyme that plays an important role in suppressing the invasion and metastasis of tumour cells. The nucleoside diphosphate kinase (NDPK) activity of NME1 is well recognized in balancing the intracellular pools of nucleotide diphosphates and triphosphates to regulate cytoskeletal rearrangement and cell motility, endocytosis, intracellular trafficking, and metastasis. In addition, NME1 was found to function as a protein-histidine kinase, 3′-5′ exonuclease and geranyl/farnesyl pyrophosphate kinase. These diverse cellular functions are regulated at the level of expression, post-translational modifications, and regulatory interactions. The NDPK activity of NME1 has been shown to be inhibited in vitro and in vivo under oxidative stress, and the inhibitory effect mediated via redox-sensitive cysteine residues. In this study, affinity purification followed by mass spectrometric analysis revealed NME1 to be a major coenzyme A (CoA) binding protein in cultured cells and rat tissues. NME1 is also found covalently modified by CoA (CoAlation) at Cys109 in the CoAlome analysis of HEK293/Pank1β cells treated with the disulfide-stress inducer, diamide. Further analysis showed that recombinant NME1 is efficiently CoAlated in vitro and in cellular response to oxidising agents and metabolic stress. In vitro CoAlation of recombinant wild type NME1, but not the C109A mutant, results in the inhibition of its NDPK activity. Moreover, CoA also functions as a competitive inhibitor of the NME1 NDPK activity by binding non-covalently to the nucleotide binding site. Taken together, our data reveal metastasis suppressor protein NME1 as a novel binding partner of the key metabolic regulator CoA, which inhibits its nucleoside diphosphate kinase activity via non-covalent and covalent interactions. NME1 is a major CoA-binding protein. CoA can bind NME1 through covalent and non-covalent interactions. NME1 CoAlation is induced by oxidative and metabolic stress in mammalian cells. CoA inhibits the NDPK activity of NME1 in vitro.
Collapse
Affiliation(s)
- Bess Yi Kun Yu
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Maria-Armineh Tossounian
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Stefan Denchev Hristov
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Ryan Lawrence
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Pallavi Arora
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Yugo Tsuchiya
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Sew Yeu Peak-Chew
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, CB2 0QH, United Kingdom
| | - Valeriy Filonenko
- Department of Cell Signaling, Institute of Molecular Biology and Genetics, Kyiv, 143, Ukraine
| | - Sally Oxenford
- School of Pharmacy, University College London, London, WC1N 1AX, United Kingdom
| | - Richard Angell
- School of Pharmacy, University College London, London, WC1N 1AX, United Kingdom
| | - Jerome Gouge
- Institute of Structural and Molecular Biology, Birkbeck College, London, WC1E 7HX, United Kingdom
| | - Mark Skehel
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, CB2 0QH, United Kingdom
| | - Ivan Gout
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom; Department of Cell Signaling, Institute of Molecular Biology and Genetics, Kyiv, 143, Ukraine.
| |
Collapse
|
33
|
The Operon Encoding Hydrolytic Dehalogenation of 4-Chlorobenzoate Is Transcriptionally Regulated by the TetR-Type Repressor FcbR and Its Ligand 4-Chlorobenzoyl Coenzyme A. Appl Environ Microbiol 2021; 87:AEM.02652-20. [PMID: 33397703 DOI: 10.1128/aem.02652-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/22/2020] [Indexed: 12/24/2022] Open
Abstract
The bacterial hydrolytic dehalogenation of 4-chlorobenzoate (4CBA) is a coenzyme A (CoA)-activation-type catabolic pathway that is usually a common part of the microbial mineralization of chlorinated aromatic compounds. Previous studies have shown that the transport and dehalogenation genes for 4CBA are typically clustered as an fcbBAT1T2T3C operon and inducibly expressed in response to 4CBA. However, the associated molecular mechanism remains unknown. In this study, a gene (fcbR) adjacent to the fcb operon was predicted to encode a TetR-type transcriptional regulator in Comamonas sediminis strain CD-2. The fcbR knockout strain exhibited constitutive expression of the fcb cluster. In the host Escherichia coli, the expression of the Pfcb -fused green fluorescent protein (gfp) reporter was repressed by the introduction of the fcbR gene, and genetic studies combining various catabolic genes suggest that the ligand for FcbR may be an intermediate metabolite. Purified FcbR could bind to the Pfcb DNA probe in vitro, and the metabolite 4-chlorobenzyl-CoA (4CBA-CoA) prevented FcbR binding to the P fcb DNA probe. Isothermal titration calorimetry (ITC) measurements showed that 4CBA-CoA could bind to FcbR at a 1:1 molar ratio. DNase I footprinting showed that FcbR protected a 42-bp DNA motif (5'-GGAAATCAATAGGTCCATAGAAAATCTATTGACTAATCGAAT-3') that consists of two sequence repeats containing four pseudopalindromic sequences (5'-TCNATNGA-3'). This binding motif overlaps with the -35 box of Pfcb and was proposed to prevent the binding of RNA polymerase. This study characterizes a transcriptional repressor of the fcb operon, together with its ligand, thus identifying halogenated benzoyl-CoA as belonging to the class of ligands of transcriptional regulators.IMPORTANCE The bacterial hydrolytic dehalogenation of 4CBA is a special CoA-activation-type catabolic pathway that plays an important role in the biodegradation of polychlorinated biphenyls and some herbicides. With genetic and biochemical approaches, the present study identified the transcriptional repressor and its cognate effector of a 4CBA hydrolytic dehalogenation operon. This work extends halogenated benzoyl-CoA as a new member of CoA-derived effector compounds that mediate allosteric regulation of transcriptional regulators.
Collapse
|
34
|
Baković J, López Martínez D, Nikolaou S, Yu BYK, Tossounian MA, Tsuchiya Y, Thrasivoulou C, Filonenko V, Gout I. Regulation of the CoA Biosynthetic Complex Assembly in Mammalian Cells. Int J Mol Sci 2021; 22:ijms22031131. [PMID: 33498827 PMCID: PMC7865483 DOI: 10.3390/ijms22031131] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/14/2022] Open
Abstract
Coenzyme A (CoA) is an essential cofactor present in all living cells. Under physiological conditions, CoA mainly functions to generate metabolically active CoA thioesters, which are indispensable for cellular metabolism, the regulation of gene expression, and the biosynthesis of neurotransmitters. When cells are exposed to oxidative or metabolic stress, CoA acts as an important cellular antioxidant that protects protein thiols from overoxidation, and this function is mediated by protein CoAlation. CoA and its derivatives are strictly maintained at levels controlled by nutrients, hormones, metabolites, and cellular stresses. Dysregulation of their biosynthesis and homeostasis has deleterious consequences and has been noted in a range of pathological conditions, including cancer, diabetes, Reye’s syndrome, cardiac hypertrophy, and neurodegeneration. The biochemistry of CoA biosynthesis, which involves five enzymatic steps, has been extensively studied. However, the existence of a CoA biosynthetic complex and the mode of its regulation in mammalian cells are unknown. In this study, we report the assembly of all five enzymes that drive CoA biosynthesis, in HEK293/Pank1β and A549 cells, using the in situ proximity ligation assay. Furthermore, we show that the association of CoA biosynthetic enzymes is strongly upregulated in response to serum starvation and oxidative stress, whereas insulin and growth factor signaling downregulate their assembly.
Collapse
Affiliation(s)
- Jovana Baković
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (J.B.); (D.L.M.); (S.N.); (B.Y.K.Y.); (M.-A.T.); (Y.T.)
| | - David López Martínez
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (J.B.); (D.L.M.); (S.N.); (B.Y.K.Y.); (M.-A.T.); (Y.T.)
| | - Savvas Nikolaou
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (J.B.); (D.L.M.); (S.N.); (B.Y.K.Y.); (M.-A.T.); (Y.T.)
| | - Bess Yi Kun Yu
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (J.B.); (D.L.M.); (S.N.); (B.Y.K.Y.); (M.-A.T.); (Y.T.)
| | - Maria-Armineh Tossounian
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (J.B.); (D.L.M.); (S.N.); (B.Y.K.Y.); (M.-A.T.); (Y.T.)
| | - Yugo Tsuchiya
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (J.B.); (D.L.M.); (S.N.); (B.Y.K.Y.); (M.-A.T.); (Y.T.)
| | - Christopher Thrasivoulou
- Research Department of Cell and Developmental Biology, The Centre for Cell and Molecular Dynamics, University College London, London WC1E 6BT, UK;
| | - Valeriy Filonenko
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03680 Kyiv, Ukraine;
| | - Ivan Gout
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (J.B.); (D.L.M.); (S.N.); (B.Y.K.Y.); (M.-A.T.); (Y.T.)
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03680 Kyiv, Ukraine;
- Correspondence:
| |
Collapse
|
35
|
Zhyvoloup A, Yu BYK, Baković J, Davis-Lunn M, Tossounian MA, Thomas N, Tsuchiya Y, Peak-Chew SY, Wigneshweraraj S, Filonenko V, Skehel M, Setlow P, Gout I. Analysis of disulphide bond linkage between CoA and protein cysteine thiols during sporulation and in spores of Bacillus species. FEMS Microbiol Lett 2020; 367:fnaa174. [PMID: 33206970 PMCID: PMC8127865 DOI: 10.1093/femsle/fnaa174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/24/2020] [Indexed: 12/17/2022] Open
Abstract
Spores of Bacillus species have novel properties, which allow them to lie dormant for years and then germinate under favourable conditions. In the current work, the role of a key metabolic integrator, coenzyme A (CoA), in redox regulation of growing cells and during spore formation in Bacillus megaterium and Bacillus subtilis is studied. Exposing these growing cells to oxidising agents or carbon deprivation resulted in extensive covalent protein modification by CoA (termed protein CoAlation), through disulphide bond formation between the CoA thiol group and a protein cysteine. Significant protein CoAlation was observed during sporulation of B. megaterium, and increased largely in parallel with loss of metabolism in spores. Mass spectrometric analysis identified four CoAlated proteins in B. subtilis spores as well as one CoAlated protein in growing B. megaterium cells. All five of these proteins have been identified as moderately abundant in spores. Based on these findings and published studies, protein CoAlation might be involved in facilitating establishment of spores' metabolic dormancy, and/or protecting sensitive sulfhydryl groups of spore enzymes.
Collapse
Affiliation(s)
- Alexander Zhyvoloup
- Department of Structural and Molecular Biology, University College London, Gower St., London WC1E 6BT, UK
| | - Bess Yi Kun Yu
- Department of Structural and Molecular Biology, University College London, Gower St., London WC1E 6BT, UK
| | - Jovana Baković
- Department of Structural and Molecular Biology, University College London, Gower St., London WC1E 6BT, UK
| | - Mathew Davis-Lunn
- Department of Structural and Molecular Biology, University College London, Gower St., London WC1E 6BT, UK
| | - Maria-Armineh Tossounian
- Department of Structural and Molecular Biology, University College London, Gower St., London WC1E 6BT, UK
| | - Naam Thomas
- Department of Structural and Molecular Biology, University College London, Gower St., London WC1E 6BT, UK
| | - Yugo Tsuchiya
- Department of Structural and Molecular Biology, University College London, Gower St., London WC1E 6BT, UK
| | - Sew Yeu Peak-Chew
- Biological Mass Spectrometry & Proteomics Cell Biology, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Trumpington, Cambridge CB2 0QH, UK
| | - Sivaramesh Wigneshweraraj
- Section of Microbiology, Faculty of Medicine and MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Flowers Building, Imperial College Road, London SW7 2AZ, UK
| | - Valeriy Filonenko
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 150 Zabolotnogo St., Kyiv 03680, Ukraine
| | - Mark Skehel
- Biological Mass Spectrometry & Proteomics Cell Biology, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Trumpington, Cambridge CB2 0QH, UK
| | - Peter Setlow
- Department of Molecular Biology and Biophysics, UConn Health, 263 Farmington Avenue, Farmington, CT 06030-3305, USA
| | - Ivan Gout
- Department of Structural and Molecular Biology, University College London, Gower St., London WC1E 6BT, UK
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 150 Zabolotnogo St., Kyiv 03680, Ukraine
| |
Collapse
|
36
|
Czumaj A, Szrok-Jurga S, Hebanowska A, Turyn J, Swierczynski J, Sledzinski T, Stelmanska E. The Pathophysiological Role of CoA. Int J Mol Sci 2020; 21:ijms21239057. [PMID: 33260564 PMCID: PMC7731229 DOI: 10.3390/ijms21239057] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022] Open
Abstract
The importance of coenzyme A (CoA) as a carrier of acyl residues in cell metabolism is well understood. Coenzyme A participates in more than 100 different catabolic and anabolic reactions, including those involved in the metabolism of lipids, carbohydrates, proteins, ethanol, bile acids, and xenobiotics. However, much less is known about the importance of the concentration of this cofactor in various cell compartments and the role of altered CoA concentration in various pathologies. Despite continuous research on these issues, the molecular mechanisms in the regulation of the intracellular level of CoA under pathological conditions are still not well understood. This review summarizes the current knowledge of (a) CoA subcellular concentrations; (b) the roles of CoA synthesis and degradation processes; and (c) protein modification by reversible CoA binding to proteins (CoAlation). Particular attention is paid to (a) the roles of changes in the level of CoA under pathological conditions, such as in neurodegenerative diseases, cancer, myopathies, and infectious diseases; and (b) the beneficial effect of CoA and pantethine (which like CoA is finally converted to Pan and cysteamine), used at pharmacological doses for the treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Aleksandra Czumaj
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdańsk, Poland;
| | - Sylwia Szrok-Jurga
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (A.H.); (J.T.)
| | - Areta Hebanowska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (A.H.); (J.T.)
| | - Jacek Turyn
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (A.H.); (J.T.)
| | - Julian Swierczynski
- State School of Higher Vocational Education in Koszalin, 75-582 Koszalin, Poland;
| | - Tomasz Sledzinski
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdańsk, Poland;
- Correspondence: (T.S.); (E.S.); Tel.: +48-(0)-583-491-479 (T.S.)
| | - Ewa Stelmanska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (A.H.); (J.T.)
- Correspondence: (T.S.); (E.S.); Tel.: +48-(0)-583-491-479 (T.S.)
| |
Collapse
|
37
|
Bellany F, Tsuchiya Y, Tran TM, Chan AWE, Allan H, Gout I, Tabor AB. Design and synthesis of Coenzyme A analogues as Aurora kinase A inhibitors: An exploration of the roles of the pyrophosphate and pantetheine moieties. Bioorg Med Chem 2020; 28:115740. [PMID: 33007553 DOI: 10.1016/j.bmc.2020.115740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022]
Abstract
Coenzyme A (CoA) is a highly selective inhibitor of the mitotic regulatory enzyme Aurora A kinase, with a novel mode of action. Herein we report the design and synthesis of analogues of CoA as inhibitors of Aurora A kinase. We have designed and synthesised modified CoA structures as potential inhibitors, combining dicarbonyl mimics of the pyrophosphate group with a conserved adenosine headgroup and different length pantetheine-based tail groups. An analogue with a -SH group at the end of the pantotheinate tail showed the best IC50, probably due to the formation of a covalent bond with Aurora A kinase Cys290.
Collapse
Affiliation(s)
- Fiona Bellany
- Department of Chemistry, UCL, Christopher Ingold Building, 20, Gordon Street, London WC1H 0AJ, UK
| | - Yugo Tsuchiya
- Department of Structural and Molecular Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Trang M Tran
- Department of Chemistry, UCL, Christopher Ingold Building, 20, Gordon Street, London WC1H 0AJ, UK
| | - A W Edith Chan
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Helen Allan
- Department of Chemistry, UCL, Christopher Ingold Building, 20, Gordon Street, London WC1H 0AJ, UK
| | - Ivan Gout
- Department of Structural and Molecular Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Alethea B Tabor
- Department of Chemistry, UCL, Christopher Ingold Building, 20, Gordon Street, London WC1H 0AJ, UK.
| |
Collapse
|
38
|
Chen N, Liu Y, Li Y, Wang C. Chemical Proteomic Profiling of Protein 4′‐Phosphopantetheinylation in Mammalian Cells. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202004105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Nan Chen
- College of Chemistry and Molecular Engineering Synthetic and Functional Biomolecules Center Beijing National Laboratory for Molecular Sciences Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
| | - Yuan Liu
- College of Chemistry and Molecular Engineering Synthetic and Functional Biomolecules Center Beijing National Laboratory for Molecular Sciences Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
| | - Yuanpei Li
- College of Chemistry and Molecular Engineering Synthetic and Functional Biomolecules Center Beijing National Laboratory for Molecular Sciences Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
| | - Chu Wang
- College of Chemistry and Molecular Engineering Synthetic and Functional Biomolecules Center Beijing National Laboratory for Molecular Sciences Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
- Peking-Tsinghua Center for Life Sciences Peking University China
| |
Collapse
|
39
|
Chen N, Liu Y, Li Y, Wang C. Chemical Proteomic Profiling of Protein 4′‐Phosphopantetheinylation in Mammalian Cells. Angew Chem Int Ed Engl 2020; 59:16069-16075. [DOI: 10.1002/anie.202004105] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/08/2020] [Indexed: 01/24/2023]
Affiliation(s)
- Nan Chen
- College of Chemistry and Molecular Engineering Synthetic and Functional Biomolecules Center Beijing National Laboratory for Molecular Sciences Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
| | - Yuan Liu
- College of Chemistry and Molecular Engineering Synthetic and Functional Biomolecules Center Beijing National Laboratory for Molecular Sciences Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
| | - Yuanpei Li
- College of Chemistry and Molecular Engineering Synthetic and Functional Biomolecules Center Beijing National Laboratory for Molecular Sciences Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
| | - Chu Wang
- College of Chemistry and Molecular Engineering Synthetic and Functional Biomolecules Center Beijing National Laboratory for Molecular Sciences Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Peking University Beijing 100871 China
- Peking-Tsinghua Center for Life Sciences Peking University China
| |
Collapse
|
40
|
Zhou S, Hao T, Xu S, Deng Y. Coenzyme A thioester-mediated carbon chain elongation as a paintbrush to draw colorful chemical compounds. Biotechnol Adv 2020; 43:107575. [PMID: 32512221 DOI: 10.1016/j.biotechadv.2020.107575] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 05/31/2020] [Accepted: 06/01/2020] [Indexed: 12/23/2022]
Abstract
The biosynthesis of various useful chemicals from simple substrates using industrial microorganisms is becoming increasingly crucial to address the challenge of dwindling non-renewable resources. As the most common intermediate substrates in organisms, Coenzyme A (CoA) thioesters play a central role in the carbon chain elongation process of their products. As a result, numerous of chemicals can be synthesized by the iterative addition of various CoA thioester extender units at a given CoA thioester primer backbone. However, these elongation reactions and the product yields are still restricted due to the low enzymatic performance and supply of CoA thioesters. This review highlights the current protein and metabolic engineering strategies used to enhance the diversity and product yield by coupling different primers, extender units, enzymes, and termination pathways, in an attempt to provide a road map for producing a more diverse range of industrial chemicals.
Collapse
Affiliation(s)
- Shenghu Zhou
- National Engineering Laboratory for Cereal Fermentation Technology (NELCF), Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Tingting Hao
- National Engineering Laboratory for Cereal Fermentation Technology (NELCF), Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Shumin Xu
- National Engineering Laboratory for Cereal Fermentation Technology (NELCF), Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yu Deng
- National Engineering Laboratory for Cereal Fermentation Technology (NELCF), Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
41
|
Thananurak P, Chuaychu-Noo N, Thélie A, Phasuk Y, Vongpralub T, Blesbois E. Different concentrations of cysteamine, ergothioneine, and serine modulate quality and fertilizing ability of cryopreserved chicken sperm. Poult Sci 2019; 99:1185-1198. [PMID: 32029149 PMCID: PMC7587801 DOI: 10.1016/j.psj.2019.10.040] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 12/01/2022] Open
Abstract
The aim of this study was to evaluate the effects of freezing diluents supplemented in three potential amines/amino acids, namely, antioxidant cysteamine (2-aminoethanethiol [AET]), ergothioneine (ERG), and serine (SER), in optimization of chicken sperm cryopreservation. The semen of 36 Pradu Hang Dum males, selected based on their motility vigor score, was frozen by a simple freezing method using nitrogen vapors and dimethylformamide (DMF). In a first experiment, a wide range of AET, ERG, and SER doses were tested. Semen quality was evaluated after incubation at 5°C or after cryopreservation in straws in the Blumberger Hahnen Sperma Verdünner (BHSV) diluent + DMF (6% v/v) with or without AET, ERG, or SER. The best targeted doses of AET, ERG, or SER were then selected for experiment 2 that was focused on cryopreserved semen. Frozen-thawed sperm quality was evaluated by different in vitro tests and by evaluation of fertility. Objective motility parameters were evaluated by computer-assisted sperm analysis. Membrane integrity, acrosome integrity, and mitochondria function were evaluated using appropriate dyes and flow cytometry. Lipid peroxide production was assessed by the thiobarbituric acid test (malondialdehyde production). Fertility obtained with frozen-thawed semen supplemented or not in AET, ERG, or SER was evaluated after artificial insemination of laying hens. ERG and AET decreased sperm lipid peroxidation and decreased fertility, even at low doses. The presence of 4 mmol of SER significantly decreased lipid peroxidation, increased the frozen-thawed sperm quality, and increased fertility after sperm cryopreservation (90% vs. control 84%, P < 0.05). In a third experiment, the use of 1 mmol of sucrose (the best result of our previous study) added to 4 mmol of SER-supplemented extender was tested. This addition allowed to the highest levels of fertility (93%). In conclusion, the addition of 4 mmol of SER in semen cryopreservation diluents decreases peroxidation and improves the efficiency of the process.
Collapse
Affiliation(s)
- Pachara Thananurak
- Department of Animal Science, Faculty of Agriculture, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Napapach Chuaychu-Noo
- Department of Animal Science, Faculty of Agriculture, Rajamangala University of Technology Srivijaya, Nakhon Sri Thammarat 80110, Thailand
| | - Aurore Thélie
- UMR-PRC, INRA, CNRS, IFCE, Université de Tours, 37380 Nouzilly, France
| | - Yupin Phasuk
- Department of Animal Science, Faculty of Agriculture, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Thevin Vongpralub
- Department of Animal Science, Faculty of Agriculture, Khon Kaen University, Khon Kaen 40002, Thailand; Research and Development Network Center for Animal Breeding (Native Chicken), Khon Kaen University, Khon Kaen 40002, Thailand.
| | | |
Collapse
|
42
|
Bartucci R, Salvati A, Olinga P, Boersma YL. Vanin 1: Its Physiological Function and Role in Diseases. Int J Mol Sci 2019; 20:E3891. [PMID: 31404995 PMCID: PMC6719204 DOI: 10.3390/ijms20163891] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/06/2019] [Accepted: 08/08/2019] [Indexed: 12/11/2022] Open
Abstract
The enzyme vascular non-inflammatory molecule-1 (vanin 1) is highly expressed at gene and protein level in many organs, such as the liver, intestine, and kidney. Its major function is related to its pantetheinase activity; vanin 1 breaks down pantetheine in cysteamine and pantothenic acid, a precursor of coenzyme A. Indeed, its physiological role seems strictly related to coenzyme A metabolism, lipid metabolism, and energy production. In recent years, many studies have elucidated the role of vanin 1 under physiological conditions in relation to oxidative stress and inflammation. Vanin's enzymatic activity was found to be of key importance in certain diseases, either for its protective effect or as a sensitizer, depending on the diseased organ. In this review, we discuss the role of vanin 1 in the liver, kidney, intestine, and lung under physiological as well as pathophysiological conditions. Thus, we provide a more complete understanding and overview of its complex function and contribution to some specific pathologies.
Collapse
Affiliation(s)
- Roberta Bartucci
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- Division of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- Division of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Anna Salvati
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Peter Olinga
- Division of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Ykelien L Boersma
- Division of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
43
|
A key metabolic integrator, coenzyme A, modulates the activity of peroxiredoxin 5 via covalent modification. Mol Cell Biochem 2019; 461:91-102. [PMID: 31375973 PMCID: PMC6790197 DOI: 10.1007/s11010-019-03593-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/20/2019] [Indexed: 01/18/2023]
Abstract
Peroxiredoxins (Prdxs) are antioxidant enzymes that catalyse the breakdown of peroxides and regulate redox activity in the cell. Peroxiredoxin 5 (Prdx5) is a unique member of Prdxs, which displays a wider subcellular distribution and substrate specificity and exhibits a different catalytic mechanism when compared to other members of the family. Here, the role of a key metabolic integrator coenzyme A (CoA) in modulating the activity of Prdx5 was investigated. We report for the first time a novel mode of Prdx5 regulation mediated via covalent and reversible attachment of CoA (CoAlation) in cellular response to oxidative and metabolic stress. The site of CoAlation in endogenous Prdx5 was mapped by mass spectrometry to peroxidatic cysteine 48. By employing an in vitro CoAlation assay, we showed that Prdx5 peroxidase activity is inhibited by covalent interaction with CoA in a dithiothreitol-sensitive manner. Collectively, these results reveal that human Prdx5 is a substrate for CoAlation in vitro and in vivo, and provide new insight into metabolic control of redox status in mammalian cells.
Collapse
|
44
|
Kosai A, Horike N, Takei Y, Yamashita A, Fujita K, Kamatani T, Tsumaki N. Changes in acetyl-CoA mediate Sik3-induced maturation of chondrocytes in endochondral bone formation. Biochem Biophys Res Commun 2019; 516:1097-1102. [PMID: 31280862 DOI: 10.1016/j.bbrc.2019.06.139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 06/25/2019] [Indexed: 12/23/2022]
Abstract
The maturation of chondrocytes is strictly regulated for proper endochondral bone formation. Although recent studies have revealed that intracellular metabolic processes regulate the proliferation and differentiation of cells, little is known about how changes in metabolite levels regulate chondrocyte maturation. To identify the metabolites which regulate chondrocyte maturation, we performed a metabolome analysis on chondrocytes of Sik3 knockout mice, in which chondrocyte maturation is delayed. Among the metabolites, acetyl-CoA was decreased in this model. Immunohistochemical analysis of the Sik3 knockout chondrocytes indicated that the expression levels of phospho-pyruvate dehydrogenase (phospho-Pdh), an inactivated form of Pdh, which is an enzyme that converts pyruvate to acetyl-CoA, and of Pdh kinase 4 (Pdk4), which phosphorylates Pdh, were increased. Inhibition of Pdh by treatment with CPI613 delayed chondrocyte maturation in metatarsal primordial cartilage in organ culture. These results collectively suggest that decreasing the acetyl-CoA level is a cause and not result of the delayed chondrocyte maturation. Sik3 appears to increase the acetyl-CoA level by decreasing the expression level of Pdk4. Blocking ATP synthesis in the TCA cycle by treatment with rotenone also delayed chondrocyte maturation in metatarsal primordial cartilage in organ culture, suggesting the possibility that depriving acetyl-CoA as a substrate for the TCA cycle is responsible for the delayed maturation. Our finding of acetyl-CoA as a regulator of chondrocyte maturation could contribute to understanding the regulatory mechanisms controlling endochondral bone formation by metabolites.
Collapse
Affiliation(s)
- Azuma Kosai
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Japan
| | - Nanao Horike
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Japan
| | - Yoshiaki Takei
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Japan
| | - Akihiro Yamashita
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Japan
| | - Kaori Fujita
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Japan
| | - Takashi Kamatani
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Japan
| | - Noriyuki Tsumaki
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Japan.
| |
Collapse
|
45
|
Xia J, Zheng M, Li L, Hou X, Zeng W. [Conjugated linoleic acid improves glucose and lipid metabolism in diabetic mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:740-746. [PMID: 31270056 DOI: 10.12122/j.issn.1673-4254.2019.06.18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To analyze the effect of conjugated linoleic acid (CLA) on glucose and lipid metabolism in obese diabetic (db/db) mice. METHODS db/db mice were randomized for treatment with saline or CLA mixture administered intragastrically. The changes in body weight, dietary intake, water intake, oral glucose tolerance, triglyceride and total cholesterol were recorded after the treatments. HE staining and oil red O staining were used to assess liver pathologies and fatty acid content. The expression levels of PPARα, PPARγ, CD36, CHREBP and SREBP-1c were detected using real-time PCR and Western blotting. HepG2 cells were treated with CLA and linoleic acid and the expressions of PPARα, ACC, P-ACC, and CD36 were detected; the level of acetyl-CoA in the cell supernatant was detected using ELISA. RESULTS CLA treatment obviously reduced the dietary and water intake of db/db mice, effectively reduced the body weight and decreased serum triglyceride and cholesterol levels (P < 0.05). CLA significantly reduced fasting blood glucose, increased glucose tolerance, reduced the accumulation of lipid droplets in the liver and improved lipid metabolism in db/db mice. The mice showed significantly increased expression of PPARα (P < 0.05) and lowered CD36 expression (P < 0.001) in the liver after CLA treatment. Cellular experiments showed that CLA significantly up-regulated PPARα (P < 0.001) and P-ACC and decreased the expression of CD36 (P < 0.01). ELISA showed that acetyl-CoA was significantly up-regulated in the cells after CLA treatment (P < 0.01). CONCLUSIONS The mixture of two conjugated linoleic acid isomers can reduce fasting blood glucose, increase glucose tolerance and improve glycolipid metabolism in db/db mice by enhancing the expression of PPARα, increasing P-ACC and inhibiting CD36 expression.
Collapse
Affiliation(s)
- Jun Xia
- Department of Cell Biology, School of Basic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Mingyue Zheng
- Department of Cell Biology, School of Basic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Lingjie Li
- Department of Cell Biology, School of Basic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xufeng Hou
- Department of Cell Biology, School of Basic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Weisen Zeng
- Department of Cell Biology, School of Basic Medicine, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
46
|
Aloum L, Brimson CA, Zhyvoloup A, Baines R, Baković J, Filonenko V, Thompson CRL, Gout I. Coenzyme A and protein CoAlation levels are regulated in response to oxidative stress and during morphogenesis in Dictyostelium discoideum. Biochem Biophys Res Commun 2019; 511:294-299. [PMID: 30797553 PMCID: PMC6416166 DOI: 10.1016/j.bbrc.2019.02.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/06/2019] [Indexed: 01/13/2023]
Abstract
Dictyostelium discoideum (D. discoideum) is a simple eukaryote with a unique life cycle in which it differentiates from unicellular amoebae into a fruiting body upon starvation. Reactive oxygen species (ROS) have been associated with bacterial predation, as well as regulatory events during D. discoideum development and differentiation. Coenzyme A (CoA) is a key metabolic integrator in all living cells. A novel function of CoA in redox regulation, mediated by covalent attachment of CoA to cellular proteins in response to oxidative or metabolic stress, has been recently discovered and termed protein CoAlation. In this study, we report that the level of CoA and protein CoAlation in D. discoideum are developmentally regulated, and correlate with the temporal expression pattern of genes implicated in CoA biosynthesis during morphogenesis. Furthermore, treatment of growing D. discoideum cells with oxidising agents results in a dose-dependent increase of protein CoAlation. However, much higher concentrations were required when compared to mammalian cells and bacteria. Increased resistance of D. discoideum to oxidative stress induced by H2O2 has previously been attributed to high levels of catalase activity. In support of this notion, we found that H2O2-induced protein CoAlation is significantly increased in CatA-deficient D. discoideum cells. Collectively, this study provides insights into the role of CoA and protein CoAlation in the maintenance of redox homeostasis in amoeba and during D. discoideum morphogenesis. D. discoideum cells are professional phagocytes and produce ROS for efficient bacterial killing. D. discoideum cells are highly resistant to oxidative stress. CoA biosynthetic genes are transcriptionally regulated during morphogenesis. The level of CoA and protein CoAlation are developmentally regulated. Oxidising agents induce protein CoAlation in D. discoideum cells.
Collapse
Affiliation(s)
- Lujain Aloum
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Christopher A Brimson
- Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, United Kingdom
| | - Alexander Zhyvoloup
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Robert Baines
- Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, United Kingdom
| | - Jovana Baković
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Valeriy Filonenko
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, 03680, Ukraine
| | - Christopher R L Thompson
- Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, United Kingdom.
| | - Ivan Gout
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom.
| |
Collapse
|
47
|
Coenzyme A: a protective thiol in bacterial antioxidant defence. Biochem Soc Trans 2019; 47:469-476. [PMID: 30783014 DOI: 10.1042/bst20180415] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/28/2018] [Accepted: 01/09/2019] [Indexed: 12/22/2022]
Abstract
Coenzyme A (CoA) is an indispensable cofactor in all living organisms. It is synthesized in an evolutionarily conserved pathway by enzymatic conjugation of cysteine, pantothenate (Vitamin B5), and ATP. This unique chemical structure allows CoA to employ its highly reactive thiol group for diverse biochemical reactions. The involvement of the CoA thiol group in the production of metabolically active CoA thioesters (e.g. acetyl CoA, malonyl CoA, and HMG CoA) and activation of carbonyl-containing compounds has been extensively studied since the discovery of this cofactor in the middle of the last century. We are, however, far behind in understanding the role of CoA as a low-molecular-weight thiol in redox regulation. This review summarizes our current knowledge of CoA function in redox regulation and thiol protection under oxidative stress in bacteria. In this context, I discuss recent findings on a novel mode of redox regulation involving covalent modification of cellular proteins by CoA, termed protein CoAlation.
Collapse
|
48
|
Nagana Gowda GA, Abell L, Tian R. Extending the Scope of 1H NMR Spectroscopy for the Analysis of Cellular Coenzyme A and Acetyl Coenzyme A. Anal Chem 2019; 91:2464-2471. [PMID: 30608643 DOI: 10.1021/acs.analchem.8b05286] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Coenzyme A (CoA) and acetyl-coenzyme A (acetyl-CoA) are ubiquitous cellular molecules, which mediate hundreds of anabolic and catabolic reactions including energy metabolism. Highly sensitive methods including absorption spectroscopy and mass spectrometry enable their analysis, albeit with many limitations. To date, however, NMR spectroscopy has not been used to analyze these important molecules. Building on our recent efforts, which enabled simultaneous analysis of a large number of metabolites in tissue and blood including many coenzymes and antioxidants ( Anal. Chem. 2016, 88, 4817-24; ibid 2017, 89, 4620-4627), we describe here a new method for identification and quantitation of CoA and acetyl-CoA ex vivo in tissue. Using mouse heart, kidney, liver, brain, and skeletal tissue, we show that a simple 1H NMR experiment can simultaneously measure these molecules. Identification of the two species involved a comprehensive analysis of the different tissue types using 1D and 2D NMR, in combination with spectral databases for standards, as well as spiking with authentic compounds. Time dependent studies showed that while the acetyl-CoA levels remain unaltered, CoA levels diminish by more than 50% within 24 h, which indicates that CoA is labile in solution; however, degassing the sample with helium gas halted its oxidation. Further, interestingly, we also identified endogenous coenzyme A glutathione disulfide (CoA-S-S-G) in tissue for the first time by NMR and show that CoA, when oxidized in tissue extract, also forms the same disulfide metabolite. The ability to simultaneously visualize absolute concentrations of CoA, acetyl-CoA, and endogenous CoA-S-S-G along with redox coenzymes (NAD+, NADH, NADP+, NADPH), energy coenzymes (ATP, ADP, AMP), antioxidants (GSH, GSSG), and a vast pool of other metabolites using a single 1D NMR spectrum offers a new avenue in the metabolomics field for investigation of cellular function in health and disease.
Collapse
|
49
|
Probing the ligand preferences of the three types of bacterial pantothenate kinase. Bioorg Med Chem 2018; 26:5896-5902. [DOI: 10.1016/j.bmc.2018.10.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/23/2018] [Accepted: 10/29/2018] [Indexed: 12/16/2022]
|
50
|
Manea E. A step closer in defining glycosylphosphatidylinositol anchored proteins role in health and glycosylation disorders. Mol Genet Metab Rep 2018; 16:67-75. [PMID: 30094187 PMCID: PMC6080220 DOI: 10.1016/j.ymgmr.2018.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/21/2018] [Accepted: 07/21/2018] [Indexed: 12/18/2022] Open
Abstract
Glycosylphosphatidylinositol anchored proteins (GPI-APs) represent a class of soluble proteins attached to the external leaflet of the plasma membrane by a post-translation modification, the GPI anchor. The 28 genes currently involved in the synthesis and remodelling of the GPI anchor add to the ever-growing class of congenital glycosylation disorders. Recent advances in next generation sequencing technology have led to the discovery of Mabry disease and CHIME syndrome genetic aetiology. Moreover, with each described mutation known phenotypes expand and new ones emerge without clear genotype-phenotype correlation. A protein database search was made for human GPI-APs with defined pathology to help building-up a physio-pathological mechanism from a clinical perspective. GPI-APs function in vitamin-B6 and folate transport, nucleotide metabolism and lipid homeostasis. Defining GPI-APs role in disease bears significant clinical implications.
Collapse
|