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] [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
|
Khan J, Bareja C, Dwivedi K, Mathur A, Kumar N, Saluja D. Identification and validation of a metabolic-related gene risk model predicting the prognosis of lung, colon, and breast cancers. Sci Rep 2025; 15:1374. [PMID: 39779736 PMCID: PMC11711664 DOI: 10.1038/s41598-025-85366-8] [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: 06/03/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Metabolic reprogramming, vital for cancer cells to adapt to the altered microenvironment, remains a topic requiring further investigation for different tumor types. Our study aims to elucidate shared metabolic reprogramming across breast (BRC), colorectal (CRC), and lung (LUC) cancers. Leveraging gene expression data from the Gene Expression Omnibus and various bioinformatics tools like MSigDB, WebGestalt, String, and Cytoscape, we identified key/hub metabolism-related genes (MRGs) and their interactions. The functional characteristics including survival parameters and expression of the key MRGs were analyzed and validated through Gene Expression Profiling Interactive Analysis 2 and qRT-PCR. In addition, we employed machine learning algorithms such as k-nearest neighbours (KNN), support vector regressor (SVR), and extreme gradient boosting (XGBoost) to assess MRGs' effectiveness in predicting overall patient survival. Among 11,384 DEGs analyzed, 540 overlapped across BRC, CRC, and LUC, with 46 MRGs and 20 key/hub MRGs involved in all studied cancer types. Of these, 11 key MRGs were prognostically significant. The qRT-PCR validation of key MRGs in specific cancer cell lines confirmed their expression profiles, with some showing cell-type-specific patterns. SVR exhibited remarkable accuracy in predicting overall survival, emphasizing its clinical utility. Our integrated approach combining bioinformatics analyses and experimental validations underscores the potential of MRGs as biomarkers for metabolic therapies, with machine learning models enhancing predictive capabilities for patient outcomes.
Collapse
Affiliation(s)
- Jiyauddin Khan
- Dr B R Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Chanchal Bareja
- Dr B R Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Kountay Dwivedi
- Department of Computer Science, FacultyofMathematicalSciences, University of Delhi, Delhi, 110007, India
| | - Ankit Mathur
- Dr B R Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
- Delhi School of Public Health (DSPH), Institution of Eminence (IoE), University of Delhi, Delhi, 110007, India
| | - Naveen Kumar
- Department of Computer Science, FacultyofMathematicalSciences, University of Delhi, Delhi, 110007, India
| | - Daman Saluja
- Dr B R Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India.
- Delhi School of Public Health (DSPH), Institution of Eminence (IoE), University of Delhi, Delhi, 110007, India.
| |
Collapse
|
3
|
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
|
4
|
Malanchuk O, Bdzhola A, Palchevskyi S, Bdzhola V, Chai P, Pardo OE, Seckl MJ, Banerjee A, Peak-Chew SY, Skehel M, Guruprasad L, Zhyvoloup A, Gout I, Filonenko V. Investigating the Regulation of Ribosomal Protein S6 Kinase 1 by CoAlation. Int J Mol Sci 2024; 25:8747. [PMID: 39201434 PMCID: PMC11354579 DOI: 10.3390/ijms25168747] [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: 07/03/2024] [Revised: 08/02/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
Ribosomal protein S6 kinases belong to a family of highly conserved enzymes in eukaryotes that regulate cell growth, proliferation, survival, and the stress response. It is well established that the activation and downstream signalling of p70S6Ks involve multiple phosphorylation events by key regulators of cell growth, survival, and energy metabolism. Here, we report for the first time the covalent modification of p70S6K1 by coenzyme A (CoA) in response to oxidative stress, which regulates its kinase activity. The site of CoA binding (CoAlation) was mapped by mass spectrometry to cysteine 217 (Cys217), located in the kinase activation loop and only one amino acid away from the tripeptide DFG motif, which facilitates ATP-binding. The CoAlation of recombinant p70S6K1 was demonstrated in vitro and was shown to inhibit its kinase activity. Our molecular docking and dynamics analysis revealed the most likely mode for CoA binding to p70S6K1. This mechanism involves the non-covalent binding of the CoA ADP moiety to the p70S6K1 nucleotide-binding pocket, positioning the CoA thiol group in close proximity to form a covalent bond with the surface-exposed Cys217 residue. These findings support a "dual anchor" mechanism for protein kinase inhibition by CoAlation in cellular response to oxidative stress. Furthermore, the inhibition of S6K1 by CoAlation may open new avenues for developing novel inhibitors.
Collapse
Affiliation(s)
- Oksana Malanchuk
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (O.M.); (A.Z.)
- Department of Cell Signalling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03143 Kyiv, Ukraine; (A.B.); (S.P.); (V.B.)
| | - Anna Bdzhola
- Department of Cell Signalling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03143 Kyiv, Ukraine; (A.B.); (S.P.); (V.B.)
| | - Sergii Palchevskyi
- Department of Cell Signalling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03143 Kyiv, Ukraine; (A.B.); (S.P.); (V.B.)
| | - Volodymyr Bdzhola
- Department of Cell Signalling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03143 Kyiv, Ukraine; (A.B.); (S.P.); (V.B.)
| | - Peng Chai
- Division of Cancer, Imperial College London, Du Cane Road, London W12 0NN, UK; (P.C.); (O.E.P.); (M.J.S.)
| | - Olivier E. Pardo
- Division of Cancer, Imperial College London, Du Cane Road, London W12 0NN, UK; (P.C.); (O.E.P.); (M.J.S.)
| | - Michael J. Seckl
- Division of Cancer, Imperial College London, Du Cane Road, London W12 0NN, UK; (P.C.); (O.E.P.); (M.J.S.)
| | - Adrija Banerjee
- School of Chemistry, University of Hyderabad, Hyderabad 500 046, India; (A.B.); (L.G.)
| | - Sew Yeu Peak-Chew
- Biological Mass Spectrometry & Proteomics Cell Biology, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Trumpington, Cambridge CB2 0QH, UK;
| | - Mark Skehel
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK;
| | - Lalitha Guruprasad
- School of Chemistry, University of Hyderabad, Hyderabad 500 046, India; (A.B.); (L.G.)
| | - Alexander Zhyvoloup
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (O.M.); (A.Z.)
| | - Ivan Gout
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (O.M.); (A.Z.)
- Department of Cell Signalling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03143 Kyiv, Ukraine; (A.B.); (S.P.); (V.B.)
| | - Valeriy Filonenko
- Department of Cell Signalling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03143 Kyiv, Ukraine; (A.B.); (S.P.); (V.B.)
| |
Collapse
|
5
|
Gary CR, Acharige NPN, Oyewumi TO, Pflum MKH. Kinase-catalyzed biotinylation for discovery and validation of substrates to multispecificity kinases NME1 and NME2. J Biol Chem 2024; 300:107588. [PMID: 39032654 PMCID: PMC11375270 DOI: 10.1016/j.jbc.2024.107588] [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: 09/18/2023] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/23/2024] Open
Abstract
Protein phosphorylation by kinases regulates mammalian cell functions, such as growth, division, and signal transduction. Among human kinases, NME1 and NME2 are associated with metastatic tumor suppression but remain understudied due to the lack of tools to monitor their cellular substrates. In particular, NME1 and NME2 are multispecificity kinases phosphorylating serine, threonine, histidine, and aspartic acid residues of substrate proteins, and the heat and acid sensitivity of phosphohistidine and phosphoaspartate complicates substrate discovery and validation. To provide new substrate monitoring tools, we established the γ-phosphate-modified ATP analog, ATP-biotin, as a cosubstrate for phosphorylbiotinylation of NME1 and NME2 cellular substrates. Building upon this ATP-biotin compatibility, the Kinase-catalyzed Biotinylation with Inactivated Lysates for Discovery of Substrates method enabled validation of a known substrate and the discovery of seven NME1 and three NME2 substrates. Given the paucity of methods to study kinase substrates, ATP-biotin and the Kinase-catalyzed Biotinylation with Inactivated Lysates for Discovery of Substrates method are valuable tools to characterize the roles of NME1 and NME2 in human cell biology.
Collapse
Affiliation(s)
- Chelsea R Gary
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA
| | | | | | - Mary Kay H Pflum
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA.
| |
Collapse
|
6
|
Proust B, Herak Bosnar M, Ćetković H, Tokarska-Schlattner M, Schlattner U. Mitochondrial NME6: A Paradigm Change within the NME/NDP Kinase Protein Family? Cells 2024; 13:1278. [PMID: 39120309 PMCID: PMC11312278 DOI: 10.3390/cells13151278] [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: 07/02/2024] [Revised: 07/27/2024] [Accepted: 07/27/2024] [Indexed: 08/10/2024] Open
Abstract
Eukaryotic NMEs/NDP kinases are a family of 10 multifunctional proteins that occur in different cellular compartments and interact with various cellular components (proteins, membranes, and DNA). In contrast to the well-studied Group I NMEs (NME1-4), little is known about the more divergent Group II NMEs (NME5-9). Three recent publications now shed new light on NME6. First, NME6 is a third mitochondrial NME, largely localized in the matrix space, associated with the mitochondrial inner membrane. Second, while its monomeric form is inactive, NME6 gains NDP kinase activity through interaction with mitochondrial RCC1L. This challenges the current notion that mammalian NMEs require the formation of hexamers to become active. The formation of complexes between NME6 and RCC1L, likely heterodimers, seemingly obviates the necessity for hexamer formation, stabilizing a NDP kinase-competent conformation. Third, NME6 is involved in mitochondrial gene maintenance and expression by providing (d)NTPs for replication and transcription (in particular the pyrimidine nucleotides) and by a less characterized mechanism that supports mitoribosome function. This review offers an overview of NME evolution and structure and highlights the new insight into NME6. The new findings position NME6 as the most comprehensively studied protein in NME Group II and may even suggest it as a new paradigm for related family members.
Collapse
Affiliation(s)
- Bastien Proust
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| | - Maja Herak Bosnar
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| | - Helena Ćetković
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| | | | - Uwe Schlattner
- Univ. Grenoble Alpes, Inserm U1055, Lab. of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France;
- Institut Universitaire de France (IUF), 75231 Paris, France
| |
Collapse
|
7
|
Ning J, Sala M, Reina J, Kalagiri R, Hunter T, McCullough BS. Histidine Phosphorylation: Protein Kinases and Phosphatases. Int J Mol Sci 2024; 25:7975. [PMID: 39063217 PMCID: PMC11277029 DOI: 10.3390/ijms25147975] [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: 06/07/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Phosphohistidine (pHis) is a reversible protein post-translational modification (PTM) that is currently poorly understood. The P-N bond in pHis is heat and acid-sensitive, making it more challenging to study than the canonical phosphoamino acids pSer, pThr, and pTyr. As advancements in the development of tools to study pHis have been made, the roles of pHis in cells are slowly being revealed. To date, a handful of enzymes responsible for controlling this modification have been identified, including the histidine kinases NME1 and NME2, as well as the phosphohistidine phosphatases PHPT1, LHPP, and PGAM5. These tools have also identified the substrates of these enzymes, granting new insights into previously unknown regulatory mechanisms. Here, we discuss the cellular function of pHis and how it is regulated on known pHis-containing proteins, as well as cellular mechanisms that regulate the activity of the pHis kinases and phosphatases themselves. We further discuss the role of the pHis kinases and phosphatases as potential tumor promoters or suppressors. Finally, we give an overview of various tools and methods currently used to study pHis biology. Given their breadth of functions, unraveling the role of pHis in mammalian systems promises radical new insights into existing and unexplored areas of cell biology.
Collapse
Affiliation(s)
- Jia Ning
- Correspondence: (J.N.); (B.S.M.)
| | | | | | | | | | - Brandon S. McCullough
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; (M.S.); (J.R.); (R.K.); (T.H.)
| |
Collapse
|
8
|
Iuso D, Guilliaumet J, Schlattner U, Khochbin S. Nucleoside Diphosphate Kinases Are ATP-Regulated Carriers of Short-Chain Acyl-CoAs. Int J Mol Sci 2024; 25:7528. [PMID: 39062771 PMCID: PMC11277454 DOI: 10.3390/ijms25147528] [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: 05/24/2024] [Revised: 06/30/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Nucleoside diphosphate (NDP) kinases 1 and 2 (NME1/2) are well-characterized enzymes known for their NDP kinase activity. Recently, these enzymes have been shown by independent studies to bind coenzyme A (CoA) or acyl-CoA. These findings suggest a hitherto unknown role for NME1/2 in the regulation of CoA/acyl-CoA-dependent metabolic pathways, in tight correlation with the cellular NTP/NDP ratio. Accordingly, the regulation of NME1/2 functions by CoA/acyl-CoA binding has been described, and additionally, NME1/2 have been shown to control the cellular pathways consuming acetyl-CoA, such as histone acetylation and fatty acid synthesis. NME1/2-controlled histone acetylation in turn mediates an important transcriptional response to metabolic changes, such as those induced following a high-fat diet (HFD). This review discusses the CoA/acyl-CoA-dependent NME1/2 activities and proposes that these enzymes be considered as the first identified carriers of CoA/short-chain acyl-CoAs.
Collapse
Affiliation(s)
- Domenico Iuso
- University of Teramo, Department of Veterinary Medicine, 64100 Teramo, Italy
| | - Julie Guilliaumet
- University Grenoble-Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, 38706 La Tronche, France; (J.G.); (S.K.)
| | - Uwe Schlattner
- University Grenoble-Alpes, INSERM U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France;
- Institut Universitaire de France, 75231 Paris, France
| | - Saadi Khochbin
- University Grenoble-Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, 38706 La Tronche, France; (J.G.); (S.K.)
| |
Collapse
|
9
|
Chi JT, Lin CC, Lin YT, Chen SY, Setayeshpour Y, Chen Y, Dunn D, Soderblom E, Zhang GF, Filonenko V, Jeong SY, Floyd S, Hayflick S, Gout I. Coenzyme A protects against ferroptosis via CoAlation of thioredoxin reductase 2. RESEARCH SQUARE 2024:rs.3.rs-4522617. [PMID: 38947036 PMCID: PMC11213209 DOI: 10.21203/rs.3.rs-4522617/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The Cystine-xCT transporter-Glutathione (GSH)-GPX4 axis is the canonical pathway to protect against ferroptosis. While not required for ferroptosis-inducing compounds (FINs) targeting GPX4, FINs targeting the xCT transporter require mitochondria and its lipid peroxidation to trigger ferroptosis. However, the mechanism underlying the difference between these FINs is still unknown. Given that cysteine is also required for coenzyme A (CoA) biosynthesis, here we show that CoA supplementation specifically prevents ferroptosis induced by xCT inhibitors but not GPX4 inhibitors. We find that, auranofin, a thioredoxin reductase inhibitor, abolishes the protective effect of CoA. We also find that CoA availability determines the enzymatic activity of thioredoxin reductase, but not thioredoxin. Importantly, the mitochondrial thioredoxin system, but not the cytosolic thioredoxin system, determines CoA-mediated ferroptosis inhibition. Our data show that the CoA regulates the in vitro enzymatic activity of mitochondrial thioredoxin reductase (TXNRD2) by covalently modifying the thiol group of cysteine (CoAlation) on Cys-483. Replacing Cys-483 with alanine on TXNRD2 abolishes its in vitro enzymatic activity and ability to protect cells from ferroptosis. Targeting xCT to limit cysteine import and, therefore, CoA biosynthesis reduced CoAlation on TXNRD2, an effect that was rescued by CoA supplementation. Furthermore, the fibroblasts from patients with disrupted CoA metabolism demonstrate increased mitochondrial lipid peroxidation. In organotypic brain slice cultures, inhibition of CoA biosynthesis leads to an oxidized thioredoxin system, mitochondrial lipid peroxidation, and loss in cell viability, which were all rescued by ferrostatin-1. These findings identify CoA-mediated post-translation modification to regulate the thioredoxin system as an alternative ferroptosis protection pathway with potential clinical relevance for patients with disrupted CoA metabolism.
Collapse
|
10
|
Barritt SA, DuBois-Coyne SE, Dibble CC. Coenzyme A biosynthesis: mechanisms of regulation, function and disease. Nat Metab 2024; 6:1008-1023. [PMID: 38871981 DOI: 10.1038/s42255-024-01059-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 04/30/2024] [Indexed: 06/15/2024]
Abstract
The tricarboxylic acid cycle, nutrient oxidation, histone acetylation and synthesis of lipids, glycans and haem all require the cofactor coenzyme A (CoA). Although the sources and regulation of the acyl groups carried by CoA for these processes are heavily studied, a key underlying question is less often considered: how is production of CoA itself controlled? Here, we discuss the many cellular roles of CoA and the regulatory mechanisms that govern its biosynthesis from cysteine, ATP and the essential nutrient pantothenate (vitamin B5), or from salvaged precursors in mammals. Metabolite feedback and signalling mechanisms involving acetyl-CoA, other acyl-CoAs, acyl-carnitines, MYC, p53, PPARα, PINK1 and insulin- and growth factor-stimulated PI3K-AKT signalling regulate the vitamin B5 transporter SLC5A6/SMVT and CoA biosynthesis enzymes PANK1, PANK2, PANK3, PANK4 and COASY. We also discuss methods for measuring CoA-related metabolites, compounds that target CoA biosynthesis and diseases caused by mutations in pathway enzymes including types of cataracts, cardiomyopathy and neurodegeneration (PKAN and COPAN).
Collapse
Affiliation(s)
- Samuel A Barritt
- Department of Pathology, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sarah E DuBois-Coyne
- Department of Medicine, Department of Biological Chemistry and Molecular Pharmacology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christian C Dibble
- Department of Pathology, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Ferrucci V, Lomada S, Wieland T, Zollo M. PRUNE1 and NME/NDPK family proteins influence energy metabolism and signaling in cancer metastases. Cancer Metastasis Rev 2024; 43:755-775. [PMID: 38180572 PMCID: PMC11156750 DOI: 10.1007/s10555-023-10165-4] [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: 10/10/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024]
Abstract
We describe here the molecular basis of the complex formation of PRUNE1 with the tumor metastasis suppressors NME1 and NME2, two isoforms appertaining to the nucleoside diphosphate kinase (NDPK) enzyme family, and how this complex regulates signaling the immune system and energy metabolism, thereby shaping the tumor microenvironment (TME). Disrupting the interaction between NME1/2 and PRUNE1, as suggested, holds the potential to be an excellent therapeutic target for the treatment of cancer and the inhibition of metastasis dissemination. Furthermore, we postulate an interaction and regulation of the other Class I NME proteins, NME3 and NME4 proteins, with PRUNE1 and discuss potential functions. Class I NME1-4 proteins are NTP/NDP transphosphorylases required for balancing the intracellular pools of nucleotide diphosphates and triphosphates. They regulate different cellular functions by interacting with a large variety of other proteins, and in cancer and metastasis processes, they can exert pro- and anti-oncogenic properties depending on the cellular context. In this review, we therefore additionally discuss general aspects of class1 NME and PRUNE1 molecular structures as well as their posttranslational modifications and subcellular localization. The current knowledge on the contributions of PRUNE1 as well as NME proteins to signaling cascades is summarized with a special regard to cancer and metastasis.
Collapse
Affiliation(s)
- Veronica Ferrucci
- Department of Molecular Medicine and Medical Biotechnology, DMMBM, University of Naples, Federico II, Via Pansini 5, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate "Franco Salvatore", Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Santosh Lomada
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
- DZHK, German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, 68167, Mannheim, Germany
| | - Thomas Wieland
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany.
- DZHK, German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, 68167, Mannheim, Germany.
- Medical Faculty Mannheim, Ludolf Krehl-Str. 13-17, 68167, Mannheim, Germany.
| | - Massimo Zollo
- Department of Molecular Medicine and Medical Biotechnology, DMMBM, University of Naples, Federico II, Via Pansini 5, 80131, Naples, Italy.
- CEINGE Biotecnologie Avanzate "Franco Salvatore", Via Gaetano Salvatore 486, 80145, Naples, Italy.
- DAI Medicina di Laboratorio e Trasfusionale, 'AOU' Federico II Policlinico, 80131, Naples, Italy.
| |
Collapse
|
12
|
Tossounian MA, Zhao Y, Yu BYK, Markey SA, Malanchuk O, Zhu Y, Cain A, Gout I. Low-molecular-weight thiol transferases in redox regulation and antioxidant defence. Redox Biol 2024; 71:103094. [PMID: 38479221 PMCID: PMC10950700 DOI: 10.1016/j.redox.2024.103094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/08/2024] [Accepted: 02/18/2024] [Indexed: 03/24/2024] Open
Abstract
Low-molecular-weight (LMW) thiols are produced in all living cells in different forms and concentrations. Glutathione (GSH), coenzyme A (CoA), bacillithiol (BSH), mycothiol (MSH), ergothioneine (ET) and trypanothione T(SH)2 are the main LMW thiols in eukaryotes and prokaryotes. LMW thiols serve as electron donors for thiol-dependent enzymes in redox-mediated metabolic and signaling processes, protect cellular macromolecules from oxidative and xenobiotic stress, and participate in the reduction of oxidative modifications. The level and function of LMW thiols, their oxidized disulfides and mixed disulfide conjugates in cells and tissues is tightly controlled by dedicated oxidoreductases, such as peroxiredoxins, glutaredoxins, disulfide reductases and LMW thiol transferases. This review provides the first summary of the current knowledge of structural and functional diversity of transferases for LMW thiols, including GSH, BSH, MSH and T(SH)2. Their role in maintaining redox homeostasis in single-cell and multicellular organisms is discussed, focusing in particular on the conjugation of specific thiols to exogenous and endogenous electrophiles, or oxidized protein substrates. Advances in the development of new research tools, analytical methodologies, and genetic models for the analysis of known LMW thiol transferases will expand our knowledge and understanding of their function in cell growth and survival under oxidative stress, nutrient deprivation, and during the detoxification of xenobiotics and harmful metabolites. The antioxidant function of CoA has been recently discovered and the breakthrough in defining the identity and functional characteristics of CoA S-transferase(s) is soon expected.
Collapse
Affiliation(s)
- Maria-Armineh Tossounian
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Yuhan Zhao
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Bess Yi Kun Yu
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Samuel A Markey
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Oksana Malanchuk
- 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
| | - Yuejia Zhu
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, United Kingdom
| | - Amanda Cain
- Department of Structural and Molecular Biology, University College London, London, WC1E 6BT, 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
|
13
|
Zhang Y, Zhao G, Yu L, Wang X, Meng Y, Mao J, Fu Z, Yin Y, Li J, Wang X, Guo C. Heat-shock protein 90α protects NME1 against degradation and suppresses metastasis of breast cancer. Br J Cancer 2023; 129:1679-1691. [PMID: 37731021 PMCID: PMC10645775 DOI: 10.1038/s41416-023-02435-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 08/28/2023] [Accepted: 09/11/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND NME1 has been exploited as a potential translational target for decades. Substantial efforts have been made to upregulate the expression of NME1 and restore its anti-metastasis function in metastatic cancer. METHODS Cycloheximide (CHX) chase assay was used to measure the steady-state protein stability of NME1 and HSP90α. The NME1-associating proteins were identified by immunoprecipitation combined with mass spectrometric analysis. Gene knockdown and overexpression were employed to examine the impact of HSP90AA1 on intracellular NME1 degradation. The motility and invasiveness of breast cancer cells were examined in vitro using wound healing and transwell invasion assays. The orthotopic spontaneous metastasis and intra-venous experimental metastasis assays were used to test the formation of metastasis in vivo, respectively. RESULTS HSP90α interacts with NME1 and increases NME1 lifetime by impeding its ubiquitin-proteasome-mediated degradation. HSP90α overexpression significantly inhibits the metastatic potential of breast cancer cells in vitro and in vivo. A novel cell-permeable peptide, OPT22 successfully mimics the HSP90α function and prolongs the life span of endogenous NME1, resulting in reduced metastasis of breast cancer. CONCLUSION These results not only reveal a new mechanism of NME1 degradation but also pave the way for the development of new and effective approaches to metastatic cancer therapy.
Collapse
Affiliation(s)
- Yanchao Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, People's Republic of China
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, People's Republic of China
| | - Guomeng Zhao
- Institute of Modern Biology, Nanjing University, Nanjing, People's Republic of China
| | - Liting Yu
- Department of Protein and Antibody Engineering, School of Pharmacy, Binzhou Medical University, Yantai, People's Republic of China
| | - Xindong Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yao Meng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jinlei Mao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Ziyi Fu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing People's Hospital, Nanjing, People's Republic of China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing People's Hospital, Nanjing, People's Republic of China.
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, People's Republic of China.
| | - Xun Wang
- Department of Hepatobiliary Surgery, National Cancer Center, National Clinical Research Center for Cancer, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| | - Changying Guo
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, People's Republic of China.
| |
Collapse
|
14
|
Iuso D, Garcia-Saez I, Couté Y, Yamaryo-Botté Y, Boeri Erba E, Adrait A, Zeaiter N, Tokarska-Schlattner M, Jilkova ZM, Boussouar F, Barral S, Signor L, Couturier K, Hajmirza A, Chuffart F, Bourova-Flin E, Vitte AL, Bargier L, Puthier D, Decaens T, Rousseaux S, Botté C, Schlattner U, Petosa C, Khochbin S. Nucleoside diphosphate kinases 1 and 2 regulate a protective liver response to a high-fat diet. SCIENCE ADVANCES 2023; 9:eadh0140. [PMID: 37672589 PMCID: PMC10482350 DOI: 10.1126/sciadv.adh0140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 08/02/2023] [Indexed: 09/08/2023]
Abstract
The synthesis of fatty acids from acetyl-coenzyme A (AcCoA) is deregulated in diverse pathologies, including cancer. Here, we report that fatty acid accumulation is negatively regulated by nucleoside diphosphate kinases 1 and 2 (NME1/2), housekeeping enzymes involved in nucleotide homeostasis that were recently found to bind CoA. We show that NME1 additionally binds AcCoA and that ligand recognition involves a unique binding mode dependent on the CoA/AcCoA 3' phosphate. We report that Nme2 knockout mice fed a high-fat diet (HFD) exhibit excessive triglyceride synthesis and liver steatosis. In liver cells, NME2 mediates a gene transcriptional response to HFD leading to the repression of fatty acid accumulation and activation of a protective gene expression program via targeted histone acetylation. Our findings implicate NME1/2 in the epigenetic regulation of a protective liver response to HFD and suggest a potential role in controlling AcCoA usage between the competing paths of histone acetylation and fatty acid synthesis.
Collapse
Affiliation(s)
- Domenico Iuso
- Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, La Tronche 38706, France
| | - Isabel Garcia-Saez
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), Grenoble 38000, France
| | - Yohann Couté
- Univ. Grenoble Alpes, INSERM, CEA, UMR BioSanté U1292, CNRS, CEA, FR2048, Grenoble 38000, France
| | - Yoshiki Yamaryo-Botté
- Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, La Tronche 38706, France
| | - Elisabetta Boeri Erba
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), Grenoble 38000, France
| | - Annie Adrait
- Univ. Grenoble Alpes, INSERM, CEA, UMR BioSanté U1292, CNRS, CEA, FR2048, Grenoble 38000, France
| | - Nour Zeaiter
- Univ. Grenoble Alpes, INSERM, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
| | | | - Zuzana Macek Jilkova
- Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, La Tronche 38706, France
- CHU Grenoble Alpes, Service d’hépato-gastroentérologie, Pôle Digidune, La Tronche 38700, France
| | - Fayçal Boussouar
- Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, La Tronche 38706, France
| | - Sophie Barral
- Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, La Tronche 38706, France
| | - Luca Signor
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), Grenoble 38000, France
| | - Karine Couturier
- Univ. Grenoble Alpes, INSERM, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
| | - Azadeh Hajmirza
- Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, La Tronche 38706, France
| | - Florent Chuffart
- Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, La Tronche 38706, France
| | - Ekaterina Bourova-Flin
- Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, La Tronche 38706, France
| | - Anne-Laure Vitte
- Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, La Tronche 38706, France
| | - Lisa Bargier
- Aix Marseille Université, INSERM, TAGC, TGML, Marseille 13288, France
| | - Denis Puthier
- Aix Marseille Université, INSERM, TAGC, TGML, Marseille 13288, France
| | - Thomas Decaens
- Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, La Tronche 38706, France
- CHU Grenoble Alpes, Service d’hépato-gastroentérologie, Pôle Digidune, La Tronche 38700, France
| | - Sophie Rousseaux
- Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, La Tronche 38706, France
| | - Cyrille Botté
- Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, La Tronche 38706, France
| | - Uwe Schlattner
- Univ. Grenoble Alpes, INSERM, Institut Universitaire de France, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
| | - Carlo Petosa
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), Grenoble 38000, France
| | - Saadi Khochbin
- Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, La Tronche 38706, France
| |
Collapse
|
15
|
Schmelter C, Fomo KN, Brueck A, Perumal N, Markowitsch SD, Govind G, Speck T, Pfeiffer N, Grus FH. Glaucoma-Associated CDR1 Peptide Promotes RGC Survival in Retinal Explants through Molecular Interaction with Acidic Leucine Rich Nuclear Phosphoprotein 32A (ANP32A). Biomolecules 2023; 13:1161. [PMID: 37509196 PMCID: PMC10377047 DOI: 10.3390/biom13071161] [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: 05/18/2023] [Revised: 07/06/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Glaucoma is a complex, multifactorial optic neuropathy mainly characterized by the progressive loss of retinal ganglion cells (RGCs) and their axons, resulting in a decline of visual function. The pathogenic molecular mechanism of glaucoma is still not well understood, and therapeutic strategies specifically addressing the neurodegenerative component of this ocular disease are urgently needed. Novel immunotherapeutics might overcome this problem by targeting specific molecular structures in the retina and providing direct neuroprotection via different modes of action. Within the scope of this research, the present study showed for the first time beneficial effects of the synthetic CDR1 peptide SCTGTSSDVGGYNYVSWYQ on the viability of RGCs ex vivo in a concentration-dependent manner compared to untreated control explants (CTRL, 50 µg/mL: p < 0.05 and 100 µg/mL: p < 0.001). Thereby, this specific peptide was identified first as a potential biomarker candidate in the serum of glaucoma patients and was significantly lower expressed in systemic IgG molecules compared to healthy control subjects. Furthermore, MS-based co-immunoprecipitation experiments confirmed the specific interaction of synthetic CDR1 with retinal acidic leucine-rich nuclear phosphoprotein 32A (ANP32A; p < 0.001 and log2 fold change > 3), which is a highly expressed protein in neurological tissues with multifactorial biological functions. In silico binding prediction analysis revealed the N-terminal leucine-rich repeat (LRR) domain of ANP32A as a significant binding site for synthetic CDR1, which was previously reported as an important docking site for protein-protein interactions (PPI). In accordance with these findings, quantitative proteomic analysis of the retinae ± CDR1 treatment resulted in the identification of 25 protein markers, which were significantly differentially distributed between both experimental groups (CTRL and CDR1, p < 0.05). Particularly, acetyl-CoA biosynthesis I-related enzymes (e.g., DLAT and PDHA1), as well as cytoskeleton-regulating proteins (e.g., MSN), were highly expressed by synthetic CDR1 treatment in the retina; on the contrary, direct ANP32A-interacting proteins (e.g., NME1 and PPP2R4), as well as neurodegenerative-related markers (e.g., CEND1), were identified with significant lower abundancy in the CDR1-treated retinae compared to CTRL. Furthermore, retinal protein phosphorylation and histone acetylation were also affected by synthetic CDR1, which are both partially controlled by ANP32A. In conclusion, the synthetic CDR1 peptide provides a great translational potential for the treatment of glaucoma in the future by eliciting its neuroprotective mechanism via specific interaction with ANP32A's N terminal LRR domain.
Collapse
Affiliation(s)
- Carsten Schmelter
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (C.S.); (K.N.F.); (A.B.); (N.P.); (N.P.)
| | - Kristian Nzogang Fomo
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (C.S.); (K.N.F.); (A.B.); (N.P.); (N.P.)
| | - Alina Brueck
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (C.S.); (K.N.F.); (A.B.); (N.P.); (N.P.)
| | - Natarajan Perumal
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (C.S.); (K.N.F.); (A.B.); (N.P.); (N.P.)
| | - Sascha D. Markowitsch
- Department of Urology and Pediatric Urology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Gokul Govind
- Institute of Physics, Johannes Gutenberg University, 55131 Mainz, Germany; (G.G.)
| | - Thomas Speck
- Institute of Physics, Johannes Gutenberg University, 55131 Mainz, Germany; (G.G.)
| | - Norbert Pfeiffer
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (C.S.); (K.N.F.); (A.B.); (N.P.); (N.P.)
| | - Franz H. Grus
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (C.S.); (K.N.F.); (A.B.); (N.P.); (N.P.)
| |
Collapse
|
16
|
Tossounian MA, Hristov SD, Semelak JA, Yu BYK, Baczynska M, Zhao Y, Estrin DA, Trujillo M, Filonenko V, Gouge J, Gout I. A Unique Mode of Coenzyme A Binding to the Nucleotide Binding Pocket of Human Metastasis Suppressor NME1. Int J Mol Sci 2023; 24:9359. [PMID: 37298313 PMCID: PMC10253429 DOI: 10.3390/ijms24119359] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/21/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Coenzyme A (CoA) is a key cellular metabolite which participates in diverse metabolic pathways, regulation of gene expression and the antioxidant defense mechanism. Human NME1 (hNME1), which is a moonlighting protein, was identified as a major CoA-binding protein. Biochemical studies showed that hNME1 is regulated by CoA through both covalent and non-covalent binding, which leads to a decrease in the hNME1 nucleoside diphosphate kinase (NDPK) activity. In this study, we expanded the knowledge on previous findings by focusing on the non-covalent mode of CoA binding to the hNME1. With X-ray crystallography, we solved the CoA bound structure of hNME1 (hNME1-CoA) and determined the stabilization interactions CoA forms within the nucleotide-binding site of hNME1. A hydrophobic patch stabilizing the CoA adenine ring, while salt bridges and hydrogen bonds stabilizing the phosphate groups of CoA were observed. With molecular dynamics studies, we extended our structural analysis by characterizing the hNME1-CoA structure and elucidating possible orientations of the pantetheine tail, which is absent in the X-ray structure due to its flexibility. Crystallographic studies suggested the involvement of arginine 58 and threonine 94 in mediating specific interactions with CoA. Site-directed mutagenesis and CoA-based affinity purifications showed that arginine 58 mutation to glutamate (R58E) and threonine 94 mutation to aspartate (T94D) prevent hNME1 from binding to CoA. Overall, our results reveal a unique mode by which hNME1 binds CoA, which differs significantly from that of ADP binding: the α- and β-phosphates of CoA are oriented away from the nucleotide-binding site, while 3'-phosphate faces catalytic histidine 118 (H118). The interactions formed by the CoA adenine ring and phosphate groups contribute to the specific mode of CoA binding to hNME1.
Collapse
Affiliation(s)
- Maria-Armineh Tossounian
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (S.D.H.); (B.Y.K.Y.); (M.B.); (Y.Z.); (I.G.)
| | - Stefan Denchev Hristov
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (S.D.H.); (B.Y.K.Y.); (M.B.); (Y.Z.); (I.G.)
| | - Jonathan Alexis Semelak
- Departmento de Química Inorgánica Analítica y Química Física, Instituto de Química Física de los Materiales, Medioambiente y Energía (INQUIMAE) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Universitaria, Pab. 2 C1428EHA, Buenos Aires 1865, Argentina; (J.A.S.); (D.A.E.)
| | - Bess Yi Kun Yu
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (S.D.H.); (B.Y.K.Y.); (M.B.); (Y.Z.); (I.G.)
| | - Maria Baczynska
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (S.D.H.); (B.Y.K.Y.); (M.B.); (Y.Z.); (I.G.)
| | - Yuhan Zhao
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (S.D.H.); (B.Y.K.Y.); (M.B.); (Y.Z.); (I.G.)
| | - Dario Ariel Estrin
- Departmento de Química Inorgánica Analítica y Química Física, Instituto de Química Física de los Materiales, Medioambiente y Energía (INQUIMAE) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Universitaria, Pab. 2 C1428EHA, Buenos Aires 1865, Argentina; (J.A.S.); (D.A.E.)
| | - Madia Trujillo
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay;
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Valeriy Filonenko
- Department of Cell Signaling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03680 Kyiv, Ukraine;
| | - Jerome Gouge
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (S.D.H.); (B.Y.K.Y.); (M.B.); (Y.Z.); (I.G.)
- Institute of Structural and Molecular Biology, Birkbeck College, University of London, London WC1E 7HX, UK
| | - Ivan Gout
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (S.D.H.); (B.Y.K.Y.); (M.B.); (Y.Z.); (I.G.)
- Department of Cell Signaling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03680 Kyiv, Ukraine;
| |
Collapse
|
17
|
Tossounian MA, Baczynska M, Dalton W, Peak-Chew SY, Undzenas K, Korza G, Filonenko V, Skehel M, Setlow P, Gout I. Bacillus subtilis YtpP and Thioredoxin A Are New Players in the Coenzyme-A-Mediated Defense Mechanism against Cellular Stress. Antioxidants (Basel) 2023; 12:antiox12040938. [PMID: 37107313 PMCID: PMC10136147 DOI: 10.3390/antiox12040938] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Coenzyme A (CoA) is an important cellular metabolite that is critical for metabolic processes and the regulation of gene expression. Recent discovery of the antioxidant function of CoA has highlighted its protective role that leads to the formation of a mixed disulfide bond with protein cysteines, which is termed protein CoAlation. To date, more than 2000 CoAlated bacterial and mammalian proteins have been identified in cellular responses to oxidative stress, with the majority being involved in metabolic pathways (60%). Studies have shown that protein CoAlation is a widespread post-translational modification which modulates the activity and conformation of the modified proteins. The induction of protein CoAlation by oxidative stress was found to be rapidly reversed after the removal of oxidizing agents from the medium of cultured cells. In this study, we developed an enzyme-linked immunosorbent assay (ELISA)-based deCoAlation assay to detect deCoAlation activity from Bacillus subtilis and Bacillus megaterium lysates. We then used a combination of ELISA-based assay and purification strategies to show that deCoAlation is an enzyme-driven mechanism. Using mass-spectrometry and deCoAlation assays, we identified B. subtilis YtpP (thioredoxin-like protein) and thioredoxin A (TrxA) as enzymes that can remove CoA from different substrates. With mutagenesis studies, we identified YtpP and TrxA catalytic cysteine residues and proposed a possible deCoAlation mechanism for CoAlated methionine sulfoxide reducatse A (MsrA) and peroxiredoxin 5 (PRDX5) proteins, which results in the release of both CoA and the reduced form of MsrA or PRDX5. Overall, this paper reveals the deCoAlation activity of YtpP and TrxA and opens doors to future studies on the CoA-mediated redox regulation of CoAlated proteins under various cellular stress conditions.
Collapse
Affiliation(s)
| | - Maria Baczynska
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - William Dalton
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Sew Yeu Peak-Chew
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Kipras Undzenas
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - George Korza
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT 06030, USA
| | - Valeriy Filonenko
- Department of Cell Signaling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03680 Kyiv, Ukraine
| | - Mark Skehel
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Peter Setlow
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT 06030, USA
| | - Ivan Gout
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK
- Department of Cell Signaling, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03680 Kyiv, Ukraine
| |
Collapse
|
18
|
Song HC, Xie CY, Kong Q, Wei L, Wang XT. Daylight ultraviolet B radiation ruptured the cell membrane, promoted nucleotide metabolism and inhibited energy metabolism in the plasma of Pacific oyster. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 862:160729. [PMID: 36496017 DOI: 10.1016/j.scitotenv.2022.160729] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/28/2022] [Accepted: 12/02/2022] [Indexed: 06/17/2023]
Abstract
The increasing and intensifying ultraviolet B (UVB) radiation in sunlight is an environmental threat to aquatic ecosystems, potentially affecting the entire life cycle of wild or aquacultural Pacific oyster Crassostrea gigas with photoreception. Due to its complex composition, plasma is an important biological specimen for investigating the degree of disturbance from its steady state caused by the external environment in the open-pipe-type hemolymph of mollusks. We performed a multi-omic analysis of C. gigas plasma exposed to daylight UVB radiation. Hub differentially expressed genes (DEGs) and differentially expressed proteins (DEPs) were identified using the functional classification of Clusters of Orthologous Groups of proteins (COGs) through the protein-protein interaction (PPI)-based maximal clique centrality (MCC) algorithm. Our results summarize three types of UVB influences (disruption of the cell membrane, promotion of nucleotide metabolism, and inhibition of energy metabolism) on C. gigas based on transcriptomic, proteomic, and metabolomic analyses. The associated hub DEGs, DEPs (e.g., nucleoside diphosphate kinase, malate dehydrogenase, and hydroxyacyl-coenzyme A dehydrogenase), and metabolites (e.g., uridine, adenine, deoxyguanosine, guanosine, and xylitol) in the plasma were identified as biomarkers of mollusk response to UVB radiation, and could be used to evaluate the influence of environmental UVB on mollusks in future studies.
Collapse
Affiliation(s)
- Hong-Ce Song
- School of Agriculture, Ludong University, Yantai City, Shandong Province 264025, China
| | - Chao-Yi Xie
- School of Agriculture, Ludong University, Yantai City, Shandong Province 264025, China
| | - Qing Kong
- School of Agriculture, Ludong University, Yantai City, Shandong Province 264025, China
| | - Lei Wei
- School of Agriculture, Ludong University, Yantai City, Shandong Province 264025, China.
| | - Xiao-Tong Wang
- School of Agriculture, Ludong University, Yantai City, Shandong Province 264025, China.
| |
Collapse
|
19
|
Dong X, Li Y, Li Q, Li W, Wu G. Identification of immune signatures in Parkinson's disease based on co-expression networks. Front Genet 2023; 14:1090382. [PMID: 36733342 PMCID: PMC9886886 DOI: 10.3389/fgene.2023.1090382] [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: 11/05/2022] [Accepted: 01/09/2023] [Indexed: 01/18/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease in middle-aged and elderly people, and there is less research on the relationship between immunity and PD. In this study, the protein-protein interaction networks (PPI) data, 2747 human immune-related genes (HIRGs), 2078 PD-related genes (PDRGs), and PD-related datasets (GSE49036 and GSE20292) were downloaded from the Human Protein Reference Database (HPRD), Amigo 2, DisGeNET, and Gene Expression Omnibus (GEO) databases, respectively. An immune- or PD-directed neighbor co-expressed network construction (IOPDNC) was drawn based on the GSE49036 dataset and HPRD database. Furthermore, a PD-directed neighbor co-expressed network was constructed. Modular clustering analysis was performed on the genes of the gene interaction network obtained in the first step to obtain the central core genes using the GraphWeb online website. The modules with the top 5 functional scores and the number of core genes greater than six were selected as PD-related gene modules. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of different module genes were performed. The single sample Gene Set Enrichment Analysis (ssGSEA) algorithm was used to calculate the immune cell infiltration of the PD and the normal samples. The quantitative Reverse Transcription Polymerase Chain Reaction (qRT-PCR) was performed to investigate the expression of module genes. An IOPDNC and PD-directed neighbor co-expressed network (PDNC network) were constructed. Furthermore, a total of 5 immune-PD modules were identified which could distinguish between PD and normal samples, and these module genes were strongly related to PD in protein interaction level or gene expression level. In addition, functional analysis indicated that module genes were involved in various neurodegenerative diseases, such as Alzheimer disease, Huntington disease, Parkinson disease, and Long-term depression. In addition, the genes of the 6 modules were significantly associated with these 4 differential immune cells (aDC cells, eosinophils, neutrophils, and Th2 cells). Finally, the result of qRT-PCR manifested that the expression of 6 module genes was significantly higher in normal samples than in PD samples. In our study, the immune-related genes were found to be strongly related to PD and might play key roles in PD.
Collapse
|
20
|
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
|
21
|
Rashed SA, Hammad SF, Eldakak MM, Khalil IA, Osman A. Assessment of the Anticancer Potentials of the Free and Metal-Organic Framework (UiO-66) - Delivered Phycocyanobilin. J Pharm Sci 2023; 112:213-224. [PMID: 36087776 DOI: 10.1016/j.xphs.2022.08.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/31/2022] [Accepted: 08/31/2022] [Indexed: 10/14/2022]
Abstract
Phycocyanin (C-PC) is a constitutive chromoprotein of Arthrospira platensis, which exhibits promising efficacy against different types of cancer. In this study, we cleaved C-PC's chromophore phycocyanobilin (PCB) and demonstrated its ability as an anti-cancer drug for Colorectal cancer (CRC). PCB displayed an anti-cancer effect for CRC (HT-29) cells with IC50 of 108 µg/ml. Assessing the transcripts levels of some biomarkers revealed that the PCB caused an upregulation in the anti-metastatic gene NME1 level and downregulation of the COX-2 level. The flow cytometric results showed the effect of PCB on the arrest of the cell cycle's G1 phase. In addition, we successfully synthesized the UiO-66 (Zr-MOF). We incorporated the PCB into UiO-66 nanoparticles with a loading percentage of 46 %. Assessment of the cytotoxic effects of UiO-66@PCB showed a 2-fold improvement in the IC50 compared to the free PCB. In conclusion, we have shown that PCB displayed a promising potential as an anti-cancer agent. Yet, it is considered a safe and natural substance that can help to mitigate cancer spread and symptoms. In the meantime, UiO-66 can be used as a safe nano-delivery tool for PCB.
Collapse
Affiliation(s)
- Suzan A Rashed
- Biotechnology Program, Institute of Basic and Applied Sciences, Egypt-Japan University of Science and Technology, Borg El-Arab, Egypt; Botany and Microbiology Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Sherif F Hammad
- Biotechnology Program, Institute of Basic and Applied Sciences, Egypt-Japan University of Science and Technology, Borg El-Arab, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Moustafa M Eldakak
- Genetics Department, Faculty of Agriculture, Alexandria University, Alexandria, Egypt
| | - Islam A Khalil
- Pharmaceutics Department, Faculty of Pharmacy and Drug Manufacturing, Misr University for Science and Technology, 6 October, Egypt
| | - Ahmed Osman
- Biotechnology Program, Institute of Basic and Applied Sciences, Egypt-Japan University of Science and Technology, Borg El-Arab, Egypt; Department of Biochemistry, Faculty of Science, Ain shams University, Cairo, Egypt
| |
Collapse
|
22
|
Tossounian MA, Baczynska M, Dalton W, Newell C, Ma Y, Das S, Semelak JA, Estrin DA, Filonenko V, Trujillo M, Peak-Chew SY, Skehel M, Fraternali F, Orengo C, Gout I. Profiling the Site of Protein CoAlation and Coenzyme A Stabilization Interactions. Antioxidants (Basel) 2022; 11:antiox11071362. [PMID: 35883853 PMCID: PMC9312308 DOI: 10.3390/antiox11071362] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/26/2022] [Accepted: 06/26/2022] [Indexed: 11/30/2022] Open
Abstract
Coenzyme A (CoA) is a key cellular metabolite known for its diverse functions in metabolism and regulation of gene expression. CoA was recently shown to play an important antioxidant role under various cellular stress conditions by forming a disulfide bond with proteins, termed CoAlation. Using anti-CoA antibodies and liquid chromatography tandem mass spectrometry (LC-MS/MS) methodologies, CoAlated proteins were identified from various organisms/tissues/cell-lines under stress conditions. In this study, we integrated currently known CoAlated proteins into mammalian and bacterial datasets (CoAlomes), resulting in a total of 2093 CoAlated proteins (2862 CoAlation sites). Functional classification of these proteins showed that CoAlation is widespread among proteins involved in cellular metabolism, stress response and protein synthesis. Using 35 published CoAlated protein structures, we studied the stabilization interactions of each CoA segment (adenosine diphosphate (ADP) moiety and pantetheine tail) within the microenvironment of the modified cysteines. Alternating polar-non-polar residues, positively charged residues and hydrophobic interactions mainly stabilize the pantetheine tail, phosphate groups and the ADP moiety, respectively. A flexible nature of CoA is observed in examined structures, allowing it to adapt its conformation through interactions with residues surrounding the CoAlation site. Based on these findings, we propose three modes of CoA binding to proteins. Overall, this study summarizes currently available knowledge on CoAlated proteins, their functional distribution and CoA-protein stabilization interactions.
Collapse
Affiliation(s)
- Maria-Armineh Tossounian
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (M.-A.T.); (M.B.); (W.D.); (C.N.); (Y.M.); (S.D.); (C.O.)
| | - Maria Baczynska
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (M.-A.T.); (M.B.); (W.D.); (C.N.); (Y.M.); (S.D.); (C.O.)
| | - William Dalton
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (M.-A.T.); (M.B.); (W.D.); (C.N.); (Y.M.); (S.D.); (C.O.)
| | - Charlie Newell
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (M.-A.T.); (M.B.); (W.D.); (C.N.); (Y.M.); (S.D.); (C.O.)
| | - Yilin Ma
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (M.-A.T.); (M.B.); (W.D.); (C.N.); (Y.M.); (S.D.); (C.O.)
| | - Sayoni Das
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (M.-A.T.); (M.B.); (W.D.); (C.N.); (Y.M.); (S.D.); (C.O.)
| | - Jonathan Alexis Semelak
- Departmento de Química Inorgánica Analítica y Química Física, INQUIMAE-CONICET, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; (J.A.S.); (D.A.E.)
| | - Dario Ariel Estrin
- Departmento de Química Inorgánica Analítica y Química Física, INQUIMAE-CONICET, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; (J.A.S.); (D.A.E.)
| | - Valeriy Filonenko
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03680 Kyiv, Ukraine;
| | - Madia Trujillo
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay;
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Sew Yeu Peak-Chew
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK;
| | - Mark Skehel
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK;
| | - Franca Fraternali
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London WC2R 2LS, UK;
| | - Christine Orengo
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (M.-A.T.); (M.B.); (W.D.); (C.N.); (Y.M.); (S.D.); (C.O.)
| | - Ivan Gout
- Department of Structural and Molecular Biology, University College London, London WC1E 6BT, UK; (M.-A.T.); (M.B.); (W.D.); (C.N.); (Y.M.); (S.D.); (C.O.)
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 03680 Kyiv, Ukraine;
- Correspondence:
| |
Collapse
|
23
|
Functional mechanism and clinical implications of miR-141 in human cancers. Cell Signal 2022; 95:110354. [PMID: 35550172 DOI: 10.1016/j.cellsig.2022.110354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/22/2022] [Accepted: 05/03/2022] [Indexed: 11/20/2022]
Abstract
Cancer is caused by the abnormal proliferation of local tissue cells under the control of many oncogenic factors. MicroRNAs (miRNAs) are a class of evolutionarily conserved, approximately 22-nucleotide noncoding small RNAs that influence transcriptional regulationby binding to the 3'-untranslated region of target messenger RNA. As a member of the miRNA family, miR-141 acts as a suppressor or an oncomiR in various cancers and regulates cancer cell proliferation, apoptosis, invasion, and metastasis through a variety of signaling pathways, such as phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) and constitutive activation of nuclear factor-κB (NF-κB). Target gene validation and pathway analysis have provided mechanistic insight into the role of this miRNA in different tissues. This review also outlines novel findings that suggest miR-141 may be useful as a noninvasive biomarker and as a therapeutic target in several cancers.
Collapse
|
24
|
Schlattner U. The Complex Functions of the NME Family-A Matter of Location and Molecular Activity. Int J Mol Sci 2021; 22:13083. [PMID: 34884887 PMCID: PMC8658066 DOI: 10.3390/ijms222313083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
The family of NME proteins represents a quite complex group of multifunctional enzymes [...].
Collapse
Affiliation(s)
- Uwe Schlattner
- University Grenoble Alpes and Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France;
- Institut Universitaire de France (IUF), 75231 Paris, France
| |
Collapse
|
25
|
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
|