1
|
Cha S, Yang Y, Du W, Jiang T, Wang R, Qu M, Ji Z, Yan C, Yang X, Gong M. Interfacial Lithium Cations Catalyze Biomimetic Aerobic Oxygenation via Short-Range Electrostatic Interaction. Angew Chem Int Ed Engl 2025; 64:e202500546. [PMID: 40178876 DOI: 10.1002/anie.202500546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/05/2025]
Abstract
Enzymes often involve short-range electrostatic interactions in the deliberate microenvironment for accelerating the catalysis. Comparatively, electrostatic interactions from ions in solutions are mostly shielded by solvent or counter-ion shells, creating negligible catalytic effects. Herein, we discovered that the interfacial Li+ cations accumulated on electrodes catalyze the selective water-involved O2 electro-reduction into peroxide anion (OOH-), forming an active side-on Li+-OOH- complex via short-range electrostatic interaction. This complex reduces the O2 reduction energy barrier and increases the nucleophilicity, expediting the aerobic oxygenation of ketones. Aside from trapping active intermediates, Li+ cations also attract the excessive water dipoles to prevent them from quenching the active Li+-OOH- complex. By using probe-assisted quantitative methods, we demonstrated the unique under-coordinative characteristics of interfacial Li+ for interacting with reaction intermediates, and the effective concentration of under-coordinative Li+ on the interface is an order of magnitude higher than in the bulk solution. These analyses provide essential evidences about the intrinsic difference between bulk ions and interfacial ions toward catalysis.
Collapse
Affiliation(s)
- Shuangshuang Cha
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, P.R. China
| | - Yizhou Yang
- National Engineering Laboratory for Industrial Wastewater Treatment, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Wei Du
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, P.R. China
| | - Tao Jiang
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, P.R. China
| | - Ran Wang
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, P.R. China
| | - Mengxin Qu
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, P.R. China
| | - Zhe Ji
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P.R. China
| | - Chang Yan
- Center for Ultrafast Science and Technology, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China
| | - Xuejing Yang
- National Engineering Laboratory for Industrial Wastewater Treatment, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Ming Gong
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, P.R. China
| |
Collapse
|
2
|
Fan X, Gu C, Gao Z, Shen L, Yang X, Song Y, Bian Y, Xu Q, Wang F, Jiang X. Active binding mechanism to superoxide dismutase and toxicological implication for environmentally prevalent phthalates and their hydrolytic products: Coupling in vitro bioassay with molecular dynamics simulation. Int J Biol Macromol 2025; 311:143607. [PMID: 40306524 DOI: 10.1016/j.ijbiomac.2025.143607] [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: 02/13/2025] [Revised: 04/23/2025] [Accepted: 04/26/2025] [Indexed: 05/02/2025]
Abstract
Phthalic acid esters (PAEs) are widely used as plasticizers to improve the flexibility and durability of plastics. However, their environmental presence poses risks by inducing oxidative stress and contributing to metabolic syndrome. Despite being linked to various diseases, the mechanisms by which PAEs disrupt antioxidant enzymes, particularly superoxide dismutase (SOD), are not well understood. This study investigated the molecular interactions between PAEs, their metabolites, and SOD using bioassays and theoretical simulations. The results showed that key metabolites, monophthalates (MAEs) and phthalic acid (PA), strongly inhibited SOD activity, with potency increasing as side chain length decreased. In contrast, PAEs caused minor changes in SOD activity. The inhibition resulted from tight binding of MAEs and PA to the residues in the enzyme's bottom cavity. PAEs and metabolites induced significant structural changes in the secondary structures, catalytic channel, and hydrogen bond network, destabilizing the protein and impairing its function. A strong correlation between SOD inhibition and Gibbs free binding energies at Arg141 was observed. Arg141 and allelic residues can serve as biomarkers for early warnings of oxidative stress. This study improves our understanding of oxidative stress mechanisms caused by PAEs and emphasizes the need for better risk management of phthalate exposure.
Collapse
Affiliation(s)
- Xiuli Fan
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 210008, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Chenggang Gu
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 210008, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Zhengyuan Gao
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 210008, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Lezu Shen
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 210008, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xinglun Yang
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 210008, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yang Song
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 210008, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yongrong Bian
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 210008, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | | | - Fang Wang
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 210008, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xin Jiang
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 210008, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| |
Collapse
|
3
|
Fan X, Gu C, Shen L, Gao Z, Yang X, Bian Y, Wang F, Jiang X. Theoretical insights into the binding of mono/di-ethyl phthalates to superoxide dismutase and associated structural changes impairing antioxidant activity: A coupled molecular docking and dynamics simulation approach. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 983:179667. [PMID: 40394786 DOI: 10.1016/j.scitotenv.2025.179667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/24/2025] [Accepted: 05/11/2025] [Indexed: 05/22/2025]
Abstract
Phthalic acid esters (PAEs) are plasticizers known to increase oxidative stress by impairing antioxidant defenses, including superoxide dismutase (SOD) activity. Since oxidative stress plays a critical role in disease development, the disruption of SOD function by PAEs presents a significant concern. However, the precise molecular mechanisms underlying the regulation of SOD activity remain unclear. This study investigated how diethyl phthalate (DEP) and its major metabolite, monoethyl phthalate (MEP), affected SOD activity using molecular docking and dynamics simulations. The results revealed that both DEP and MEP bound to SOD through weak hydrophilic interactions and hydrogen bonds with residues Lys9, Thr17, Asn51, Thr52, and Arg141 in the bottom of the enzyme's two subunit cavities. These interactions triggered structural changes, particularly in the electrostatic loop and catalytic channels, destabilizing SOD. DEP and MEP increased the enzyme's radius of gyration and solvent-accessible surface area while disrupting intra-protein interactions. MEP showed a stronger inhibitory effect, significantly altering SOD's conformation. This change correlated with reduced catalytic activity (R2 > 0.9). Consequently, the inhibition of the enzyme is primarily due to the disruption of Arg141's conformation and function, which weakens SOD's antioxidant defense and potentially contributes to diseases related to oxidative damage. These results underscore the health risks posed by PAEs, especially following metabolic transformation, and highlight the importance of addressing their oxidative impact.
Collapse
Affiliation(s)
- Xiuli Fan
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 211135, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Chenggang Gu
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 211135, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Lezu Shen
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 211135, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Zhengyuan Gao
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 211135, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xinglun Yang
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 211135, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yongrong Bian
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 211135, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Fang Wang
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 211135, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xin Jiang
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing 211135, PR China; University of the Chinese Academy of Sciences, Beijing 100049, PR China
| |
Collapse
|
4
|
Yapici I, Tokur AG, Sever B, Ciftci H, Basak AN, DeMirci H. Structural Insights into the Dynamics of Water in SOD1 Catalysis and Drug Interactions. Int J Mol Sci 2025; 26:4228. [PMID: 40362464 PMCID: PMC12071975 DOI: 10.3390/ijms26094228] [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: 02/18/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 05/15/2025] Open
Abstract
Superoxide dismutase 1 (SOD1) is a crucial enzyme that protects cells from oxidative damage by converting superoxide radicals into H2O2 and O2. This detoxification process, essential for cellular homeostasis, relies on a precisely orchestrated catalytic mechanism involving the copper cation, while the zinc cation contributes to the structural integrity of the enzyme. This study presents the 2.3 Å crystal structure of human SOD1 (PDB ID: 9IYK), revealing an assembly of six homodimers and twelve distinct active sites. The water molecules form a complex hydrogen-bonding network that drives proton transfer and sustains active site dynamics. Our structure also uncovers subtle conformational changes that highlight the intrinsic flexibility of SOD1, which is essential for its function. Additionally, we observe how these dynamic structural features may be linked to pathological mutations associated with amyotrophic lateral sclerosis (ALS). By advancing our understanding of hSOD1's mechanistic intricacies and the influence of water coordination, this study offers valuable insights for developing therapeutic strategies targeting ALS. Our structure's unique conformations and active site interactions illuminate new facets of hSOD1 function, underscoring the critical role of structural dynamics in enzyme catalysis. Moreover, we conducted a molecular docking analysis using SOD1 for potential radical scavengers and Abelson non-receptor tyrosine kinase (c-Abl, Abl1) inhibitors targeting misfolded SOD1 aggregation along with oxidative stress and apoptosis, respectively. The results showed that CHEMBL1075867, a free radical scavenger derivative, showed the most promising docking results and interactions at the binding site of hSOD1, highlighting its promising role for further studies against SOD1-mediated ALS.
Collapse
Affiliation(s)
- Ilkin Yapici
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Türkiye; (I.Y.); (A.G.T.)
| | - Arda Gorkem Tokur
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Türkiye; (I.Y.); (A.G.T.)
| | - Belgin Sever
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Türkiye;
| | - Halilibrahim Ciftci
- Department of Molecular Biology and Genetics, Burdur Mehmet Akif Ersoy University, Burdur 15030, Türkiye;
| | - Ayse Nazli Basak
- Suna and İnan Kıraç Foundation, Neurodegeneration Research Laboratory (KUTTAM-NDAL), School of Medicine, Koc University, Istanbul 34450, Türkiye
| | - Hasan DeMirci
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Türkiye; (I.Y.); (A.G.T.)
- Stanford PULSE Institute, SLAC National Laboratory, Menlo Park, CA 94025, USA
| |
Collapse
|
5
|
Roy I, Mathur S, Deb S, Rathnam SSV, Tuti N, Shaji UP, L K, Roy A, Maji S. Benzimidazole-based mononuclear polypyridyl Cu(II) complexes: DNA binding, cleavage, and in vitro antiproliferative studies. Dalton Trans 2025; 54:6386-6401. [PMID: 40062962 DOI: 10.1039/d4dt03379e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
This paper addresses the synthesis, characterization, DNA binding, cleavage, and in vitro antiproliferative activity studies of a series of heteroleptic mononuclear copper(II) complexes [Cu(L)(bpy)](ClO4)2, {1}; [Cu(L)(phen)](ClO4)2, {2}; and [Cu(L)(Mephen)](ClO4)2, {3} derived from different polypyridyl ligands, where in the complex architecture, one 2,6-bis(1-methyl-1H-benzo[d]imidazol-2-yl)pyridine(Mebzimpy) (L) moiety is connected to the central Cu metal in a tridentate fashion and the bidentate co-ligands are 2,2'-bipyridine (bpy), 1,10-phenanthroline (phen) and 2,9-dimethyl-1,10-phenanthroline (Mephen). All the synthesized complexes were characterized using various spectroscopic and analytical methods, along with the single-crystal X-ray diffraction (SCXRD) technique. The complexes crystallize in a penta-coordinated distorted square pyramidal geometry. The redox properties of the complexes were also studied by using cyclic voltammetry (CV) and differential pulse voltammetry (DPV). The DNA binding nature of the complexes was investigated utilizing absorbance spectral measurement and fluorescence quenching experiments with ethidium bromide (EB) as a DNA intercalator, employing double-stranded salmon sperm DNA (ss-DNA). Both the binding constant (Kb) and the Stern-Volmer constant (KSV) were found to be in the order of 104. In silico molecular docking analysis confirmed that all the complexes could interact with the minor groove of duplex DNA. The DNA cleaving ability of the complexes was studied by gel electrophoresis using supercoiled plasmid DNA; however, no DNA cleavage was found. DNA-binding polypyridyl complexes are well known to disrupt DNA metabolic pathways and cause cytotoxicity to rapidly growing cancer cells. Hence, cell viability analysis was also carried out with complexes 1-3. It was observed that complexes 2 and 3 prevented the proliferation of the human osteosarcoma cell line U2OS and the triple-negative breast cancer cell line MDA-MB-231. Overall, these findings could be beneficial in the design and development of future antitumor agents.
Collapse
Affiliation(s)
- Indrajit Roy
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India.
| | - Shobhit Mathur
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India.
| | - Sourav Deb
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India.
| | | | - Nikhil Tuti
- Department of Biotechnology, Indian Institute of Technology, Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| | | | - Karthikeyan L
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India.
| | - Anindya Roy
- Department of Biotechnology, Indian Institute of Technology, Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| | - Somnath Maji
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India.
| |
Collapse
|
6
|
Zuercher EC, Poore AT, Prajapat D, Palazzo J, Thomas A, Birthright C, Lawrence J, Chen M, Tian S. Secondary sphere interactions modulate peroxynitrite scavenging by the E2 domain of amyloid precursor protein. Dalton Trans 2025; 54:571-581. [PMID: 39670805 PMCID: PMC11640295 DOI: 10.1039/d4dt02552k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Peroxynitrite (ONOO-) is a highly reactive nitrogen species that can cause significant damage to proteins, lipids, and DNA. Various enzymes, including metalloenzymes, play crucial roles in reducing ONOO- concentrations to protect cellular components. While the interaction of ONOO- with heme proteins is well known, the reduction by Cu-containing proteins is less studied. Amyloid precursor protein (APP), implicated in Alzheimer's disease, has an E2 domain that binds copper ions with a dissociation constant of KD ∼ 10-12 M and is proposed to be involved in iron homeostasis, copper trafficking, and oxidative stress response. Our recent studies using EXAFS, UV-Vis, and EPR spectroscopy revealed a previously unidentified labile water ligand in the Cu(II) site of the E2 domain, suggesting reactivity with anionic substrates like ONOO-. Experimental data showed that Cu(I)-E2 reduces ONOO- at a significant rate (1.1 × 105 M-1 s-1), comparable to native peroxynitrite scavengers, while maintaining active site integrity through multiple redox cycles. This study further investigates the mechanism of ONOO- reduction by Cu(I)-E2 using the Griess assay, demonstrating that reduction occurs via single electron transfer, forming nitrite and nitrate. This process aligns with previous findings that Cu(I)-E2 is oxidized to Cu(II)-E2 upon ONOO- reduction. Mutations at Lys435, affecting secondary sphere interactions, revealed that factors beyond electrostatics are involved in substrate recruitment. MD simulations suggest that steric hindrance from a newly formed hydrogen bond also plays a role. Understanding ONOO- reduction by the E2 domain of APP expands our knowledge of copper proteins in mitigating oxidative stress and elucidates their physiological and pathological roles, particularly in Alzheimer's disease.
Collapse
Affiliation(s)
- Eli C Zuercher
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, USA.
| | - Andrew T Poore
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, USA.
| | - Devendra Prajapat
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, USA.
| | - Joseph Palazzo
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, USA.
| | - Alana Thomas
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, USA.
| | - Caitlin Birthright
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, USA.
| | - Jack Lawrence
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, USA.
| | - Ming Chen
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, USA.
| | - Shiliang Tian
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, USA.
| |
Collapse
|
7
|
McCaig CD. Neurological Diseases can be Regulated by Phase Separation. Rev Physiol Biochem Pharmacol 2025; 187:273-338. [PMID: 39838017 DOI: 10.1007/978-3-031-68827-0_16] [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] [Indexed: 01/23/2025]
Abstract
Several neurological diseases arise from abnormal protein aggregation within neurones and this is closely regulated by phase separation. One such is motor neurone disease and aberrant aggregation of superoxide dismutase. Again these events are regulated by electrical forces that are examined.
Collapse
Affiliation(s)
- Colin D McCaig
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| |
Collapse
|
8
|
Marques HM. Electron transfer in biological systems. J Biol Inorg Chem 2024; 29:641-683. [PMID: 39424709 PMCID: PMC11638306 DOI: 10.1007/s00775-024-02076-8] [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: 04/21/2024] [Accepted: 09/27/2024] [Indexed: 10/21/2024]
Abstract
Examples of how metalloproteins feature in electron transfer processes in biological systems are reviewed. Attention is focused on the electron transport chains of cellular respiration and photosynthesis, and on metalloproteins that directly couple electron transfer to a chemical reaction. Brief mention is also made of extracellular electron transport. While covering highlights of the recent and the current literature, this review is aimed primarily at introducing the senior undergraduate and the novice postgraduate student to this important aspect of bioinorganic chemistry.
Collapse
Affiliation(s)
- Helder M Marques
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, Johannesburg, 2050, South Africa.
| |
Collapse
|
9
|
Guinard P, Hostachy S, Diebold L, Pécaut J, Le Goff A, Duboc C, Delangle P. Outstanding Superoxide Dismutase Catalytic Activity Of Simple Peptide-Based Nickel(II) Complexes. Angew Chem Int Ed Engl 2024; 63:e202409343. [PMID: 39012328 DOI: 10.1002/anie.202409343] [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: 05/17/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/17/2024]
Abstract
We present here the most active synthetic Ni superoxide dismutase (NiSOD) mimic reported to date. Reactive oxygen species are aggressive compounds, which concentrations are tightly regulated in vivo. Among them, the superoxide anion, O2⋅-, is controlled by superoxide dismutases. Capitalizing on the versatility of the Amino-Terminal CuII- and NiII-binding (ATCUN) peptide motif, we introduced positive charges around the NiII center to favor the interaction with the superoxide radical anion. At physiological pH, the pentapeptide H-Cys-His-Cys-Arg-Arg-NH2 coordinates NiII after the deprotonation of one thiol, two amides, and either the second thiol or the N-terminal ammonium, leading to an equilibrium between the two N3S1 and N2S2 coordination modes. Under catalytic conditions, a kcat value of 8.6(4)×106 L.mol-1.s-1 was measured. Within the first second, the catalyst remained undegraded with quantitative consumption of O2⋅- (completed up to 37 catalytic cycles). An extra arginine (Arg) was introduced at the peptide C-terminus to increase the global charge of the NiII complex from +1 to +2. This had no effect on the catalytic performance, highlighting the critical role of charge distribution in space as a determining factor influencing the reactivity.
Collapse
Affiliation(s)
- Pawel Guinard
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SyMMES, 38000, Grenoble, France
- Univ. Grenoble Alpes, CNRS, DCM, 38000, Grenoble, France
| | - Sarah Hostachy
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SyMMES, 38000, Grenoble, France
| | - Léa Diebold
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SyMMES, 38000, Grenoble, France
| | - Jacques Pécaut
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SyMMES, 38000, Grenoble, France
| | - Alan Le Goff
- Univ. Grenoble Alpes, CNRS, DCM, 38000, Grenoble, France
| | - Carole Duboc
- Univ. Grenoble Alpes, CNRS, DCM, 38000, Grenoble, France
| | - Pascale Delangle
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SyMMES, 38000, Grenoble, France
| |
Collapse
|
10
|
Watanabe S, Amporndanai K, Awais R, Latham C, Awais M, O'Neill PM, Yamanaka K, Hasnain SS. Ebselen analogues delay disease onset and its course in fALS by on-target SOD-1 engagement. Sci Rep 2024; 14:12118. [PMID: 38802492 PMCID: PMC11130262 DOI: 10.1038/s41598-024-62903-5] [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: 02/14/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) selectively affects motor neurons. SOD1 is the first causative gene to be identified for ALS and accounts for at least 20% of the familial (fALS) and up to 4% of sporadic (sALS) cases globally with some geographical variability. The destabilisation of the SOD1 dimer is a key driving force in fALS and sALS. Protein aggregation resulting from the destabilised SOD1 is arrested by the clinical drug ebselen and its analogues (MR6-8-2 and MR6-26-2) by redeeming the stability of the SOD1 dimer. The in vitro target engagement of these compounds is demonstrated using the bimolecular fluorescence complementation assay with protein-ligand binding directly visualised by co-crystallography in G93A SOD1. MR6-26-2 offers neuroprotection slowing disease onset of SOD1G93A mice by approximately 15 days. It also protected neuromuscular junction from muscle denervation in SOD1G93A mice clearly indicating functional improvement.
Collapse
Affiliation(s)
- Seiji Watanabe
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8601, Japan
| | - Kangsa Amporndanai
- Molecular Biophysics Group, Department of Biochemistry and System Biology, Institute of System, M0polecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, L69 7ZB, UK
- Department of Biochemistry, School of Medicine, Vanderbilt University, Nashville, TN, 37232, USA
| | - Raheela Awais
- School of Life Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Caroline Latham
- School of Life Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Muhammad Awais
- Department of Molecular and Clinical Cancer Medicine, Institute of System, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Paul M O'Neill
- Department of Chemistry, Faculty of Science and Engineering, University of Liverpool, Liverpool, L69 7ZD, UK.
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8601, Japan.
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, Japan.
- Center for One Medicine Innovative Translational Research (COMIT), Nagoya University, Nagoya, Japan.
| | - S Samar Hasnain
- Molecular Biophysics Group, Department of Biochemistry and System Biology, Institute of System, M0polecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, L69 7ZB, UK.
| |
Collapse
|
11
|
Orrico F, Laurance S, Lopez AC, Lefevre SD, Thomson L, Möller MN, Ostuni MA. Oxidative Stress in Healthy and Pathological Red Blood Cells. Biomolecules 2023; 13:1262. [PMID: 37627327 PMCID: PMC10452114 DOI: 10.3390/biom13081262] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Red cell diseases encompass a group of inherited or acquired erythrocyte disorders that affect the structure, function, or production of red blood cells (RBCs). These disorders can lead to various clinical manifestations, including anemia, hemolysis, inflammation, and impaired oxygen-carrying capacity. Oxidative stress, characterized by an imbalance between the production of reactive oxygen species (ROS) and the antioxidant defense mechanisms, plays a significant role in the pathophysiology of red cell diseases. In this review, we discuss the most relevant oxidant species involved in RBC damage, the enzymatic and low molecular weight antioxidant systems that protect RBCs against oxidative injury, and finally, the role of oxidative stress in different red cell diseases, including sickle cell disease, glucose 6-phosphate dehydrogenase deficiency, and pyruvate kinase deficiency, highlighting the underlying mechanisms leading to pathological RBC phenotypes.
Collapse
Affiliation(s)
- Florencia Orrico
- Laboratorio de Fisicoquímica Biológica, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay; (F.O.); (A.C.L.); (M.N.M.)
- Laboratorio de Enzimología, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay;
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Sandrine Laurance
- Université Paris Cité and Université des Antilles, UMR_S1134, BIGR, Inserm, F-75014 Paris, France; (S.L.); (S.D.L.)
| | - Ana C. Lopez
- Laboratorio de Fisicoquímica Biológica, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay; (F.O.); (A.C.L.); (M.N.M.)
- Laboratorio de Enzimología, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay;
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Sophie D. Lefevre
- Université Paris Cité and Université des Antilles, UMR_S1134, BIGR, Inserm, F-75014 Paris, France; (S.L.); (S.D.L.)
| | - Leonor Thomson
- Laboratorio de Enzimología, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay;
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Matias N. Möller
- Laboratorio de Fisicoquímica Biológica, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay; (F.O.); (A.C.L.); (M.N.M.)
- Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Mariano A. Ostuni
- Université Paris Cité and Université des Antilles, UMR_S1134, BIGR, Inserm, F-75014 Paris, France; (S.L.); (S.D.L.)
| |
Collapse
|
12
|
Wu DD, Jin S, Cheng RX, Cai WJ, Xue WL, Zhang QQ, Yang LJ, Zhu Q, Li MY, Lin G, Wang YZ, Mu XP, Wang Y, Zhang IY, Zhang Q, Chen Y, Cai SY, Tan B, Li Y, Chen YQ, Zhang PJ, Sun C, Yin Y, Wang MJ, Zhu YZ, Tao BB, Zhou JH, Huang WX, Zhu YC. Hydrogen sulfide functions as a micro-modulator bound at the copper active site of Cu/Zn-SOD to regulate the catalytic activity of the enzyme. Cell Rep 2023; 42:112750. [PMID: 37421623 DOI: 10.1016/j.celrep.2023.112750] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 05/16/2023] [Accepted: 06/21/2023] [Indexed: 07/10/2023] Open
Abstract
The present study examines whether there is a mechanism beyond the current concept of post-translational modifications to regulate the function of a protein. A small gas molecule, hydrogen sulfide (H2S), was found to bind at active-site copper of Cu/Zn-SOD using a series of methods including radiolabeled binding assay, X-ray absorption near-edge structure (XANES), and crystallography. Such an H2S binding enhanced the electrostatic forces to guide the negatively charged substrate superoxide radicals to the catalytic copper ion, changed the geometry and energy of the frontier molecular orbitals of the active site, and subsequently facilitated the transfer of an electron from the superoxide radical to the catalytic copper ion and the breakage of the copper-His61 bridge. The physiological relevance of such an H2S effect was also examined in both in vitro and in vivo models where the cardioprotective effects of H2S were dependent on Cu/Zn-SOD.
Collapse
Affiliation(s)
- Dong-Dong Wu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China; School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Sheng Jin
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China; Department of Physiology, Hebei Medical University, 361 Zhongshan Road, Shijiazhuang 050017, China
| | - Ruo-Xiao Cheng
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Wen-Jie Cai
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China; School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Wen-Long Xue
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Qing-Qing Zhang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Le-Jie Yang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Qi Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Meng-Yao Li
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Ge Lin
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Yi-Zhen Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Xue-Pan Mu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Yu Wang
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| | - Igor Ying Zhang
- Department of Chemistry, Fudan University, Shanghai 200438, China
| | - Qi Zhang
- Department of Chemistry, Fudan University, Shanghai 200438, China
| | - Ying Chen
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Sheng-Yang Cai
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Bo Tan
- Clinical Pharmacokinetic Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ye Li
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Yun-Qian Chen
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, Shanghai 200438, China
| | - Pu-Juan Zhang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Chen Sun
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Yue Yin
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai 201210, China
| | - Ming-Jie Wang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Yi-Zhun Zhu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 200433, China; State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Avenida WaiLong, Taipa, Macau 999078, China
| | - Bei-Bei Tao
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Jia-Hai Zhou
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Wei-Xue Huang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China.
| | - Yi-Chun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China.
| |
Collapse
|
13
|
Sim KS, Inoue T. Structure of a superoxide dismutase from a tardigrade: Ramazzottius varieornatus strain YOKOZUNA-1. Acta Crystallogr F Struct Biol Commun 2023; 79:169-179. [PMID: 37358501 PMCID: PMC10327573 DOI: 10.1107/s2053230x2300523x] [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: 04/02/2023] [Accepted: 06/12/2023] [Indexed: 06/27/2023] Open
Abstract
Superoxide dismutase (SOD) is an essential and ubiquitous antioxidant protein that is widely present in biological systems. The anhydrobiotic tardigrades are some of the toughest micro-animals. They have an expanded set of genes for antioxidant proteins such as SODs. These proteins are thought to play an essential role in oxidative stress resistance in critical situations such as desiccation, although their functions at the molecular level have yet to be explored. Here, crystal structures of a copper/zinc-containing SOD (RvSOD15) from an anhydrobiotic tardigrade, Ramazzottius varieornatus strain YOKOZUNA-1, are reported. In RvSOD15, one of the histidine ligands of the catalytic copper center is replaced by a valine (Val87). The crystal structures of the wild type and the V87H mutant show that even though a histidine is placed at position 87, a nearby flexible loop can destabilize the coordination of His87 to the Cu atom. Model structures of other RvSODs were investigated and it was found that some of them are also unusual SODs, with features such as deletion of the electrostatic loop or β3 sheet and unusual metal-binding residues. These studies show that RvSOD15 and some other RvSODs may have evolved to lose the SOD function, suggesting that gene duplications of antioxidant proteins do not solely explain the high stress tolerance of anhydrobiotic tardigrades.
Collapse
Affiliation(s)
- Kee-Shin Sim
- Graduate School of Pharmaceutical Science, Osaka University, Suita City, Osaka 565-0871, Japan
| | - Tsuyoshi Inoue
- Graduate School of Pharmaceutical Science, Osaka University, Suita City, Osaka 565-0871, Japan
| |
Collapse
|
14
|
Vishnu J, Kesavan P, Shankar B, Dembińska K, Swiontek Brzezinska M, Kaczmarek-Szczepańska B. Engineering Antioxidant Surfaces for Titanium-Based Metallic Biomaterials. J Funct Biomater 2023; 14:344. [PMID: 37504839 PMCID: PMC10381466 DOI: 10.3390/jfb14070344] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023] Open
Abstract
Prolonged inflammation induced by orthopedic metallic implants can critically affect the success rates, which can even lead to aseptic loosening and consequent implant failure. In the case of adverse clinical conditions involving osteoporosis, orthopedic trauma and implant corrosion-wear in peri-implant region, the reactive oxygen species (ROS) activity is enhanced which leads to increased oxidative stress. Metallic implant materials (such as titanium and its alloys) can induce increased amount of ROS, thereby critically influencing the healing process. This will consequently affect the bone remodeling process and increase healing time. The current review explores the ROS generation aspects associated with Ti-based metallic biomaterials and the various surface modification strategies developed specifically to improve antioxidant aspects of Ti surfaces. The initial part of this review explores the ROS generation associated with Ti implant materials and the associated ROS metabolism resulting in the formation of superoxide anion, hydroxyl radical and hydrogen peroxide radicals. This is followed by a comprehensive overview of various organic and inorganic coatings/materials for effective antioxidant surfaces and outlook in this research direction. Overall, this review highlights the critical need to consider the aspects of ROS generation as well as oxidative stress while designing an implant material and its effective surface engineering.
Collapse
Affiliation(s)
- Jithin Vishnu
- Department of Mechanical Engineering, Amrita Vishwa Vidyapeetham, Amritapuri Campus, Clappana 690525, India
| | - Praveenkumar Kesavan
- Department of Materials Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Balakrishnan Shankar
- Department of Mechanical Engineering, Amrita Vishwa Vidyapeetham, Amritapuri Campus, Clappana 690525, India
| | - Katarzyna Dembińska
- Department of Environmental Microbiology and Biotechnology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland
| | - Maria Swiontek Brzezinska
- Department of Environmental Microbiology and Biotechnology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland
| | - Beata Kaczmarek-Szczepańska
- Department of Biomaterials and Cosmetic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland
| |
Collapse
|
15
|
Mavadat E, Seyedalipour B, Hosseinkhani S, Colagar AH. Role of charged residues of the "electrostatic loop" of hSOD1 in promotion of aggregation: Implications for the mechanism of ALS-associated mutations under amyloidogenic conditions. Int J Biol Macromol 2023:125289. [PMID: 37307969 DOI: 10.1016/j.ijbiomac.2023.125289] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 09/14/2022] [Accepted: 06/07/2023] [Indexed: 06/14/2023]
Abstract
Protein misfolding and amyloid formation are hallmarks of numerous diseases, including amyotrophic lateral sclerosis (ALS), in which hSOD1 aggregation is involved in pathogenesis. We used two point mutations in the electrostatic loop, G138E and T137R, to analyze charge distribution under destabilizing circumstances to gain more about how ALS-linked mutations affect SOD1 protein stability or net repulsive charge. We show that protein charge is important in the ALS disease process using bioinformatics and experiments. The MD simulation findings demonstrate that the mutant protein differs significantly from WT SOD1, which is consistent with the experimental evidence. The specific activity of the wild type was 1.61 and 1.48 times higher than that of the G138E and T137R mutants, respectively. Under amyloid induction conditions, the intensity of intrinsic and ANS fluorescence in both mutants reduced. Increasing the content of β-sheet structures in mutants can be attributed to aggregation propensity, which was confirmed using CD polarimetry and FTIR spectroscopy. Our findings show that two ALS-related mutations promote the formation of amyloid-like aggregates at near physiological pH under destabilizing conditions, which were detected using spectroscopic probes such as Congo red and ThT fluorescence, and also further confirmation of amyloid-like species by TEM. Overall, our results provide evidence supporting the notion that negative charge changes combined with other destabilizing factors play an important role in increasing protein aggregation by reducing repulsive negative charges.
Collapse
Affiliation(s)
- Elaheh Mavadat
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran
| | - Bagher Seyedalipour
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran.
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
16
|
Sharma KK, Singh D, Mohite SV, Williamson PR, Kennedy JF. Metal manipulators and regulators in human pathogens: A comprehensive review on microbial redox copper metalloenzymes "multicopper oxidases and superoxide dismutases". Int J Biol Macromol 2023; 233:123534. [PMID: 36740121 DOI: 10.1016/j.ijbiomac.2023.123534] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/17/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
The chemistry of metal ions with human pathogens is essential for their survival, energy generation, redox signaling, and niche dominance. To regulate and manipulate the metal ions, various enzymes and metal chelators are present in pathogenic bacteria. Metalloenzymes incorporate transition metal such as iron, zinc, cobalt, and copper in their reaction centers to perform essential metabolic functions; however, iron and copper have gained more importance. Multicopper oxidases have the ability to perform redox reaction on phenolic substrates with the help of copper ions. They have been reported from Enterobacteriaceae, namely Salmonella enterica, Escherichia coli, and Yersinia enterocolitica, but their role in virulence is still poorly understood. Similarly, superoxide dismutases participate in reducing oxidative stress and allow the survival of pathogens. Their role in virulence and survival is well established in Salmonella typhimurium and Mycobacterium tuberculosis. Further, to ensure survival against stress, like metal starvation or metal toxicity, redox metalloenzymes and metal transportation systems of pathogens actively participate in metal homeostasis. Recently, the omics and protein structure biology studies have helped to predict new targets for regulation the colonization potential of the pathogenic strains. The current review is focused on the major roles of redox metalloenzymes, especially MCOs and SODs of human pathogenic bacteria.
Collapse
Affiliation(s)
- Krishna Kant Sharma
- Laboratory of Enzymology and Gut Microbiology, Department of Microbiology, Maharshi Dayanand University, Rohtak 124001, Haryana, India.
| | - Deepti Singh
- Laboratory of Enzymology and Gut Microbiology, Department of Microbiology, Maharshi Dayanand University, Rohtak 124001, Haryana, India
| | - Shreya Vishwas Mohite
- Laboratory of Enzymology and Gut Microbiology, Department of Microbiology, Maharshi Dayanand University, Rohtak 124001, Haryana, India
| | - Peter R Williamson
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John F Kennedy
- Chembiotech Laboratories, Advanced Science and Technology Institute, 5 the Croft, Buntsford Drive, Stoke Heath, Bromsgrove, Worcs B60 4JE, UK
| |
Collapse
|
17
|
Andrés CMC, Pérez de la Lastra JM, Andrés Juan C, Plou FJ, Pérez-Lebeña E. Superoxide Anion Chemistry-Its Role at the Core of the Innate Immunity. Int J Mol Sci 2023; 24:1841. [PMID: 36768162 PMCID: PMC9916283 DOI: 10.3390/ijms24031841] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/09/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Classically, superoxide anion O2•- and reactive oxygen species ROS play a dual role. At the physiological balance level, they are a by-product of O2 reduction, necessary for cell signalling, and at the pathological level they are considered harmful, as they can induce disease and apoptosis, necrosis, ferroptosis, pyroptosis and autophagic cell death. This revision focuses on understanding the main characteristics of the superoxide O2•-, its generation pathways, the biomolecules it oxidizes and how it may contribute to their modification and toxicity. The role of superoxide dismutase, the enzyme responsible for the removal of most of the superoxide produced in living organisms, is studied. At the same time, the toxicity induced by superoxide and derived radicals is beneficial in the oxidative death of microbial pathogens, which are subsequently engulfed by specialized immune cells, such as neutrophils or macrophages, during the activation of innate immunity. Ultimately, this review describes in some depth the chemistry related to O2•- and how it is harnessed by the innate immune system to produce lysis of microbial agents.
Collapse
Affiliation(s)
| | - José Manuel Pérez de la Lastra
- Institute of Natural Products and Agrobiology, CSIC—Spanish Research Council, Avda. Astrofísico Fco. Sánchez, 3, 38206 La Laguna, Spain
| | - Celia Andrés Juan
- Cinquima Institute and Department of Organic Chemistry, Faculty of Sciences, Valladolid University, Paseo de Belén, 7, 47011 Valladolid, Spain
| | - Francisco J. Plou
- Institute of Catalysis and Petrochemistry, CSIC—Spanish Research Council, 28049 Madrid, Spain
| | | |
Collapse
|
18
|
Tellechea E, Asensio AC, Ciaurriz P, Buezo J, López-Gómez P, Urra M, Moran JF. A Study of the Interface of Gold Nanoparticles Conjugated to Cowpea Fe-Superoxide Dismutase. Antioxidants (Basel) 2022; 11:2082. [PMID: 36358454 PMCID: PMC9686739 DOI: 10.3390/antiox11112082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 10/16/2023] Open
Abstract
The iron superoxide dismutase (FeSOD) is a first barrier to defend photosynthetic organisms from superoxide radicals. Although it is broadly present in plants and bacteria, FeSODs are absent in animals. They belong to the same phylogenic family as Mn-containing SODs, which are also highly efficient at detoxifying superoxide radicals. In addition, SODs can react with peroxynitrite, and FeSOD enzyme has already been used to evaluate the anti-nitrative capacity of plant antioxidants. Gold nanoparticles (AuNPs) have been shown to significantly improve the functionality and the efficiency of ligands, providing they are properly assembled. In this work, the characteristics of the recombinant cowpea (Vigna unguiculata) FeSOD (rVuFeSOD) immobilized onto AuNPs were investigated as a function of (1) NP surface chemistry and (2) biofunctionalization methods, either physical adsorption or covalent bonding. The NP surface chemistry was studied by varying the concentration of the ligand molecule 11-mercaptoundecanoic acid (MUA) on the NP surface. The coverage and activity of the protein on AuNPs was determined and correlated to the surface chemistry and the two biofunctionalization methods. rVuFeSOD-AuNPs conjugate stability was monitored through absorption measurements, agarose gel electrophoresis and DLS, enzymatic activity by a colorimetric assay and by in-gel activity assay, and coverage was measured by colorimetric assay. When using physical adsorption, the NP is the most perturbing agent for the activity of the enzyme. In contrast, only the NP coverage was affected by MUA ligand concentration. However, during covalent attachment, both the NP and the concentration of MUA on the surface influenced the enzyme activity, while the coverage of the NP remained constant. The results evidence the importance of the biomolecule and AuNP interaction for the functionality of the hybrid. These strategies can be used to develop electrochemical biosensors for O2•- and for peroxynitrite in biomedical applications.
Collapse
Affiliation(s)
- Edurne Tellechea
- NAITEC-Technological Center of Automotive and Mechatronics, C/Tajonar 20, 31006 Pamplona, Spain
| | - Aaron C. Asensio
- NAITEC-Technological Center of Automotive and Mechatronics, C/Tajonar 20, 31006 Pamplona, Spain
| | - Paula Ciaurriz
- NAITEC-Technological Center of Automotive and Mechatronics, C/Tajonar 20, 31006 Pamplona, Spain
| | - Javier Buezo
- Institute for Multidisciplinary Research in Applied Biology (IMAB), Department of Sciences, Public University of Navarre (UPNA); Avda. de Pamplona 123, 31192 Mutilva, Spain
| | - Pedro López-Gómez
- Institute for Multidisciplinary Research in Applied Biology (IMAB), Department of Sciences, Public University of Navarre (UPNA); Avda. de Pamplona 123, 31192 Mutilva, Spain
| | - Marina Urra
- Institute for Multidisciplinary Research in Applied Biology (IMAB), Department of Sciences, Public University of Navarre (UPNA); Avda. de Pamplona 123, 31192 Mutilva, Spain
| | - Jose F. Moran
- Institute for Multidisciplinary Research in Applied Biology (IMAB), Department of Sciences, Public University of Navarre (UPNA); Avda. de Pamplona 123, 31192 Mutilva, Spain
| |
Collapse
|
19
|
Reprint of: Oxygen Free Radicals and Iron in Relation to Biology and Medicine: Some Problems and Concepts. Arch Biochem Biophys 2022; 726:109246. [PMID: 35680438 DOI: 10.1016/j.abb.2022.109246] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
20
|
Superoxide Radicals in the Execution of Cell Death. Antioxidants (Basel) 2022; 11:antiox11030501. [PMID: 35326151 PMCID: PMC8944419 DOI: 10.3390/antiox11030501] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/01/2022] [Accepted: 03/01/2022] [Indexed: 12/24/2022] Open
Abstract
Superoxide is a primary oxygen radical that is produced when an oxygen molecule receives one electron. Superoxide dismutase (SOD) plays a primary role in the cellular defense against an oxidative insult by ROS. However, the resulting hydrogen peroxide is still reactive and, in the presence of free ferrous iron, may produce hydroxyl radicals and exacerbate diseases. Polyunsaturated fatty acids are the preferred target of hydroxyl radicals. Ferroptosis, a type of necrotic cell death induced by lipid peroxides in the presence of free iron, has attracted considerable interest because of its role in the pathogenesis of many diseases. Radical electrons, namely those released from mitochondrial electron transfer complexes, and those produced by enzymatic reactions, such as lipoxygenases, appear to cause lipid peroxidation. While GPX4 is the most potent anti-ferroptotic enzyme that is known to reduce lipid peroxides to alcohols, other antioxidative enzymes are also indirectly involved in protection against ferroptosis. Moreover, several low molecular weight compounds that include α-tocopherol, ascorbate, and nitric oxide also efficiently neutralize radical electrons, thereby suppressing ferroptosis. The removal of radical electrons in the early stages is of primary importance in protecting against ferroptosis and other diseases that are related to oxidative stress.
Collapse
|
21
|
Wu H, Liu J, Zhang X, Zhang X, Zhang J, Ma E. Four alternative splicing transcripts of intracellular copper/zinc superoxide dismutase 1 in Oxya chinensis. Int J Biol Macromol 2021; 193:1600-1609. [PMID: 34740682 DOI: 10.1016/j.ijbiomac.2021.10.222] [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: 05/16/2021] [Revised: 08/09/2021] [Accepted: 10/29/2021] [Indexed: 11/20/2022]
Abstract
In this study, we obtained four alternative splicing transcripts of intracellular copper/zinc superoxide dismutase 1 (icCuZnSOD1) in Oxya chinensis. OcicCuZnSOD1a has all common characteristics of CuZnSOD family and is a canonical CuZnSOD. OcicCuZnSOD1b is missing a Zn binding site. OcicCuZnSOD1c lacks Zn ion and is a Cu-only SOD. OcicCuZnSOD1d is missing a CuZnSOD conserved sequence and lacks the E-loop, a conserved disulfide bond, and an active site arginine. OcicCuZnSOD1a was the most heat-resistant and OcicCuZnSOD1c was the most unstable at high temperatures above 55 °C. They were stable at a wide pH range, especially in alkaline conditions. The four variants expressed at the throughout developmental stages and had various tissue expression patterns. OcicCuZnSOD1a and OcicCuZnSOD1d were significantly induced by 8.79 mM CuCl2 and OcicCuZnSOD1b was significantly up-regulated by 14.67 mM CuCl2. OcicCuZnSOD1a was significantly inhibited by 19.13 mM ZnSO4 while OcicCuZnSOD1d were significantly induced by 22.61 mM ZnSO4. Disc diffusion assay showed that the four isoforms of OcicCuZnSOD1 made the killing zones smaller surrounding the CdCl2-soaked filter discs. However, the reduction ratios of OcicCuZnSOD1a were the highest. These results implied that the four transcripts played roles in defense against CdCl2-induced oxidative stress while OcicCuZnSOD1a had stronger antioxidant capacity.
Collapse
Affiliation(s)
- Haihua Wu
- Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi 030006, China.
| | - Jing Liu
- Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi 030006, China; College of Life Science, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Xuhan Zhang
- Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi 030006, China; College of Life Science, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Xueyao Zhang
- Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Jianzhen Zhang
- Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Enbo Ma
- Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi 030006, China.
| |
Collapse
|
22
|
Sirisena DMKP, Gayashani Sandamalika WM, Neranjan Tharuka MD, Madusanka RK, Jeong JB, Lee J. A copper-zinc-superoxide dismutase (CuZnSOD) from redlip mullet, Liza haematocheila: Insights to its structural characteristics, immune responses, antioxidant activity, and potent antibacterial properties. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 123:104165. [PMID: 34116115 DOI: 10.1016/j.dci.2021.104165] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/05/2021] [Accepted: 06/05/2021] [Indexed: 06/12/2023]
Abstract
Copper-zinc superoxide dismutase (CuZnSOD) is a nuclear-encoded metalloenzyme responsible for scavenging harmful reactive oxygen species (ROS). In this study, the CuZnSOD homolog from redlip mullet (Liza haematochelia) (MuCuZnSOD) was structurally and functionally characterized to evaluate its antioxidant capacity, antibacterial properties, and protective level in various pathogenic stress conditions. Structural characteristics of MuCuZnSOD were evaluated using different bioinformatics tools. Pairwise sequence comparison and evolutionary tree structure revealed that the MuCuZnSOD sequence was closely related to the CuZnSOD sequence of Oplegnathus fasciatus with a 94.2% sequence identity. Sequence alignment analysis indicated that the CuZnSOD domain was well conserved. The highest transcriptional expression of MuCuZnSOD was identified in the blood. Immune challenge with lipopolysaccharide (LPS), Lactococcus garvieae, and polyinosinic-polycytidylic acid (poly I:C) exhibited an increased MuCuZnSOD mRNA expression in the blood and liver. Transfected green fluorescent protein-fused MuCuZnSOD was localized in the cytoplasm. Recombinant MuCuZnSOD (rMuCuZnSOD) was overexpressed in a bacterial system. The rMuCuZnSOD possessed significant antioxidant properties as determined by conventional xanthine oxidase assay. The optimum pH and temperature of rMuCuZnSOD were found to be pH 9 and 25 °C, respectively. rMuCuZnSOD enzyme activity increased in a concentration-dependent manner. Treatment with potassium cyanide highly inhibited the rMuCuZnSOD activity. rMuCuZnSOD possessed a significant peroxidation activity in the presence of HCO3- ions as demonstrated by the increased viability in cells treated with the enzyme in the presence of hydrogen peroxide. Antibacterial assays showed that rMuCuZnSOD had significant growth-inhibitory effects on both gram-positive and gram-negative bacteria. Collectively, our findings demonstrate that MuCuZnSOD is an essential antioxidant protein, which regulates the host defense mechanisms and innate immunity under oxidative stress.
Collapse
Affiliation(s)
- D M K P Sirisena
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - W M Gayashani Sandamalika
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - M D Neranjan Tharuka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - Rajamanthrilage Kasun Madusanka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - Joon Bum Jeong
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea.
| | - Jehee Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea.
| |
Collapse
|
23
|
Sea KW, Taylor AB, Thomas ST, Liba A, Bergman IB, Holloway SP, Cao X, Gralla EB, Valentine JS, Hart PJ, Galaleldeen A. A pH Switch Controls Zinc Binding in Tomato Copper-Zinc Superoxide Dismutase. Biochemistry 2021; 60:1597-1608. [PMID: 33961402 PMCID: PMC8754426 DOI: 10.1021/acs.biochem.1c00133] [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] [Indexed: 11/29/2022]
Abstract
Copper-zinc superoxide dismutase (SOD1) is a major antioxidant metalloenzyme that protects cells from oxidative damage by superoxide anions (O2-). Structural, biophysical, and other characteristics have in the past been compiled for mammalian SOD1s and for the highly homologous fungal and bovine SOD1s. Here, we characterize the biophysical properties of a plant SOD1 from tomato chloroplasts and present several of its crystal structures. The most unusual of these structures is a structure at low pH in which tSOD1 harbors zinc in the copper-binding site but contains no metal in the zinc-binding site. The side chain of D83, normally a zinc ligand, adopts an alternate rotameric conformation to form an unusual bidentate hydrogen bond with the side chain of D124, precluding metal binding in the zinc-binding site. This alternate conformation of D83 appears to be responsible for the previously observed pH-dependent loss of zinc from the zinc-binding site of SOD1. Titrations of cobalt into apo tSOD1 at a similar pH support the lack of an intact zinc-binding site. Further characterization of tSOD1 reveals that it is a weaker dimer relative to human SOD1 and that it can be activated in vivo through a copper chaperone for the SOD1-independent mechanism.
Collapse
Affiliation(s)
- Kevin W. Sea
- Department of Agriculture and Natural Resources, Santa Rosa Junior College, Santa Rosa, California 95401
| | - Alexander B. Taylor
- Department of Biochemistry and the X-ray Crystallography Core Laboratory, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
| | - Susan T. Thomas
- Department of Biochemistry and the X-ray Crystallography Core Laboratory, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
| | - Amir Liba
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095
| | - Isabelle B. Bergman
- Department of Biological Sciences, St. Mary’s University, San Antonio, TX 78228
| | - Stephen P. Holloway
- Department of Biochemistry and the X-ray Crystallography Core Laboratory, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
| | - Xiaohang Cao
- Department of Biochemistry and the X-ray Crystallography Core Laboratory, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
| | - Edith B. Gralla
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095
| | - Joan S. Valentine
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095
| | - P. John Hart
- Department of Biochemistry and the X-ray Crystallography Core Laboratory, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
- Geriatric Research, Education and Clinical Center, Department of Veterans Affairs, South Texas Veterans Health Care System, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Ahmad Galaleldeen
- Department of Biochemistry and the X-ray Crystallography Core Laboratory, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
- Department of Biological Sciences, St. Mary’s University, San Antonio, TX 78228
| |
Collapse
|
24
|
Trist BG, Hilton JB, Hare DJ, Crouch PJ, Double KL. Superoxide Dismutase 1 in Health and Disease: How a Frontline Antioxidant Becomes Neurotoxic. Angew Chem Int Ed Engl 2021; 60:9215-9246. [PMID: 32144830 PMCID: PMC8247289 DOI: 10.1002/anie.202000451] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Indexed: 12/11/2022]
Abstract
Cu/Zn superoxide dismutase (SOD1) is a frontline antioxidant enzyme catalysing superoxide breakdown and is important for most forms of eukaryotic life. The evolution of aerobic respiration by mitochondria increased cellular production of superoxide, resulting in an increased reliance upon SOD1. Consistent with the importance of SOD1 for cellular health, many human diseases of the central nervous system involve perturbations in SOD1 biology. But far from providing a simple demonstration of how disease arises from SOD1 loss-of-function, attempts to elucidate pathways by which atypical SOD1 biology leads to neurodegeneration have revealed unexpectedly complex molecular characteristics delineating healthy, functional SOD1 protein from that which likely contributes to central nervous system disease. This review summarises current understanding of SOD1 biology from SOD1 genetics through to protein function and stability.
Collapse
Affiliation(s)
- Benjamin G. Trist
- Brain and Mind Centre and Discipline of PharmacologyThe University of Sydney, CamperdownSydneyNew South Wales2050Australia
| | - James B. Hilton
- Department of Pharmacology and TherapeuticsThe University of MelbourneParkvilleVictoria3052Australia
| | - Dominic J. Hare
- Brain and Mind Centre and Discipline of PharmacologyThe University of Sydney, CamperdownSydneyNew South Wales2050Australia
- School of BioSciencesThe University of MelbourneParkvilleVictoria3052Australia
- Atomic Medicine InitiativeThe University of Technology SydneyBroadwayNew South Wales2007Australia
| | - Peter J. Crouch
- Department of Pharmacology and TherapeuticsThe University of MelbourneParkvilleVictoria3052Australia
| | - Kay L. Double
- Brain and Mind Centre and Discipline of PharmacologyThe University of Sydney, CamperdownSydneyNew South Wales2050Australia
| |
Collapse
|
25
|
Azizian H, Forooghian S, Amanlou A, Pérez-Sánchez H, Amanlou M. Phenothiazine as novel human superoxide dismutase modulators: discovery, optimization, and biological evaluation. J Biomol Struct Dyn 2021; 40:7070-7083. [PMID: 33663349 DOI: 10.1080/07391102.2021.1893819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Superoxide dismutases (SODs) are regarded as important antioxidants for protecting cells against damage arising from oxidative stress. Much research is focused on finding new chemicals with an ability to boost human SOD activity. In the research described herein a structure-based approach was used to identify new human Cu-Zn superoxide dismutase (SOD1) modulators based on previously reported plasmodium falciparum iron SOD inhibitors using induced fit docking and molecular dynamic (MD) protocols. The compound with the highest docking binding energy was selected for further structure simplification followed by structural similarity and MD in order to find a new activator/inhibitor scaffold of the SOD1 enzyme. According to the docking survey of the mentioned series, 1,4-bis(3-(1,4,8-trichloro-10Hphenothiazin-10-yl) propyl) piperazine (DS88) was the top scoring compound interacting with the SOD1 active site channel. Following structure simplification and similarity search, the most promising scaffold which is closely related to the phenothiazine antipsychotic class, was identified. Compared with the normal blood SOD1 activity, the percent of O2 production increased with trifluoperazine, while it decreased with the chlorpromazine. The molecular dynamic investigation shows that trifluoperazine exerts its SOD1 activating effect by stabilizing electrostatic loop while chlorpromazine employs SOD1 inhibition activity through repositioning of the electrostatic loop and increasing its distance from the catalytic metal site which diminished substrate specificity and catalytic activity of the SOD1 enzyme. The results identified the preferred region, orientation, and types of interaction for each activator or inhibitor compound.
Collapse
Affiliation(s)
- Homa Azizian
- Department of Medicinal Chemistry, School of Pharmacy, Iran University of Medical Sciences, Tehran Iran
| | - Simin Forooghian
- Faculty of Basic Science, Department of Biology, Tehran Payame Noor University, Tehran, Iran
| | - Arash Amanlou
- Faculty of Specialized Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Horacio Pérez-Sánchez
- Structural Bioinformatics and High-Performance Computing Research Group (BIO-HPC), Computer Engineering Department, Universidad Católica de Murcia (UCAM), Murcia, Spain
| | - Massoud Amanlou
- Faculty of Pharmacy, Department of Medicinal Chemistry, Tehran University of Medical Sciences, Tehran, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Ibrahim MM, El-Kemary MA, Al-Harbi SA, Al-Saidi HM, Sallam SA, Ramadan AEMM. Synthesis and Structural Characterization of Pyridine-based Mn(III), Fe(III), and Co(III) Complexes as SOD Mimics and BSA Binding Studies. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129706] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
27
|
Bonavita R, Laukkanen MO. Common Signal Transduction Molecules Activated by Bacterial Entry into a Host Cell and by Reactive Oxygen Species. Antioxid Redox Signal 2021; 34:486-503. [PMID: 32600071 DOI: 10.1089/ars.2019.7968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Significance: An increasing number of pathogens are acquiring resistance to antibiotics. Efficient antimicrobial drug regimens are important even for the most advanced therapies, which range from cutting-edge invasive clinical protocols, such as robotic surgeries, to the treatment of harmless bacterial diseases and to minor scratches to the skin. Therefore, there is an urgent need to survey alternative antimicrobial drugs that can reinforce or replace existing antibiotics. Recent Advances: Bacterial proteins that are critical for energy metabolism, promising novel anticancer thiourea derivatives, and the use of synthetic molecules that increase the sensitivity of currently used antibiotics are among the recently discovered antimicrobial drugs. Critical Issues: In the development of new drugs, serious consideration should be given to the previous bacterial evolutionary selection caused by antibiotics, by the high proliferation rate of bacteria, and by the simple prokaryotic structure of bacteria. Future Directions: The survey of drug targets has mainly focused on bacterial proteins, although host signaling molecules involved in the treatment of various pathologies may have unknown antimicrobial characteristics. Recent data have suggested that small molecule inhibitors might enhance the effect of antibiotics, for example, by limiting bacterial entry into host cells. Phagocytosis, the mechanism by which host cells internalize pathogens through β-actin cytoskeletal rearrangement, induces calcium signaling, small GTPase activation, and phosphorylation of the phosphatidylinositol 3-kinase-serine/threonine-specific protein kinase B pathway. Antioxid. Redox Signal. 34, 486-503.
Collapse
Affiliation(s)
- Raffaella Bonavita
- Experimental Institute of Endocrinology and Oncology G. Salvatore, IEOS CNR, Naples, Italy
| | | |
Collapse
|
28
|
Flohé L. Looking Back at the Early Stages of Redox Biology. Antioxidants (Basel) 2020; 9:E1254. [PMID: 33317108 PMCID: PMC7763103 DOI: 10.3390/antiox9121254] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/12/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
The beginnings of redox biology are recalled with special emphasis on formation, metabolism and function of reactive oxygen and nitrogen species in mammalian systems. The review covers the early history of heme peroxidases and the metabolism of hydrogen peroxide, the discovery of selenium as integral part of glutathione peroxidases, which expanded the scope of the field to other hydroperoxides including lipid hydroperoxides, the discovery of superoxide dismutases and superoxide radicals in biological systems and their role in host defense, tissue damage, metabolic regulation and signaling, the identification of the endothelial-derived relaxing factor as the nitrogen monoxide radical (more commonly named nitric oxide) and its physiological and pathological implications. The article highlights the perception of hydrogen peroxide and other hydroperoxides as signaling molecules, which marks the beginning of the flourishing fields of redox regulation and redox signaling. Final comments describe the development of the redox language. In the 18th and 19th century, it was highly individualized and hard to translate into modern terminology. In the 20th century, the redox language co-developed with the chemical terminology and became clearer. More recently, the introduction and inflationary use of poorly defined terms has unfortunately impaired the understanding of redox events in biological systems.
Collapse
Affiliation(s)
- Leopold Flohé
- Dipartimento di Medicina Molecolare, Università degli Studi di Padova, v.le G. Colombo 3, 35121 Padova, Italy;
- Departamento de Bioquímica, Universidad de la República, Avda. General Flores 2125, 11800 Montevideo, Uruguay
| |
Collapse
|
29
|
Eleutherio ECA, Silva Magalhães RS, de Araújo Brasil A, Monteiro Neto JR, de Holanda Paranhos L. SOD1, more than just an antioxidant. Arch Biochem Biophys 2020; 697:108701. [PMID: 33259795 DOI: 10.1016/j.abb.2020.108701] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022]
Abstract
During cellular respiration, radicals, such as superoxide, are produced, and in a large concentration, they may cause cell damage. To combat this threat, the cell employs the enzyme Cu/Zn Superoxide Dismutase (SOD1), which converts the radical superoxide into molecular oxygen and hydrogen peroxide, through redox reactions. Although this is its main function, recent studies have shown that the SOD1 has other functions that deviates from its original one including activation of nuclear gene transcription or as an RNA binding protein. This comprehensive review looks at the most important aspects of human SOD1 (hSOD1), including the structure, properties, and characteristics as well as transcriptional and post-translational modifications (PTM) that the enzyme can receive and their effects, and its many functions. We also discuss the strategies currently used to analyze it to better understand its participation in diseases linked to hSOD1 including Amyotrophic Lateral Sclerosis (ALS), cancer, and Parkinson.
Collapse
|
30
|
Trist BG, Hilton JB, Hare DJ, Crouch PJ, Double KL. Superoxide Dismutase 1 in Health and Disease: How a Frontline Antioxidant Becomes Neurotoxic. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202000451] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Benjamin G. Trist
- Brain and Mind Centre and Discipline of Pharmacology The University of Sydney, Camperdown Sydney New South Wales 2050 Australia
| | - James B. Hilton
- Department of Pharmacology and Therapeutics The University of Melbourne Parkville Victoria 3052 Australia
| | - Dominic J. Hare
- Brain and Mind Centre and Discipline of Pharmacology The University of Sydney, Camperdown Sydney New South Wales 2050 Australia
- School of BioSciences The University of Melbourne Parkville Victoria 3052 Australia
- Atomic Medicine Initiative The University of Technology Sydney Broadway New South Wales 2007 Australia
| | - Peter J. Crouch
- Department of Pharmacology and Therapeutics The University of Melbourne Parkville Victoria 3052 Australia
| | - Kay L. Double
- Brain and Mind Centre and Discipline of Pharmacology The University of Sydney, Camperdown Sydney New South Wales 2050 Australia
| |
Collapse
|
31
|
Gastric healing effect of p-coumaric acid isolated from Baccharis dracunculifolia DC on animal model. Naunyn Schmiedebergs Arch Pharmacol 2020; 394:49-57. [PMID: 32780226 DOI: 10.1007/s00210-020-01928-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/18/2020] [Indexed: 02/07/2023]
Abstract
The p-coumaric acid is a phenolic compound present in large quantities in the extract of Baccharis dracunculifolia DC, a Brazilian medicinal plant used to treat gastric ulcer. Given the necessity for finding new chemical components capable of accelerating gastric healing, in this study, the effects of the p-coumaric acid were evaluated in the acetic acid-induced ulcer model in rats, where histological, inflammatory, and oxidative parameters were analyzed. The healing property was also evaluated in the scratch assay on fibroblast cells (L929) and the cytotoxicity of p-coumaric acid was assessed in both L929 and human gastric adenocarcinoma (AGS) cells by MTT assay. The treatment with p-coumaric acid (10 mg/kg, p.o.) for 7 days, twice a day, decreased by 44.6% the acetic acid-induced gastric ulcer compared with the vehicle-treated group. The vehicle control-treated group showed a larger extension of the ulcer base and an extensive damage into the mucosa and submucosa layers, which were mitigated by the treatment with p-coumaric acid. This beneficial effect was also associated with increased levels of mucin and reduced glutathione, decreased amount of lipid hydroperoxides, and increased superoxide dismutase and catalase activities without interfering with the activity of myeloperoxidase in the gastric tissue. The compound promoted the restructuring of the cell monolayer in the scratch test and did not show toxicity in the L929 cell line, while reduced the viability of the AGS, a lineage of human gastric adenocarcinoma. Thus, p-coumaric acid may be considered a natural source for the treatment of gastric ulcers, by reinforcing protective factors of gastric mucosa and by accelerating gastric healing.
Collapse
|
32
|
Malik R, Corrales C, Linsenmeier M, Alalami H, Sepanj N, Bitan G. Examination of SOD1 aggregation modulators and their effect on SOD1 enzymatic activity as a proxy for potential toxicity. FASEB J 2020; 34:11957-11969. [PMID: 32701214 DOI: 10.1096/fj.202000948r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/17/2020] [Accepted: 06/24/2020] [Indexed: 01/08/2023]
Abstract
Small-molecule inhibitors of abnormal protein self-assembly are promising candidates for developing therapy against proteinopathies. Such compounds have been examined primarily as inhibitors of amyloid β-protein (Aβ), whereas testing of inhibitors of other amyloidogenic proteins has lagged behind. An important issue with screening compound libraries is that although an inhibitor suitable for therapy must be both effective and nontoxic, typical screening focuses on efficacy, whereas safety typically is tested at a later stage using cells and/or animals. In addition, typical thioflavin T (ThT)-fluorescence-based screens use the final fluorescence value as a readout, potentially missing important kinetic information. Here, we examined potential inhibitors of superoxide dismutase 1 (SOD1) using ThT-fluorescence including the different phases of fluorescence change and added a parallel screen of SOD1 activity as a potential proxy for compound toxicity. Some compounds previously reported to inhibit other amyloidogenic proteins also inhibited SOD1 aggregation at low micromolar concentrations, whereas others were ineffective. Analysis of the lag phase and exponential slope added important information that could help exclude false-positive or false-negative results. SOD1 was highly resistant to inhibition of its activity, and therefore, did not have the necessary sensitivity to serve as a proxy for examining potential toxicity.
Collapse
Affiliation(s)
- Ravinder Malik
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA, USA
| | - Christian Corrales
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA, USA
| | - Miriam Linsenmeier
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA, USA
| | - Huda Alalami
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA, USA
| | - Niki Sepanj
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA, USA
| | - Gal Bitan
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA, USA.,Brain Research Institute, University of California at Los Angeles, Los Angeles, CA, USA.,Molecular Biology Institute, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
33
|
Zhang AY, Koone JC, Dashnaw CM, Zahler CT, Shaw BF. Complete Charge Regulation by a Redox Enzyme Upon Single Electron Transfer. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202001452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ao Yun Zhang
- Department of Chemistry and Biochemistry Baylor University Waco TX USA
| | - Jordan C. Koone
- Department of Chemistry and Biochemistry Baylor University Waco TX USA
| | - Chad M. Dashnaw
- Department of Chemistry and Biochemistry Baylor University Waco TX USA
| | - Collin T. Zahler
- Department of Chemistry and Biochemistry Baylor University Waco TX USA
| | - Bryan F. Shaw
- Department of Chemistry and Biochemistry Baylor University Waco TX USA
| |
Collapse
|
34
|
Zhang AY, Koone JC, Dashnaw CM, Zahler CT, Shaw BF. Complete Charge Regulation by a Redox Enzyme Upon Single Electron Transfer. Angew Chem Int Ed Engl 2020; 59:10989-10995. [PMID: 32212239 DOI: 10.1002/anie.202001452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/28/2020] [Indexed: 01/22/2023]
Abstract
The degree by which metalloproteins partially regulate net charge (Z) upon electron transfer (ET) was recently measured for the first time using "protein charge ladders" of azurin, cytochrome c, and myoglobin [Angew. Chem. Int. Ed. 2018, 57(19), 5364-5368; Angew. Chem. 2018, 130, 5462-5466]. Here, we show that Cu, Zn superoxide dismutase (SOD1) is unique among proteins in its ability to resist changes in net charge upon single ET (e.g., ΔZET(SOD1) =0.05±0.08 per electron, compared to ΔZET(Cyt-c) =1.19±0.02). This total regulation of net charge by SOD1 is attributed to the protonation of the bridging histidine upon copper reduction, yielding redox centers that are isoelectric at both copper oxidation states. Charge regulation by SOD1 would prevent long range coulombic perturbations to residue pKa 's upon ET at copper, allowing SOD1's "electrostatic loop" to attract superoxide with equal affinity (at both redox states of copper) during diffusion-limited reduction and oxidation of superoxide.
Collapse
Affiliation(s)
- Ao Yun Zhang
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - Jordan C Koone
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - Chad M Dashnaw
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - Collin T Zahler
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - Bryan F Shaw
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| |
Collapse
|
35
|
Chatzidimitriou E, Bisaccia P, Corrà F, Bonato M, Irato P, Manuto L, Toppo S, Bakiu R, Santovito G. Copper/Zinc Superoxide Dismutase from the Crocodile Icefish Chionodraco hamatus: Antioxidant Defense at Constant Sub-Zero Temperature. Antioxidants (Basel) 2020; 9:325. [PMID: 32316382 PMCID: PMC7222407 DOI: 10.3390/antiox9040325] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
In the present study, we describe the purification and molecular characterization of Cu,Zn superoxide dismutase (SOD) from Chionodraco hamatus, an Antarctic teleost widely distributed in many areas of the Ross Sea that plays a pivotal role in the Antarctic food chain. The primary sequence was obtained using biochemical and molecular biology approaches and compared with Cu,Zn SODs from other organisms. Multiple sequence alignment using the amino acid sequence revealed that Cu,Zn SOD showed considerable sequence similarity with its orthologues from various vertebrate species, but also some specific substitutions directly linked to cold adaptation. Phylogenetic analyses presented the monophyletic status of Antartic Teleostei among the Perciformes, confirming the erratic differentiation of these proteins and concurring with the theory of the "unclock-like" behavior of Cu,Zn SOD evolution. Expression of C. hamatus Cu,Zn SOD at both the mRNA and protein levels were analyzed in various tissues, highlighting the regulation of gene expression related to environmental stress conditions and also animal physiology. The data presented are the first on the antioxidant enzymes of a fish belonging to the Channichthyidae family and represent an important starting point in understanding the antioxidant systems of these organisms that are subject to constant risk of oxidative stress.
Collapse
Affiliation(s)
- Evangelia Chatzidimitriou
- Institute of Natural Resource Sciences, ZHAW Zurich University of Applied Sciences, 8820 Wädenswil, Switzerland;
| | - Paola Bisaccia
- Department of Biology, University of Padova, 35131 Padova, Italy; (P.B.); (F.C.); (M.B.); (P.I.)
| | - Francesca Corrà
- Department of Biology, University of Padova, 35131 Padova, Italy; (P.B.); (F.C.); (M.B.); (P.I.)
| | - Marco Bonato
- Department of Biology, University of Padova, 35131 Padova, Italy; (P.B.); (F.C.); (M.B.); (P.I.)
| | - Paola Irato
- Department of Biology, University of Padova, 35131 Padova, Italy; (P.B.); (F.C.); (M.B.); (P.I.)
| | - Laura Manuto
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy; (L.M.); (S.T.)
| | - Stefano Toppo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy; (L.M.); (S.T.)
- CRIBI Biotech Centre, University of Padova, 35131 Padova, Italy
| | - Rigers Bakiu
- Department of Aquaculture and Fisheries, Agricultural University of Tirana, 1000 Tiranë, Albania;
| | - Gianfranco Santovito
- Department of Biology, University of Padova, 35131 Padova, Italy; (P.B.); (F.C.); (M.B.); (P.I.)
| |
Collapse
|
36
|
Abstract
Few proteins have come under such intense scrutiny as superoxide dismutase-1 (SOD1). For almost a century, scientists have dissected its form, function and then later its malfunction in the neurodegenerative disease amyotrophic lateral sclerosis (ALS). We now know SOD1 is a zinc and copper metalloenzyme that clears superoxide as part of our antioxidant defence and respiratory regulation systems. The possibility of reduced structural integrity was suggested by the first crystal structures of human SOD1 even before deleterious mutations in the sod1 gene were linked to the ALS. This concept evolved in the intervening years as an impressive array of biophysical studies examined the characteristics of mutant SOD1 in great detail. We now recognise how ALS-related mutations perturb the SOD1 maturation processes, reduce its ability to fold and reduce its thermal stability and half-life. Mutant SOD1 is therefore predisposed to monomerisation, non-canonical self-interactions, the formation of small misfolded oligomers and ultimately accumulation in the tell-tale insoluble inclusions found within the neurons of ALS patients. We have also seen that several post-translational modifications could push wild-type SOD1 down this toxic pathway. Recently we have come to view ALS as a prion-like disease where both the symptoms, and indeed SOD1 misfolding itself, are transmitted to neighbouring cells. This raises the possibility of intervention after the initial disease presentation. Several small-molecule and biologic-based strategies have been devised which directly target the SOD1 molecule to change the behaviour thought to be responsible for ALS. Here we provide a comprehensive review of the many biophysical advances that sculpted our view of SOD1 biology and the recent work that aims to apply this knowledge for therapeutic outcomes in ALS.
Collapse
|
37
|
della Valle E, Marracino P, Pakhomova O, Liberti M, Apollonio F. Nanosecond pulsed electric signals can affect electrostatic environment of proteins below the threshold of conformational effects: The case study of SOD1 with a molecular simulation study. PLoS One 2019; 14:e0221685. [PMID: 31454403 PMCID: PMC6711501 DOI: 10.1371/journal.pone.0221685] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/13/2019] [Indexed: 12/19/2022] Open
Abstract
Electric fields can be a powerful tool to interact with enzymes or proteins, with an intriguing perspective to allow protein manipulation. Recently, researchers have focused the interest on intracellular enzyme modifications triggered by the application of nanosecond pulsed electric fields. These findings were also supported by theoretical predictions from molecular dynamics simulations focussing on significant variations in protein secondary structures. In this work, a theoretical study utilizing molecular dynamics simulations is proposed to explore effects of electric fields of high intensity and very short nanosecond duration applied to the superoxide dismutase (Cu/Zn-SOD or SOD-1), an important enzyme involved in the cellular antioxidant defence mechanism. The effects of 100-nanosecond pulsed electric fields, with intensities ranging from 108 to 7x108 V/m, on a single SOD1 enzyme are presented. We demonstrated that the lowest intensity of 108 V/m, although not inducing structural changes, can produce electrostatic modifications on the reaction centre of the enzyme, as apparent from the dipolar response and the electric field distribution of the protein active site. Electric pulses above 5x108 V/m produced a fast transition between the folded and a partially denatured state, as inferred by the secondary structures analysis. Finally, for the highest field intensity used (7x108 V/m), a not reversible transition toward an unfolded state was observed.
Collapse
Affiliation(s)
- Elena della Valle
- BioElectronic Vision Lab, University of Michigan, Ann Arbor, Michigan, United States of America
| | | | - Olga Pakhomova
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia, United States of America
| | - Micaela Liberti
- Department of Information Engineering, Electronics and Telecommunications, Sapienza University of Rome, Rome, Italy
| | - Francesca Apollonio
- Department of Information Engineering, Electronics and Telecommunications, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
38
|
Banks CJ, Andersen JL. Mechanisms of SOD1 regulation by post-translational modifications. Redox Biol 2019; 26:101270. [PMID: 31344643 PMCID: PMC6658992 DOI: 10.1016/j.redox.2019.101270] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 12/18/2022] Open
Abstract
SOD1 is commonly known for its ROS scavenging activity, but recent work has uncovered additional roles in modulating metabolism, maintaining redox balance, and regulating transcription. This new paradigm of expanded SOD1 function raises questions regarding the regulation of SOD1 and the cellular partitioning of its biological roles. Despite decades of research on SOD1, much of which focuses on its pathogenic role in amyotrophic lateral sclerosis, relatively little is known about its regulation by post-translational modifications (PTMs). However, over the last decade, advancements in mass spectrometry have led to a boom in PTM discovery across the proteome, which has also revealed new mechanisms of SOD1 regulation by PTMs and an array of SOD1 PTMs with high likelihood of biological function. In this review, we address emerging mechanisms of SOD1 regulation by post-translational modifications, many of which begin to shed light on how the various functions of SOD1 are regulated within the cell.
Collapse
Affiliation(s)
- C J Banks
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - J L Andersen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA.
| |
Collapse
|
39
|
Tompa DR, Kadhirvel S. Far positioned ALS associated mutants of Cu/Zn SOD forms partially metallated, destabilized misfolding intermediates. Biochem Biophys Res Commun 2019; 516:494-499. [PMID: 31230748 DOI: 10.1016/j.bbrc.2019.06.086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 06/16/2019] [Indexed: 11/18/2022]
Abstract
Loss of stability of proteins is associated with their misfolding and aggregation which results in disease. Despite of the higher stability of Cu/Zn superoxide dismutase (SOD1), the point mutations destabilize its structure, results in oligomerization and the aggregation of SOD1 which is closely associated with the motor neuron disorder, amyotrophic lateral sclerosis. In the present study, we analyzed the role of two SOD1 mutants V14G and E100G which are located far away from the metal sites, dimer interface and disulfide region. The SOD1 mutants were recombinantly produced and their activity, structure and stability were investigated using biochemical methods, CD and DSC methods. In comparison with wild-type SOD1, the mutants exhibited reduced activity and the CD data showed comparable secondary structures composition. However, the stability studies using chemical and thermal denaturation methods showed the mutants are destabilized. Interestingly, our DSC data strongly suggested the destabilization of the mutants is due to the partial metalation of Cu/Zn ions. This observation emphasizes that although the mutations V14G and E100G are located away from the metal sites, they could affect the metal binding similar to metal binding region mutants, which are more susceptible to misfold and aggregate.
Collapse
Affiliation(s)
- Dharma Rao Tompa
- Biomolecular Crystallography Laboratory, Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, 613401, Tamil Nadu, India
| | - Saraboji Kadhirvel
- Biomolecular Crystallography Laboratory, Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, 613401, Tamil Nadu, India.
| |
Collapse
|
40
|
Wang W, Wu S, Wang J, Li Z, Cui H, Lin S, Zhu J, Chen Q. Superoxide dismutase transcellular shuttle constructed from dendritic MOF and charge reversible protein derivatives. Chem Sci 2019; 10:4476-4485. [PMID: 31057775 PMCID: PMC6482591 DOI: 10.1039/c8sc04160a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 03/07/2019] [Indexed: 11/21/2022] Open
Abstract
The development of molecular biology has led to the identification of protein-based therapeutics as an intriguing approach for the treatment of a wide range of diseases. To manufacture transcellular protein delivery shuttles, we attempted charge reversal chemistry on native proteins [e.g., superoxide dismutase (SOD): an enzyme capable of scavenging detrimental reactive oxygen species] by the selective conversion of the positively charged amino residues of native SOD to conjugated negatively charged citraconic moieties, eliciting overall negatively charged polyelectrolytes for the subsequent electrostatic self-assembly with cationic metal-organic framework (MOF) derivatives into protein delivery systems. Please note that the charge conversion was reversible, restoring the original amino groups in intracellular acidic endosome compartments (pH 5), which afforded facile charge reversible functions to reclaim the active SOD in the cell interior. In particular, the strategic manufacture of dendritic MOF supramolecular architectures as transcellular shuttles for the aforementioned charge-reversible SOD derivatives is noteworthy. The MOF was surface-functionalized with several polycationic segments, thus contributing to the hyper-charged architecture for the easy accommodation of the negatively charged SOD derivatives. Consequently, the SOD derivatives managed to internalize into cells by hitchhiking via endocytosis of the positively charged MOF. Once they resided in the acidic endosomes, the charge reversal of the SOD derivatives could occur smoothly and result in reduced interactions between the charged-reversed SOD and MOF, leading to the release of active SOD. Simultaneously, the dendritic MOF due to protein release presented a highly positive-charged architecture to provoke endosome membrane disruption, consequently spurring the translocation of SOD to the cytosol for the execution of its enzymatic activities. Herein, the intracellular ROS level of the activated macrophages was validated to be markedly suppressed by our proposed transcellular SOD shuttles, thereby indicating their wide availability to diverse functional proteins for biomedical applications.
Collapse
Affiliation(s)
- Wei Wang
- State Key Laboratory of Fine Chemicals , Dalian University of Technology , No. 2 Linggong Road , Dalian 116024 , China .
- School of Life Science and Biotechnology , Dalian University of Technology , No. 2 Linggong Road , Dalian 116024 , China
| | - Sudong Wu
- Academy for Advanced Interdisciplinary Studies , Southern University of Science and Technology , Shenzhen 518055 , China
- Department of Materials Science and Engineering , Southern University of Science and Technology , Shenzhen 518055 , China
| | - Jingyun Wang
- State Key Laboratory of Fine Chemicals , Dalian University of Technology , No. 2 Linggong Road , Dalian 116024 , China .
- School of Life Science and Biotechnology , Dalian University of Technology , No. 2 Linggong Road , Dalian 116024 , China
| | - Zhen Li
- College of Pharmacy , Dalian Medical University , No. 9 West Section Lvshun South Road , Dalian 116044 , China
| | - Hongyan Cui
- School of Life Science and Biotechnology , Dalian University of Technology , No. 2 Linggong Road , Dalian 116024 , China
| | - Shuseng Lin
- School of Life Science and Biotechnology , Dalian University of Technology , No. 2 Linggong Road , Dalian 116024 , China
| | - Jingyi Zhu
- School of Life Science and Biotechnology , Dalian University of Technology , No. 2 Linggong Road , Dalian 116024 , China
| | - Qixian Chen
- State Key Laboratory of Fine Chemicals , Dalian University of Technology , No. 2 Linggong Road , Dalian 116024 , China .
- School of Life Science and Biotechnology , Dalian University of Technology , No. 2 Linggong Road , Dalian 116024 , China
| |
Collapse
|
41
|
Zahler CT, Shaw BF. What Are We Missing by Not Measuring the Net Charge of Proteins? Chemistry 2019; 25:7581-7590. [PMID: 30779227 DOI: 10.1002/chem.201900178] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Indexed: 12/21/2022]
Abstract
The net electrostatic charge (Z) of a folded protein in solution represents a bird's eye view of its surface potentials-including contributions from tightly bound metal, solvent, buffer, and cosolvent ions-and remains one of its most enigmatic properties. Few tools are available to the average biochemist to rapidly and accurately measure Z at pH≠pI. Tools that have been developed more recently seem to go unnoticed. Most scientists are content with this void and estimate the net charge of a protein from its amino acid sequence, using textbook values of pKa . Thus, Z remains unmeasured for nearly all folded proteins at pH≠pI. When marveling at all that has been learned from accurately measuring the other fundamental property of a protein-its mass-one wonders: what are we missing by not measuring the net charge of folded, solvated proteins? A few big questions immediately emerge in bioinorganic chemistry. When a single electron is transferred to a metalloprotein, does the net charge of the protein change by approximately one elementary unit of charge or does charge regulation dominate, that is, do the pKa values of most ionizable residues (or just a few residues) adjust in response to (or in concert with) electron transfer? Would the free energy of charge regulation (ΔΔGz ) account for most of the outer sphere reorganization energy associated with electron transfer? Or would ΔΔGz contribute more to the redox potential? And what about metal binding itself? When an apo-metalloprotein, bearing minimal net negative charge (e.g., Z=-2.0) binds one or more metal cations, is the net charge abolished or inverted to positive? Or do metalloproteins regulate net charge when coordinating metal ions? The author's group has recently dusted off a relatively obscure tool-the "protein charge ladder"-and used it to begin to answer these basic questions.
Collapse
Affiliation(s)
- Collin T Zahler
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, 76706, USA
| | - Bryan F Shaw
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, 76706, USA
| |
Collapse
|
42
|
Electronic and Functional Structure of Copper in Plant Cu/Zn Superoxide Dismutase with Combined Site-directed Mutagenesis and Electron Paramagnetic Resonance. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2019. [DOI: 10.1016/s1872-2040(19)61143-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
43
|
Oliveira AP, Souza LKM, Araújo TSL, Araújo SD, Nogueira KM, Sousa FBM, Silva RO, Pacífico DM, Martins CS, Brito GADC, Souza MHLP, Medeiros JVR. Lactobacillus reuteri DSM 17938 Protects against Gastric Damage Induced by Ethanol Administration in Mice: Role of TRPV1/Substance P Axis. Nutrients 2019; 11:nu11010208. [PMID: 30669695 PMCID: PMC6356937 DOI: 10.3390/nu11010208] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/05/2018] [Accepted: 11/20/2018] [Indexed: 12/13/2022] Open
Abstract
This study aimed to evaluate the effect of Lactobacillus reuteri DSM 17938 (DSM) on ethanol-induced gastric injury, and if its possible mechanism of action is related to inhibiting the transient receptor potential vanilloid type 1 (TRPV1). We evaluated the effect of supplementing 10⁸ CFU•g body wt-1•day-1 of DSM on ethanol-induced gastric injury. DSM significantly reduced the ulcer area (1.940 ± 1.121 mm²) with 3 days of pretreatment. The effects of DSM supplementation were reversed by Resiniferatoxin (RTX), TRPV1 agonist (3 nmol/kg p.o.). Substance P (SP) (1 μmol/L per 20 g) plus 50% ethanol resulted in hemorrhagic lesions, and DSM supplementation did not reverse the lesion area induced by administering SP. TRPV1 staining intensity was lower, SP, malondialdehyde (MDA) and nitrite levels were reduced, and restored normal levels of antioxidant parameters (glutathione and superoxide dismutase) in the gastric mucosa in mice treated with DSM. In conclusion, DSM exhibited gastroprotective activity through decreased expression of TRPV1 receptor and decreasing SP levels, with a consequent reduction of oxidative stress.
Collapse
Affiliation(s)
- Ana P Oliveira
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Federal University of Piauí, Av. São Sebastião, nº 2819, CEP 64202-02, Parnaíba, PI, Brazil.
| | - Luan K M Souza
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Federal University of Piauí, Av. São Sebastião, nº 2819, CEP 64202-02, Parnaíba, PI, Brazil.
| | - Thiago S L Araújo
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Federal University of Piauí, Av. São Sebastião, nº 2819, CEP 64202-02, Parnaíba, PI, Brazil.
| | - Simone de Araújo
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Federal University of Piauí, Av. São Sebastião, nº 2819, CEP 64202-02, Parnaíba, PI, Brazil.
| | - Kerolayne M Nogueira
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Federal University of Piauí, Av. São Sebastião, nº 2819, CEP 64202-02, Parnaíba, PI, Brazil.
| | - Francisca Beatriz M Sousa
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Federal University of Piauí, Av. São Sebastião, nº 2819, CEP 64202-02, Parnaíba, PI, Brazil.
| | - Renan O Silva
- Department of Physiology and Pharmacology, Federal University of Ceará, CEP 60430-270, Fortaleza, Ceará, Brazil.
| | - Dvison M Pacífico
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, Faculty of Medicine, Federal University Ceará, CEP 60430-170, Fortaleza-CE, Brazil.
| | - Conceição S Martins
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, Faculty of Medicine, Federal University Ceará, CEP 60430-170, Fortaleza-CE, Brazil.
| | - Gerly Anne de C Brito
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, Faculty of Medicine, Federal University Ceará, CEP 60430-170, Fortaleza-CE, Brazil.
| | - Marcellus H L P Souza
- Department of Physiology and Pharmacology, Federal University of Ceará, CEP 60430-270, Fortaleza, Ceará, Brazil.
| | - Jand Venes R Medeiros
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (Lafidg), Federal University of Piauí, Av. São Sebastião, nº 2819, CEP 64202-02, Parnaíba, PI, Brazil.
| |
Collapse
|
44
|
Parascandolo A, Laukkanen MO. Carcinogenesis and Reactive Oxygen Species Signaling: Interaction of the NADPH Oxidase NOX1-5 and Superoxide Dismutase 1-3 Signal Transduction Pathways. Antioxid Redox Signal 2019; 30:443-486. [PMID: 29478325 PMCID: PMC6393772 DOI: 10.1089/ars.2017.7268] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Reduction/oxidation (redox) balance could be defined as an even distribution of reduction and oxidation complementary processes and their reaction end products. There is a consensus that aberrant levels of reactive oxygen species (ROS), commonly observed in cancer, stimulate primary cell immortalization and progression of carcinogenesis. However, the mechanism how different ROS regulate redox balance is not completely understood. Recent Advances: In the current review, we have summarized the main signaling cascades inducing NADPH oxidase NOX1-5 and superoxide dismutase (SOD) 1-3 expression and their connection to cell proliferation, immortalization, transformation, and CD34+ cell differentiation in thyroid, colon, lung, breast, and hematological cancers. CRITICAL ISSUES Interestingly, many of the signaling pathways activating redox enzymes or mediating the effect of ROS are common, such as pathways initiated from G protein-coupled receptors and tyrosine kinase receptors involving protein kinase A, phospholipase C, calcium, and small GTPase signaling molecules. FUTURE DIRECTIONS The clarification of interaction of signal transduction pathways could explain how cells regulate redox balance and may even provide means to inhibit the accumulation of harmful levels of ROS in human pathologies.
Collapse
|
45
|
Robinett NG, Culbertson EM, Peterson RL, Sanchez H, Andes DR, Nett JE, Culotta VC. Exploiting the vulnerable active site of a copper-only superoxide dismutase to disrupt fungal pathogenesis. J Biol Chem 2018; 294:2700-2713. [PMID: 30593499 DOI: 10.1074/jbc.ra118.007095] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 12/21/2018] [Indexed: 01/08/2023] Open
Abstract
Copper-only superoxide dismutases (SODs) represent a new class of SOD enzymes that are exclusively extracellular and unique to fungi and oomycetes. These SODs are essential for virulence of fungal pathogens in pulmonary and disseminated infections, and we show here an additional role for copper-only SODs in promoting survival of fungal biofilms. The opportunistic fungal pathogen Candida albicans expresses three copper-only SODs, and deletion of one of them, SOD5, eradicated candidal biofilms on venous catheters in a rodent model. Fungal copper-only SODs harbor an irregular active site that, unlike their Cu,Zn-SOD counterparts, contains a copper co-factor unusually open to solvent and lacks zinc for stabilizing copper binding, making fungal copper-only SODs highly vulnerable to metal chelators. We found that unlike mammalian Cu,Zn-SOD1, C. albicans SOD5 indeed rapidly loses its copper to metal chelators such as EDTA, and binding constants for Cu(II) predict that copper-only SOD5 has a much lower affinity for copper than does Cu,Zn-SOD1. We screened compounds with a variety of indications and identified several metal-binding compounds, including the ionophore pyrithione zinc (PZ), that effectively inhibit C. albicans SOD5 but not mammalian Cu,Zn-SOD1. We observed that PZ both acts as an ionophore that promotes uptake of toxic metals and inhibits copper-only SODs. The pros and cons of a vulnerable active site for copper-only SODs and the possible exploitation of this vulnerability in antifungal drug design are discussed.
Collapse
Affiliation(s)
- Natalie G Robinett
- From the Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205 and
| | - Edward M Culbertson
- From the Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205 and
| | - Ryan L Peterson
- From the Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205 and
| | - Hiram Sanchez
- the Departments of Medicine and Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53726
| | - David R Andes
- the Departments of Medicine and Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53726
| | - Jeniel E Nett
- the Departments of Medicine and Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53726
| | - Valeria C Culotta
- From the Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205 and
| |
Collapse
|
46
|
Tang HYH, Shin DS, Hura GL, Yang Y, Hu X, Lightstone FC, McGee MD, Padgett HS, Yannone SM, Tainer JA. Structural Control of Nonnative Ligand Binding in Engineered Mutants of Phosphoenolpyruvate Carboxykinase. Biochemistry 2018; 57:6688-6700. [PMID: 30376300 PMCID: PMC6642699 DOI: 10.1021/acs.biochem.8b00963] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Protein engineering to alter recognition underlying ligand binding and activity has enormous potential. Here, ligand binding for Escherichia coli phosphoenolpyruvate carboxykinase (PEPCK), which converts oxaloacetate into CO2 and phosphoenolpyruvate as the first committed step in gluconeogenesis, was engineered to accommodate alternative ligands as an exemplary system with structural information. From our identification of bicarbonate binding in the PEPCK active site at the supposed CO2 binding site, we probed binding of nonnative ligands with three oxygen atoms arranged to resemble the bicarbonate geometry. Crystal structures of PEPCK and point mutants with bound nonnative ligands thiosulfate and methanesulfonate along with strained ATP and reoriented oxaloacetate intermediates and unexpected bicarbonate were determined and analyzed. The mutations successfully altered the bound ligand position and orientation and its specificity: mutated PEPCKs bound either thiosulfate or methanesulfonate but never both. Computational calculations predicted a methanesulfonate binding mutant and revealed that release of the active site ordered solvent exerts a strong influence on ligand binding. Besides nonnative ligand binding, one mutant altered the Mn2+ coordination sphere: instead of the canonical octahedral ligand arrangement, the mutant in question had an only five-coordinate arrangement. From this work, critical features of ligand binding, position, and metal ion cofactor geometry required for all downstream events can be engineered with small numbers of mutations to provide insights into fundamental underpinnings of protein-ligand recognition. Through structural and computational knowledge, the combination of designed and random mutations aids in the robust design of predetermined changes to ligand binding and activity to engineer protein function.
Collapse
Affiliation(s)
- Henry Y. H. Tang
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
- Department of Chemistry, University of California, Berkeley, California 94720, United States
| | - David S. Shin
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Greg L. Hura
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
- Department of Biochemistry and Chemistry, University of California, Santa Cruz, California 95064, United States
| | - Yue Yang
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California 94550, United States
| | - Xiaoyu Hu
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Felice C. Lightstone
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California 94550, United States
| | | | - Hal S. Padgett
- Novici Biotech LLC, Vacaville, California 95688, United States
| | - Steven M. Yannone
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - John A. Tainer
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, United States
| |
Collapse
|
47
|
Olson AJ. Perspectives on Structural Molecular Biology Visualization: From Past to Present. J Mol Biol 2018; 430:3997-4012. [PMID: 30009769 PMCID: PMC6186497 DOI: 10.1016/j.jmb.2018.07.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/03/2018] [Accepted: 07/06/2018] [Indexed: 12/15/2022]
Abstract
Visualization has been a key technology in the progress of structural molecular biology for as long as the field has existed. This perspective describes the nature of the visualization process in structural studies, how it has evolved over the years, and its relationship to the changes in technology that have supported and driven it. It focuses on how technical advances have changed the way we look at and interact with molecular structure, and how structural biology has fostered and challenged that technology.
Collapse
Affiliation(s)
- Arthur J Olson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
48
|
Wexler AG, Schofield WB, Degnan PH, Folta-Stogniew E, Barry NA, Goodman AL. Human gut Bacteroides capture vitamin B 12 via cell surface-exposed lipoproteins. eLife 2018; 7:37138. [PMID: 30226189 PMCID: PMC6143338 DOI: 10.7554/elife.37138] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/26/2018] [Indexed: 01/02/2023] Open
Abstract
Human gut Bacteroides use surface-exposed lipoproteins to bind and metabolize complex polysaccharides. Although vitamins and other nutrients are also essential for commensal fitness, much less is known about how commensal bacteria compete with each other or the host for these critical resources. Unlike in Escherichia coli, transport loci for vitamin B12 (cobalamin) and other corrinoids in human gut Bacteroides are replete with conserved genes encoding proteins whose functions are unknown. Here we report that one of these proteins, BtuG, is a surface-exposed lipoprotein that is essential for efficient B12 transport in B. thetaiotaomicron. BtuG binds B12 with femtomolar affinity and can remove B12 from intrinsic factor, a critical B12 transport protein in humans. Our studies suggest that Bacteroides use surface-exposed lipoproteins not only for capturing polysaccharides, but also to acquire key vitamins in the gut. Eating is the first step in an hours-long process that extracts the nutrients we need to live. It not only nourishes us, but also a vast community of bacteria in our gut called the microbiota. The gut microbiota acts like an extension of our immune system and helps us stay healthy in many ways. For example, it blocks pathogens from making us sick. But too many gut bacteria in the wrong parts of our intestines can be harmful. Some people are prone to developing a dangerous overgrowth of bacteria in their small intestine where most of our dietary nutrients get absorbed. This overgrowth can lead to many problems including vitamin B12 deficiency even when they eat plenty of it. To understand why, scientists must learn how microbes affect our ability to absorb nutrients from food and how the microbes themselves capture nutrients like vitamin B12 as they pass through our digestive tract. Now, Wexler et al. show that some gut microbes may be able to pirate vitamin B12 from us as it passes through the digestive tract. Wexler et al. showed that a protein called BtuG on the surface of a type of gut bacteria called Bacteriodes grabs onto vitamin B12 with extraordinary strength. In fact, these bacterial proteins bind to vitamin B12 so strongly that they can even pry it away from our own vitamin B12 collecting protein. When Bacteriodes with and without BtuG were placed in mice with no gut bacteria of their own, bacteria with BtuG rapidly outcompeted those lacking the protein. The experiments suggest that competition for vitamin B12 among microbes has favored bacteria that are better at capturing the nutrient. More studies are needed to learn whether BtuG contributes to vitamin B12 deficiencies in humans with gut bacteria overgrowth and determine the best ways to combat such deficiencies.
Collapse
Affiliation(s)
- Aaron G Wexler
- Department of Microbial Pathogenesis, Yale University, New Haven, United States.,Microbial Sciences Institute, Yale University, New Haven, United States
| | - Whitman B Schofield
- Department of Microbial Pathogenesis, Yale University, New Haven, United States.,Microbial Sciences Institute, Yale University, New Haven, United States
| | - Patrick H Degnan
- Department of Microbial Pathogenesis, Yale University, New Haven, United States.,Microbial Sciences Institute, Yale University, New Haven, United States
| | - Ewa Folta-Stogniew
- W.M. Keck Biotechnology Resource Laboratory, Yale University School of Medicine, New Haven, United States
| | - Natasha A Barry
- Department of Microbial Pathogenesis, Yale University, New Haven, United States.,Microbial Sciences Institute, Yale University, New Haven, United States
| | - Andrew L Goodman
- Department of Microbial Pathogenesis, Yale University, New Haven, United States.,Microbial Sciences Institute, Yale University, New Haven, United States
| |
Collapse
|
49
|
Perera NCN, Godahewa GI, Nam BH, Park JY, Lee J. Two metalloenzymes from rockfish (Sebastes schligellii): Deciphering their potential involvement in redox homeostasis against oxidative stress. FISH & SHELLFISH IMMUNOLOGY 2018; 80:31-45. [PMID: 29859306 DOI: 10.1016/j.fsi.2018.05.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/21/2018] [Accepted: 05/29/2018] [Indexed: 06/08/2023]
Abstract
Disturbance in the balance between pro-oxidants and anti-oxidants result oxidative stress in aerobic organisms. However, oxidative stress can be inhibited by enzymatic and non-enzymatic defense mechanisms. Superoxide dismutases (SODs) are well-known scavengers of superoxide radicals, and they protect cells by detoxifying hazardous reactive oxygen species. Here, we have identified and characterized two different SODs, CuZnSOD and MnSOD, from black rockfish (RfCuZnSOD and RfMnSOD, respectively). In silico analysis revealed the well-conserved molecular structures comprising all essential properties of CuZnSOD and MnSOD. Phylogenetic analysis revealed that both RfCuZnSOD and RfMnSOD cladded with their fish counterparts. The recombinant RfSOD proteins demonstrated their potential superoxide scavenging abilities through a xanthine oxidase assay. The optimum temperature and pH conditions for both rRfSODs were 25 °C and pH 8, respectively. Moreover, the potential peroxidation function of rRfCuZnSOD was observed in the presence of HCO3-. The highest peroxidation activity was observed at 100 μg/mL of rRfCuZnSOD using the MTT cell viability assay and flow cytometry. The analogous tissue-specific expression profile indicated ubiquitous expression of both RfCuZnSOD and RfMnSOD in selected tissues of healthy juvenile rockfish. An immune challenge experiment illustrated the altered expression profiles of both RfCuZnSOD and RfMnSOD against lipopolysaccharide, Streptococcus iniae, and polyinosinic-polycytidylic acid (poly I:C). Collectively, these results strengthen the general understanding of the structural and functional characteristics of SODs within the host defense system.
Collapse
Affiliation(s)
- N C N Perera
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - G I Godahewa
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - Bo-Hye Nam
- Biotechnology Research Division, National Institute of Fisheries Science, 408-1 Sirang-ri, Gijang-up, Gijang-gun, Busan, 46083, Republic of Korea
| | - Jung Youn Park
- Biotechnology Research Division, National Institute of Fisheries Science, 408-1 Sirang-ri, Gijang-up, Gijang-gun, Busan, 46083, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea.
| |
Collapse
|
50
|
Lancaster L, Abdallah W, Banta S, Wheeldon I. Engineering enzyme microenvironments for enhanced biocatalysis. Chem Soc Rev 2018; 47:5177-5186. [PMID: 29796541 DOI: 10.1039/c8cs00085a] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Protein engineering provides a means to alter protein structure leading to new functions. Much work has focused on the engineering of enzyme active sites to enhance catalytic activity, however there is an increasing trend towards engineering other aspects of biocatalysts as these efforts can also lead to useful improvements. This tutorial discusses recent advances in engineering an enzyme's local chemical and physical environment, with the goal of enhancing enzyme reaction kinetics, substrate selectivity, and activity in harsh conditions (e.g., low or high pH). By introducing stimuli-responsiveness to these enzyme modifications, dynamic control of activity also becomes possible. These new biomolecular and protein engineering techniques are separate and independent from traditional active site engineering and can therefore be applied synergistically to create new biocatalyst technologies with novel functions.
Collapse
Affiliation(s)
- Louis Lancaster
- Department of Chemical and Environmental Engineering, University of California, Riverside, Riverside, CA 92521, USA.
| | | | | | | |
Collapse
|