1
|
Xu Z, Chen L, Luo Y, Wei YM, Wu NY, Luo LF, Wei YB, Huang J. Advances in metal-organic framework-based nanozymes in ROS scavenging medicine. NANOTECHNOLOGY 2024; 35:362006. [PMID: 38865988 DOI: 10.1088/1361-6528/ad572a] [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: 03/08/2024] [Accepted: 06/12/2024] [Indexed: 06/14/2024]
Abstract
Reactive oxygen species (ROS) play important roles in regulating various physiological functions in the human body, however, excessive ROS can cause serious damage to the human body, considering the various limitations of natural enzymes as scavengers of ROS in the body, the development of better materials for the scavenging of ROS is of great significance to the biomedical field, and nanozymes, as a kind of nanomaterials which can show the activity of natural enzymes. Have a good potential for the development in the area of ROS scavenging. Metal-organic frameworks (MOFs), which are porous crystalline materials with a periodic network structure composed of metal nodes and organic ligands, have been developed with a variety of active nanozymes including catalase-like, superoxide dismutase-like, and glutathione peroxidase-like enzymes due to the adjustability of active sites, structural diversity, excellent biocompatibility, and they have shown a wide range of applications and prospects. In the present review, we first introduce three representative natural enzymes for ROS scavenging in the human body, methods for the detection of relevant enzyme-like activities and mechanisms of enzyme-like clearance are discussed, meanwhile, we systematically summarize the progress of the research on MOF-based nanozymes, including the design strategy, mechanism of action, and medical application, etc. Finally, the current challenges of MOF-based nanozymes are summarized, and the future development direction is anticipated. We hope that this review can contribute to the research of MOF-based nanozymes in the medical field related to the scavenging of ROS.
Collapse
Affiliation(s)
- Zhong Xu
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, People's Republic of China
| | - Liang Chen
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, People's Republic of China
| | - Yan Luo
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, People's Republic of China
| | - Yan-Mei Wei
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, People's Republic of China
| | - Ning-Yuan Wu
- Guangxi Medical University Life Sciences Institute, Guangxi Medical University, Nanning 530021, People's Republic of China
| | - Lan-Fang Luo
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, People's Republic of China
| | - Yong-Biao Wei
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, People's Republic of China
| | - Jin Huang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, People's Republic of China
| |
Collapse
|
2
|
Borgstahl G, Azadmanesh J, Slobodnik K, Struble L, Cone E, Dasgupta M, Lutz W, Kumar S, Natarajan A, Coates L, Weiss K, Myles D, Kroll T. The role of Tyr34 in proton-coupled electron transfer of human manganese superoxide dismutase. RESEARCH SQUARE 2024:rs.3.rs-4494128. [PMID: 38946943 PMCID: PMC11213228 DOI: 10.21203/rs.3.rs-4494128/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Human manganese superoxide dismutase (MnSOD) plays a crucial role in controlling levels of reactive oxygen species (ROS) by converting superoxide (O2 ●-) to molecular oxygen (O2) and hydrogen peroxide (H2O2) with proton-coupled electron transfers (PCETs). The reactivity of human MnSOD is determined by the state of a key catalytic residue, Tyr34, that becomes post-translationally inactivated by nitration in various diseases associated with mitochondrial dysfunction. We previously reported that Tyr34 has an unusual pKa due to its proximity to the Mn metal and undergoes cyclic deprotonation and protonation events to promote the electron transfers of MnSOD. To shed light on the role of Tyr34 MnSOD catalysis, we performed neutron diffraction, X-ray spectroscopy, and quantum chemistry calculations of Tyr34Phe MnSOD in various enzymatic states. The data identifies the contributions of Tyr34 in MnSOD activity that support mitochondrial function and presents a thorough characterization of how a single tyrosine modulates PCET catalysis.
Collapse
|
3
|
Azadmanesh J, Slobodnik K, Struble LR, Cone EA, Dasgupta M, Lutz WE, Kumar S, Natarajan A, Coates L, Weiss KL, Myles DAA, Kroll T, Borgstahl GEO. The role of Tyr34 in proton-coupled electron transfer of human manganese superoxide dismutase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596464. [PMID: 38853997 PMCID: PMC11160768 DOI: 10.1101/2024.05.29.596464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Human manganese superoxide dismutase (MnSOD) plays a crucial role in controlling levels of reactive oxygen species (ROS) by converting superoxide (O 2 •- ) to molecular oxygen (O 2 ) and hydrogen peroxide (H 2 O 2 ) with proton-coupled electron transfers (PCETs). The reactivity of human MnSOD is determined by the state of a key catalytic residue, Tyr34, that becomes post-translationally inactivated by nitration in various diseases associated with mitochondrial dysfunction. We previously reported that Tyr34 has an unusual pK a due to its proximity to the Mn metal and undergoes cyclic deprotonation and protonation events to promote the electron transfers of MnSOD. To shed light on the role of Tyr34 MnSOD catalysis, we performed neutron diffraction, X-ray spectroscopy, and quantum chemistry calculations of Tyr34Phe MnSOD in various enzymatic states. The data identifies the contributions of Tyr34 in MnSOD activity that support mitochondrial function and presents a thorough characterization of how a single tyrosine modulates PCET catalysis.
Collapse
|
4
|
Miranda ER, Shahtout JL, Funai K. Chicken or Egg? Mitochondrial Phospholipids and Oxidative Stress in Disuse-Induced Skeletal Muscle Atrophy. Antioxid Redox Signal 2023; 38:338-351. [PMID: 36301935 PMCID: PMC9986029 DOI: 10.1089/ars.2022.0151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 09/25/2022] [Indexed: 11/13/2022]
Abstract
Significance: Accumulation of reactive oxygen species (ROS) is known to promote cellular damage in multiple cell types. In skeletal muscle, ROS has been implicated in disuse-induced muscle atrophy. However, the molecular origin and mechanism of how disuse promotes ROS and muscle dysfunction remains unclear. Recent Advances: Recently, we implicated membrane lipids of mitochondria to be a potential source of ROS to promote muscle atrophy. Critical Issues: In this review, we discuss evidence that changes in mitochondrial lipids represent a physiologically relevant process by which disuse promotes mitochondrial electron leak and oxidative stress. Future Directions: We further discuss lipid hydroperoxides as a potential downstream mediator of ROS to induce muscle atrophy. Antioxid. Redox Signal. 38, 338-351.
Collapse
Affiliation(s)
- Edwin R. Miranda
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA
| | - Justin L. Shahtout
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA
| | - Katsuhiko Funai
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
5
|
Neupane T, Chambers LR, Godfrey AJ, Monlux MM, Jacobs EJ, Whitworth S, Spawn JE, Clingman SHK, Vergunst KL, Niven FM, Townley JJ, Orion IW, Goodspeed CR, Cooper KA, Cronk JD, Shepherd JN, Langelaan DN. Microbial rhodoquinone biosynthesis proceeds via an atypical RquA-catalyzed amino transfer from S-adenosyl-L-methionine to ubiquinone. Commun Chem 2022; 5:89. [PMID: 36697674 PMCID: PMC9814641 DOI: 10.1038/s42004-022-00711-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 07/20/2022] [Indexed: 01/28/2023] Open
Abstract
Rhodoquinone (RQ) is a close analogue of ubiquinone (UQ) that confers diverse bacterial and eukaryotic taxa the ability to utilize fumarate as an electron acceptor in hypoxic conditions. The RquA protein, identified in a Rhodospirillum rubrum RQ-deficient mutant, has been shown to be required for RQ biosynthesis in bacteria. In this report, we demonstrate that RquA, homologous to SAM-dependent methyltransferases, is necessary and sufficient to catalyze RQ biosynthesis from UQ in vitro. Remarkably, we show that RquA uses SAM as the amino group donor in a substitution reaction that converts UQ to RQ. In contrast to known aminotransferases, RquA does not use pyridoxal 5'-phosphate (PLP) as a coenzyme, but requires the presence of Mn2+ as a cofactor. As these findings reveal, RquA provides an example of a non-canonical SAM-dependent enzyme that does not catalyze methyl transfer, instead it uses SAM in an atypical amino transfer mechanism.
Collapse
Affiliation(s)
- Trilok Neupane
- grid.55602.340000 0004 1936 8200Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS Canada
| | - Lydia R. Chambers
- grid.256410.40000 0001 0668 7980Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA USA
| | - Alexander J. Godfrey
- grid.256410.40000 0001 0668 7980Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA USA
| | - Melina M. Monlux
- grid.256410.40000 0001 0668 7980Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA USA
| | - Evan J. Jacobs
- grid.256410.40000 0001 0668 7980Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA USA
| | - Sophia Whitworth
- grid.256410.40000 0001 0668 7980Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA USA
| | - Jamie E. Spawn
- grid.256410.40000 0001 0668 7980Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA USA
| | - Seo Hee K. Clingman
- grid.256410.40000 0001 0668 7980Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA USA
| | - Kathleen L. Vergunst
- grid.55602.340000 0004 1936 8200Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS Canada
| | - Fair M. Niven
- grid.256410.40000 0001 0668 7980Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA USA
| | - James J. Townley
- grid.256410.40000 0001 0668 7980Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA USA
| | - Iris W. Orion
- grid.256410.40000 0001 0668 7980Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA USA
| | - Carly R. Goodspeed
- grid.256410.40000 0001 0668 7980Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA USA
| | - Kathryn A. Cooper
- grid.256410.40000 0001 0668 7980Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA USA
| | - Jeff D. Cronk
- grid.256410.40000 0001 0668 7980Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA USA
| | - Jennifer N. Shepherd
- grid.256410.40000 0001 0668 7980Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA USA
| | - David N. Langelaan
- grid.55602.340000 0004 1936 8200Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS Canada
| |
Collapse
|
6
|
Shahraki S. Schiff base compounds as artificial metalloenzymes. Colloids Surf B Biointerfaces 2022; 218:112727. [PMID: 35921691 DOI: 10.1016/j.colsurfb.2022.112727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 07/19/2022] [Accepted: 07/24/2022] [Indexed: 11/16/2022]
Abstract
Much research has been done on traditional homogeneous metal catalysts and enzymatic catalysts, but recently a new class of hybrid catalysts called synthetic (artificial) metalloenzymes has been considered by researchers. Metalloenzymes as hybrid catalysts (host-guest systems) have been shown that combine the properties of a homogeneous and also enzymatic catalyst. The hybrid catalyst will have added value such as enantioselectivity or chemo-selectivity. This review focuses on Schiff base complexes that either act as homogeneous artificial enzymes or contribute to the structure of a host in the preparation of hybrid metalloenzymes. Because this approach can virtually be applied to any bio- or synthetic host or guest coordination complex, the details of hybrid catalysts seem important for advance in catalysis.
Collapse
|
7
|
Kumar R, Jafri MS. Computational Modeling of Mitochondria to Understand the Dynamics of Oxidative Stress. Methods Mol Biol 2022; 2497:363-422. [PMID: 35771458 PMCID: PMC9811848 DOI: 10.1007/978-1-0716-2309-1_27] [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: 01/14/2023]
Abstract
Mitochondria are complex organelles that use catabolic metabolism to produce ATP which is the critical energy source for cell function. Oxidative phosphorylation by the electron transport chain, which receives reducing equivalents (NADH and FADH2) from the tricarboxylic acid cycle, also produces reactive oxygen species (ROS) as a by-product at complex I and III. ROS play a significant role in health and disease. In order to better understand this process, a computational model of mitochondrial energy metabolism and the production of ROS has been developed. The model demonstrates the process regulating ROS production and removal and how different energy substrates can affect ROS production.
Collapse
Affiliation(s)
- Rashmi Kumar
- School of Systems Biology, George Mason University, Fairfax, VA, USA
| | - Mohsin S Jafri
- School of Systems Biology, George Mason University, Fairfax, VA, USA.
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
8
|
MnSOD functions as a thermoreceptor activated by low temperature. J Inorg Biochem 2022; 229:111745. [DOI: 10.1016/j.jinorgbio.2022.111745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/22/2022] [Accepted: 01/22/2022] [Indexed: 11/20/2022]
|
9
|
Hennigan JN, Lynch MD. The past, present, and future of enzyme-based therapies. Drug Discov Today 2022; 27:117-133. [PMID: 34537332 PMCID: PMC8714691 DOI: 10.1016/j.drudis.2021.09.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/15/2021] [Accepted: 09/10/2021] [Indexed: 01/03/2023]
Abstract
Enzyme-based therapeutics (EBTs) have the potential to tap into an almost unmeasurable amount of enzyme biodiversity and treat myriad conditions. Although EBTs were some of the first biologics used clinically, the rate of development of newer EBTs has lagged behind that of other biologics. Here, we review the history of EBTs, and discuss the state of each class of EBT, their potential clinical advantages, and the unique challenges to their development. Additionally, we discuss key remaining technical barriers that, if addressed, could increase the diversity and rate of the development of EBTs.
Collapse
Affiliation(s)
| | - Michael D Lynch
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
10
|
Kim YE, Kim J. ROS-Scavenging Therapeutic Hydrogels for Modulation of the Inflammatory Response. ACS APPLIED MATERIALS & INTERFACES 2021; 14:23002-23021. [PMID: 34962774 DOI: 10.1021/acsami.1c18261] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Although reactive oxygen species (ROS) are essential for cellular processes, excessive ROS could be a major cause of various inflammatory diseases because of the oxidation of proteins, DNA, and membrane lipids. It has recently been suggested that the amount of ROS could thus be regulated to treat such physiological disorders. A ROS-scavenging hydrogel is a promising candidate for therapeutic applications because of its high biocompatibility, 3D matrix, and ability to be modified. Approaches to conferring antioxidant properties to normal hydrogels include embedding ROS-scavenging catalytic nanoparticles, modifying hydrogel polymer chains with ROS-adsorbing organic moieties, and incorporating ROS-labile linkers in polymer backbones. Such therapeutic hydrogels can be used for wound healing, cardiovascular diseases, bone repair, ocular diseases, and neurodegenerative disorders. ROS-scavenging hydrogels could eliminate oxidative stress, accelerate the regeneration process, and show synergetic effects with other drugs or therapeutic molecules. In this review, the mechanisms by which ROS are generated and scavenged in the body are outlined, and the effects of high levels of ROS and the resulting oxidative stress on inflammatory diseases are described. Next, the mechanism of ROS scavenging by hydrogels is explained depending on the ROS-scavenging agents embedded within the hydrogel. Lastly, the recent achievements in the development of ROS-scavenging hydrogels to treat various inflammation-associated diseases are presented.
Collapse
Affiliation(s)
- Ye Eun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| |
Collapse
|
11
|
Yuan S, Hahn SA, Miller MP, Sanker S, Calderon MJ, Sullivan M, Dosunmu-Ogunbi AM, Fazzari M, Li Y, Reynolds M, Wood KC, St Croix CM, Stolz D, Cifuentes-Pagano E, Navas P, Shiva S, Schopfer FJ, Pagano PJ, Straub AC. Cooperation between CYB5R3 and NOX4 via coenzyme Q mitigates endothelial inflammation. Redox Biol 2021; 47:102166. [PMID: 34656824 PMCID: PMC8577475 DOI: 10.1016/j.redox.2021.102166] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
NADPH oxidase 4 (NOX4) regulates endothelial inflammation by producing hydrogen peroxide (H2O2) and to a lesser extent O2•-. The ratio of NOX4-derived H2O2 and O2•- can be altered by coenzyme Q (CoQ) mimics. Therefore, we hypothesize that cytochrome b5 reductase 3 (CYB5R3), a CoQ reductase abundant in vascular endothelial cells, regulates inflammatory activation. To examine endothelial CYB5R3 in vivo, we created tamoxifen-inducible endothelium-specific Cyb5r3 knockout mice (R3 KO). Radiotelemetry measurements of systolic blood pressure showed systemic hypotension in lipopolysaccharides (LPS) challenged mice, which was exacerbated in R3 KO mice. Meanwhile, LPS treatment caused greater endothelial dysfunction in R3 KO mice, evaluated by acetylcholine-induced vasodilation in the isolated aorta, accompanied by elevated mRNA expression of vascular adhesion molecule 1 (Vcam-1). Similarly, in cultured human aortic endothelial cells (HAEC), LPS and tumor necrosis factor α (TNF-α) induced VCAM-1 protein expression was enhanced by Cyb5r3 siRNA, which was ablated by silencing the Nox4 gene simultaneously. Moreover, super-resolution confocal microscopy indicated mitochondrial co-localization of CYB5R3 and NOX4 in HAECs. APEX2-based electron microscopy and proximity biotinylation also demonstrated CYB5R3's localization on the mitochondrial outer membrane and its interaction with NOX4, which was further confirmed by the proximity ligation assay. Notably, Cyb5r3 knockdown HAECs showed less total H2O2 but more mitochondrial O2•-. Using inactive or non-membrane bound active CYB5R3, we found that CYB5R3 activity and membrane translocation are needed for optimal generation of H2O2 by NOX4. Lastly, cells lacking the CoQ synthesizing enzyme COQ6 showed decreased NOX4-derived H2O2, indicating a requirement for endogenous CoQ in NOX4 activity. In conclusion, CYB5R3 mitigates endothelial inflammatory activation by assisting in NOX4-dependent H2O2 generation via CoQ.
Collapse
Affiliation(s)
- Shuai Yuan
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Scott A Hahn
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Megan P Miller
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Subramaniam Sanker
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael J Calderon
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mara Sullivan
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - Atinuke M Dosunmu-Ogunbi
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Marco Fazzari
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yao Li
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael Reynolds
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Katherine C Wood
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Donna Stolz
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eugenia Cifuentes-Pagano
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Placido Navas
- Centro Andaluz de Biología del Desarrollo and CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain, Spain
| | - Sruti Shiva
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Francisco J Schopfer
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Liver Research Center (PLRC), University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick J Pagano
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Center for Microvascular Research, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
12
|
Bonetta R, Hunter GJ, Trinh CH, Borowski T, Fenech AG, Kulp M, Tabares LC, Un S, Hunter T. Substitution of histidine 30 by asparagine in manganese superoxide dismutase alters biophysical properties and supports proliferation in a K562 leukemia cell line. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2021; 50:571-585. [PMID: 34021366 PMCID: PMC8190026 DOI: 10.1007/s00249-021-01544-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023]
Abstract
We have generated a mutant of C. elegans manganese superoxide dismutase at histidine 30 by site-directed mutagenesis. The structure was solved at a resolution of 1.52 Å by X-ray crystallography (pdb: 6S0D). His30 was targeted, as it forms as a gateway residue at the top of the solvent access funnel to the active site, together with Tyr34. In the wild-type protein, these gateway residues are involved in the hydrogen-bonding network providing the protons necessary for the catalytic reaction at the metal center. However, biophysical characterization and cell viability experiments reveal that a mutation from histidine to asparagine in the H30N mutant modifies metal selectivity in the protein, favoring the uptake of iron over manganese in minimal media conditions, alters active-site coordination from the characteristic trigonal bipyramidal to octahedral geometry, and encourages cellular proliferation in K562 cells, when added exogenously to the cells.
Collapse
Affiliation(s)
- Rosalin Bonetta
- Centre of Molecular Medicine & Biobanking, University of Malta, Msida, Malta. .,Barts and the London, School of Medicine and Dentistry, QMUL, Victoria, Malta.
| | - Gary J Hunter
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Chi H Trinh
- Astbury Centre for Structural Molecular Biology, Institute of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Tomasz Borowski
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Krakow, Poland
| | - Anthony G Fenech
- Department of Clinical Pharmacology and Therapeutics, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Maria Kulp
- Department of Chemistry, Tallinn University of Technology, Tallinn, Estonia
| | - Leandro C Tabares
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, 91198, Gif-sur-Yvette, France
| | - Sun Un
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, 91198, Gif-sur-Yvette, France
| | - Thérèse Hunter
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| |
Collapse
|
13
|
Azadmanesh J, Lutz WE, Coates L, Weiss KL, Borgstahl GEO. Direct detection of coupled proton and electron transfers in human manganese superoxide dismutase. Nat Commun 2021; 12:2079. [PMID: 33824320 PMCID: PMC8024262 DOI: 10.1038/s41467-021-22290-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/26/2021] [Indexed: 11/30/2022] Open
Abstract
Human manganese superoxide dismutase is a critical oxidoreductase found in the mitochondrial matrix. Concerted proton and electron transfers are used by the enzyme to rid the mitochondria of O2•-. The mechanisms of concerted transfer enzymes are typically unknown due to the difficulties in detecting the protonation states of specific residues and solvent molecules at particular redox states. Here, neutron diffraction of two redox-controlled manganese superoxide dismutase crystals reveal the all-atom structures of Mn3+ and Mn2+ enzyme forms. The structures deliver direct data on protonation changes between oxidation states of the metal. Observations include glutamine deprotonation, the involvement of tyrosine and histidine with altered pKas, and four unusual strong-short hydrogen bonds, including a low barrier hydrogen bond. We report a concerted proton and electron transfer mechanism for human manganese superoxide dismutase from the direct visualization of active site protons in Mn3+ and Mn2+ redox states.
Collapse
Affiliation(s)
- Jahaun Azadmanesh
- Department of Biochemistry and Molecular Biology, 985870 Nebraska Medical Center, Omaha, NE, USA
| | - William E Lutz
- Eppley Institute for Cancer and Allied Diseases, 986805 Nebraska Medical Center, Omaha, NE, USA
| | - Leighton Coates
- Second Target Station, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Kevin L Weiss
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Gloria E O Borgstahl
- Department of Biochemistry and Molecular Biology, 985870 Nebraska Medical Center, Omaha, NE, USA.
- Eppley Institute for Cancer and Allied Diseases, 986805 Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
14
|
Sconyers DJ, Shaughnessy CI, Lee HJ, Subramaniam B, Leonard KC, Blakemore JD. Enhancing Molecular Electrocatalysis of CO 2 Reduction with Pressure-Tunable CO 2 -Expanded Electrolytes. CHEMSUSCHEM 2020; 13:6338-6345. [PMID: 32196939 DOI: 10.1002/cssc.202000390] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/19/2020] [Indexed: 06/10/2023]
Abstract
Electrochemical studies of CO2 conversion by molecular catalysts are typically carried out in a narrow range of near-ambient CO2 pressures wherein low CO2 solubilities in the liquid phase can limit the rate of CO2 reduction. In this study, five-fold rate enhancements are enabled by pairing CO2 -expanded electrolytes (CXEs), a class of media that accommodate multimolar concentrations of CO2 in organic solvents at modest pressures, with a homogeneous molecular electrocatalyst, [Re(CO)3 (bpy)Cl] (1, bpy=2,2'-bipyridyl). Analysis of cyclic voltammetry data reveals pressure-tunable rate behavior, with first-order kinetics at moderate CO2 pressures giving way to zero-order kinetics at higher pressures. The significant enhancement in the space-time yield of CO demonstrates that CXEs offer a simple yet powerful strategy for unlocking the intrinsic potential of molecular catalysts by mitigating CO2 solubility limitations commonly encountered in conventional liquid electrolytes. Moreover, our findings reveal that 1, a workhorse molecular catalyst, performs with intrinsic kinetic behavior, which is competitive with fast enzymes under optimal conditions in CXEs.
Collapse
Affiliation(s)
- David J Sconyers
- Center for Environmentally Beneficial Catalysis, University of Kansas, 1501 Wakarusa Drive, Lawrence, Kansas, 66047, USA
- Department of Chemistry, University of Kansas, 1567 Irving Hill Road, Lawrence, Kansas, 66045, USA
| | - Charles I Shaughnessy
- Center for Environmentally Beneficial Catalysis, University of Kansas, 1501 Wakarusa Drive, Lawrence, Kansas, 66047, USA
- Department of Chemical and Petroleum Engineering, University of Kansas, 1530 W 15th Street, Lawrence, Kansas, 66045, USA
| | - Hyun-Jin Lee
- Center for Environmentally Beneficial Catalysis, University of Kansas, 1501 Wakarusa Drive, Lawrence, Kansas, 66047, USA
| | - Bala Subramaniam
- Center for Environmentally Beneficial Catalysis, University of Kansas, 1501 Wakarusa Drive, Lawrence, Kansas, 66047, USA
- Department of Chemical and Petroleum Engineering, University of Kansas, 1530 W 15th Street, Lawrence, Kansas, 66045, USA
| | - Kevin C Leonard
- Center for Environmentally Beneficial Catalysis, University of Kansas, 1501 Wakarusa Drive, Lawrence, Kansas, 66047, USA
- Department of Chemical and Petroleum Engineering, University of Kansas, 1530 W 15th Street, Lawrence, Kansas, 66045, USA
| | - James D Blakemore
- Center for Environmentally Beneficial Catalysis, University of Kansas, 1501 Wakarusa Drive, Lawrence, Kansas, 66047, USA
- Department of Chemistry, University of Kansas, 1567 Irving Hill Road, Lawrence, Kansas, 66045, USA
| |
Collapse
|
15
|
Eshima H, Siripoksup P, Mahmassani ZS, Johnson JM, Ferrara PJ, Verkerke ARP, Salcedo A, Drummond MJ, Funai K. Neutralizing mitochondrial ROS does not rescue muscle atrophy induced by hindlimb unloading in female mice. J Appl Physiol (1985) 2020; 129:124-132. [PMID: 32552434 DOI: 10.1152/japplphysiol.00456.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Excess reactive oxygen species (ROS) induced by physical inactivity is associated with muscle atrophy and muscle weakness. However, the role of mitochondrial ROS on disuse-induced muscle atrophy is not fully understood. The purpose of this study was to utilize a genetic strategy to examine the effect of neutralizing mitochondrial ROS on disuse-induced skeletal muscle atrophy. This was accomplished by placing wild-type (WT) and mitochondrial-targeted catalase-expressing (MCAT) littermate mice on 7 days of hindlimb unloading. After assessment of body weight and composition, muscles were analyzed for individual muscle mass, force-generating capacity, fiber type, cross-sectional area, and mitochondrial function, including H2O2 production. Despite a successful attenuation of mitochondrial ROS, MCAT mice were not protected from muscle atrophy. No differences were observed in body composition, lean mass, individual muscle masses, force-generating capacity, or muscle fiber cross-sectional area. These data suggest that neutralizing mitochondrial ROS is insufficient to suppress disuse-induced loss of skeletal muscle mass and contractile function.NEW & NOTEWORTHY The premise of this study was to examine the efficacy of genetic suppression of mitochondrial reactive oxygen species (ROS) to attenuate disuse-induced muscle atrophy and muscle weakness. Neutralization of mitochondrial ROS by MCAT expression was insufficient to rescue muscle atrophy and muscle weakness.
Collapse
Affiliation(s)
- Hiroaki Eshima
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah.,Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Piyarat Siripoksup
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah.,Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Ziad S Mahmassani
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah.,Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Jordan M Johnson
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Patrick J Ferrara
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Anthony R P Verkerke
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Anahy Salcedo
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Micah J Drummond
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah
| | - Katsuhiko Funai
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah
| |
Collapse
|
16
|
Fuchs P, Rugen N, Carrie C, Elsässer M, Finkemeier I, Giese J, Hildebrandt TM, Kühn K, Maurino VG, Ruberti C, Schallenberg-Rüdinger M, Steinbeck J, Braun HP, Eubel H, Meyer EH, Müller-Schüssele SJ, Schwarzländer M. Single organelle function and organization as estimated from Arabidopsis mitochondrial proteomics. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2020; 101:420-441. [PMID: 31520498 DOI: 10.1111/tpj.14534] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/23/2019] [Accepted: 08/28/2019] [Indexed: 05/14/2023]
Abstract
Mitochondria host vital cellular functions, including oxidative phosphorylation and co-factor biosynthesis, which are reflected in their proteome. At the cellular level plant mitochondria are organized into hundreds of discrete functional entities, which undergo dynamic fission and fusion. It is the individual organelle that operates in the living cell, yet biochemical and physiological assessments have exclusively focused on the characteristics of large populations of mitochondria. Here, we explore the protein composition of an individual average plant mitochondrion to deduce principles of functional and structural organisation. We perform proteomics on purified mitochondria from cultured heterotrophic Arabidopsis cells with intensity-based absolute quantification and scale the dataset to the single organelle based on criteria that are justified by experimental evidence and theoretical considerations. We estimate that a total of 1.4 million protein molecules make up a single Arabidopsis mitochondrion on average. Copy numbers of the individual proteins span five orders of magnitude, ranging from >40 000 for Voltage-Dependent Anion Channel 1 to sub-stoichiometric copy numbers, i.e. less than a single copy per single mitochondrion, for several pentatricopeptide repeat proteins that modify mitochondrial transcripts. For our analysis, we consider the physical and chemical constraints of the single organelle and discuss prominent features of mitochondrial architecture, protein biogenesis, oxidative phosphorylation, metabolism, antioxidant defence, genome maintenance, gene expression, and dynamics. While assessing the limitations of our considerations, we exemplify how our understanding of biochemical function and structural organization of plant mitochondria can be connected in order to obtain global and specific insights into how organelles work.
Collapse
Affiliation(s)
- Philippe Fuchs
- Institut für Biologie und Biotechnologie der Pflanzen (IBBP), Westfälische Wilhelms-Universität, Schlossplatz 7-8, 48143, Münster, Germany
- Institut für Nutzpflanzenforschung und Ressourcenschutz (INRES), Rheinische Friedrich-Wilhelms-Universität Bonn, Friedrich-Ebert-Allee 144, 53113, Bonn, Germany
| | - Nils Rugen
- Institut für Pflanzengenetik, Leibniz Universität Hannover, Herrenhäuser Str. 2, 30419, Hannover, Germany
| | - Chris Carrie
- Department Biologie I - Botanik, Ludwig-Maximilians-Universität München, Grosshadernerstr. 2-4, 82152, Planegg-Martinsried, Germany
| | - Marlene Elsässer
- Institut für Biologie und Biotechnologie der Pflanzen (IBBP), Westfälische Wilhelms-Universität, Schlossplatz 7-8, 48143, Münster, Germany
- Institut für Nutzpflanzenforschung und Ressourcenschutz (INRES), Rheinische Friedrich-Wilhelms-Universität Bonn, Friedrich-Ebert-Allee 144, 53113, Bonn, Germany
- Institut für Zelluläre und Molekulare Botanik (IZMB), Rheinische Friedrich-Wilhelms-Universität Bonn, Kirschallee 1, 53115, Bonn, Germany
| | - Iris Finkemeier
- Institut für Biologie und Biotechnologie der Pflanzen (IBBP), Westfälische Wilhelms-Universität, Schlossplatz 7-8, 48143, Münster, Germany
| | - Jonas Giese
- Institut für Biologie und Biotechnologie der Pflanzen (IBBP), Westfälische Wilhelms-Universität, Schlossplatz 7-8, 48143, Münster, Germany
| | - Tatjana M Hildebrandt
- Institut für Pflanzengenetik, Leibniz Universität Hannover, Herrenhäuser Str. 2, 30419, Hannover, Germany
| | - Kristina Kühn
- Institut für Biologie, Martin-Luther-Universität Halle-Wittenberg, Weinbergweg 10, 06120, Halle/Saale, Germany
| | - Veronica G Maurino
- Institute of Developmental and Molecular Biology of Plants, and Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Cristina Ruberti
- Institut für Biologie und Biotechnologie der Pflanzen (IBBP), Westfälische Wilhelms-Universität, Schlossplatz 7-8, 48143, Münster, Germany
| | - Mareike Schallenberg-Rüdinger
- Institut für Zelluläre und Molekulare Botanik (IZMB), Rheinische Friedrich-Wilhelms-Universität Bonn, Kirschallee 1, 53115, Bonn, Germany
| | - Janina Steinbeck
- Institut für Biologie und Biotechnologie der Pflanzen (IBBP), Westfälische Wilhelms-Universität, Schlossplatz 7-8, 48143, Münster, Germany
| | - Hans-Peter Braun
- Institut für Pflanzengenetik, Leibniz Universität Hannover, Herrenhäuser Str. 2, 30419, Hannover, Germany
| | - Holger Eubel
- Institut für Pflanzengenetik, Leibniz Universität Hannover, Herrenhäuser Str. 2, 30419, Hannover, Germany
| | - Etienne H Meyer
- Institut für Biologie, Martin-Luther-Universität Halle-Wittenberg, Weinbergweg 10, 06120, Halle/Saale, Germany
| | - Stefanie J Müller-Schüssele
- Institut für Nutzpflanzenforschung und Ressourcenschutz (INRES), Rheinische Friedrich-Wilhelms-Universität Bonn, Friedrich-Ebert-Allee 144, 53113, Bonn, Germany
| | - Markus Schwarzländer
- Institut für Biologie und Biotechnologie der Pflanzen (IBBP), Westfälische Wilhelms-Universität, Schlossplatz 7-8, 48143, Münster, Germany
| |
Collapse
|
17
|
Heden TD, Johnson JM, Ferrara PJ, Eshima H, Verkerke ARP, Wentzler EJ, Siripoksup P, Narowski TM, Coleman CB, Lin CT, Ryan TE, Reidy PT, de Castro Brás LE, Karner CM, Burant CF, Maschek JA, Cox JE, Mashek DG, Kardon G, Boudina S, Zeczycki TN, Rutter J, Shaikh SR, Vance JE, Drummond MJ, Neufer PD, Funai K. Mitochondrial PE potentiates respiratory enzymes to amplify skeletal muscle aerobic capacity. SCIENCE ADVANCES 2019; 5:eaax8352. [PMID: 31535029 PMCID: PMC6739096 DOI: 10.1126/sciadv.aax8352] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 08/15/2019] [Indexed: 05/08/2023]
Abstract
Exercise capacity is a strong predictor of all-cause mortality. Skeletal muscle mitochondrial respiratory capacity, its biggest contributor, adapts robustly to changes in energy demands induced by contractile activity. While transcriptional regulation of mitochondrial enzymes has been extensively studied, there is limited information on how mitochondrial membrane lipids are regulated. Here, we show that exercise training or muscle disuse alters mitochondrial membrane phospholipids including phosphatidylethanolamine (PE). Addition of PE promoted, whereas removal of PE diminished, mitochondrial respiratory capacity. Unexpectedly, skeletal muscle-specific inhibition of mitochondria-autonomous synthesis of PE caused respiratory failure because of metabolic insults in the diaphragm muscle. While mitochondrial PE deficiency coincided with increased oxidative stress, neutralization of the latter did not rescue lethality. These findings highlight the previously underappreciated role of mitochondrial membrane phospholipids in dynamically controlling skeletal muscle energetics and function.
Collapse
Affiliation(s)
- Timothy D. Heden
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Kinesiology, East Carolina University, Greenville, NC, USA
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Jordan M. Johnson
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Kinesiology, East Carolina University, Greenville, NC, USA
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, USA
| | - Patrick J. Ferrara
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Kinesiology, East Carolina University, Greenville, NC, USA
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, USA
| | - Hiroaki Eshima
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
| | - Anthony R. P. Verkerke
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Kinesiology, East Carolina University, Greenville, NC, USA
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, USA
| | - Edward J. Wentzler
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Kinesiology, East Carolina University, Greenville, NC, USA
| | - Piyarat Siripoksup
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, USA
| | - Tara M. Narowski
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Kinesiology, East Carolina University, Greenville, NC, USA
| | - Chanel B. Coleman
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Kinesiology, East Carolina University, Greenville, NC, USA
| | - Chien-Te Lin
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Physiology, East Carolina University, Greenville, NC, USA
| | - Terence E. Ryan
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Physiology, East Carolina University, Greenville, NC, USA
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL, USA
| | - Paul T. Reidy
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, USA
| | | | - Courtney M. Karner
- Department of Orthopedic Surgery & Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Charles F. Burant
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, USA
| | - J. Alan Maschek
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, USA
| | - James E. Cox
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, USA
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Douglas G. Mashek
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Sihem Boudina
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
| | - Tonya N. Zeczycki
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Biochemistry and Molecular Biology, East Carolina University, Greenville, NC, USA
| | - Jared Rutter
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Saame Raza Shaikh
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Biochemistry and Molecular Biology, East Carolina University, Greenville, NC, USA
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, USA
| | - Jean E. Vance
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Micah J. Drummond
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
| | - P. Darrell Neufer
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Kinesiology, East Carolina University, Greenville, NC, USA
- Department of Physiology, East Carolina University, Greenville, NC, USA
| | - Katsuhiko Funai
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Kinesiology, East Carolina University, Greenville, NC, USA
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, USA
- Department of Physiology, East Carolina University, Greenville, NC, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Corresponding author.
| |
Collapse
|
18
|
Rudin D, Roos NJ, Duthaler U, Krähenbühl S. Toxicity of metamizole on differentiating HL60 cells and human neutrophil granulocytes. Toxicology 2019; 426:152254. [PMID: 31356851 DOI: 10.1016/j.tox.2019.152254] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/15/2019] [Accepted: 07/24/2019] [Indexed: 12/18/2022]
Abstract
Metamizole is an analgesic and antipyretic with a superior analgesic efficacy than paracetamol. Since metamizole can cause neutropenia and agranulocytosis, it is currently used in only few countries. In a previous study, we have shown that N-methyl-4-aminoantipyrine (MAA), the active metamizole metabolite, reacts with hemin and forms an electrophilic metabolite that is toxic for HL60 cells, but not for mature neutrophil granulocytes. In the current study, we investigated the toxicity of hemin (12.5 μM) and MAA (100 μM) on differentiating HL60 cells. In undifferentiated HL60 cells, hemin decreased the viability and this effect was significantly increased by MAA. Similarly, hemin/MAA was more toxic than hemin alone on human cord blood cells. At 3 days (metamyelocyte stage) and 5 days of differentiation (mature neutrophils), hemin/MAA was not toxic on HL60 cells, whereas hemin alone was still toxic. No toxicity was observed on freshly isolated human neutrophils. The protein expression of enzymes responsible for hemin metabolism increased with HL60 cell differentiation. Inhibition of heme oxygenase-1 or cytochrome P450 reductase increased the toxicity of hemin and hemin/MAA in undifferentiated, but only for hemin in differentiated HL60 cells. Similar to the enzymes involved in hemin metabolism, the protein expression of enzymes involved in antioxidative defense and the cellular glutathione pool increased with HL60 cell differentiation. In conclusion, HL60 cells become resistant to the toxicity of hemin/MAA and partly also of hemin during their differentiation. This resistance is associated with the development of heme metabolism and of the antioxidative defense system including the cellular glutathione pool.
Collapse
Affiliation(s)
- Deborah Rudin
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, Schanzenstrasse 55, 4031, Basel, Switzerland; Department of Biomedicine, University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland.
| | - Noëmi Johanna Roos
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, Schanzenstrasse 55, 4031, Basel, Switzerland; Department of Biomedicine, University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland.
| | - Urs Duthaler
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, Schanzenstrasse 55, 4031, Basel, Switzerland; Department of Biomedicine, University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland.
| | - Stephan Krähenbühl
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, Schanzenstrasse 55, 4031, Basel, Switzerland; Department of Biomedicine, University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Missionsstrasse 64, 4055, Basel, Switzerland.
| |
Collapse
|
19
|
Chemical Basis of Reactive Oxygen Species Reactivity and Involvement in Neurodegenerative Diseases. Int J Mol Sci 2019; 20:ijms20102407. [PMID: 31096608 PMCID: PMC6566277 DOI: 10.3390/ijms20102407] [Citation(s) in RCA: 456] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/09/2019] [Accepted: 05/13/2019] [Indexed: 12/12/2022] Open
Abstract
Increasing numbers of individuals suffer from neurodegenerative diseases, which are characterized by progressive loss of neurons. Oxidative stress, in particular, the overproduction of Reactive Oxygen Species (ROS), play an important role in the development of these diseases, as evidenced by the detection of products of lipid, protein and DNA oxidation in vivo. Even if they participate in cell signaling and metabolism regulation, ROS are also formidable weapons against most of the biological materials because of their intrinsic nature. By nature too, neurons are particularly sensitive to oxidation because of their high polyunsaturated fatty acid content, weak antioxidant defense and high oxygen consumption. Thus, the overproduction of ROS in neurons appears as particularly deleterious and the mechanisms involved in oxidative degradation of biomolecules are numerous and complexes. This review highlights the production and regulation of ROS, their chemical properties, both from kinetic and thermodynamic points of view, the links between them, and their implication in neurodegenerative diseases.
Collapse
|
20
|
Kizhuveetil U, Palukuri MV, Sharma P, Karunagaran D, Rengaswamy R, Suraishkumar GK. Entrainment of superoxide rhythm by menadione in HCT116 colon cancer cells. Sci Rep 2019; 9:3347. [PMID: 30833672 PMCID: PMC6399287 DOI: 10.1038/s41598-019-40017-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 02/07/2019] [Indexed: 11/08/2022] Open
Abstract
Reactive oxygen species (ROS) are primary effectors of cytotoxicity induced by many anti-cancer drugs. Rhythms in the pseudo-steady-state (PSS) levels of particular intracellular ROS in cancer cells and their relevance to drug effectiveness are unknown thus far. We report that the PSS levels of intracellular superoxide (SOX), an important ROS, exhibit an inherent rhythm in HCT116 colon cancer cells, which is entrained (reset) by the SOX inducer, menadione (MD). This reset was dependent on the expression of p53, and it doubled the sensitivity of the cells to MD. The period of oscillation was found to have a linear correlation with MD concentration, given by the equation, T, in h = 23.52 - 1.05 [MD concentration in µM]. Further, we developed a mathematical model to better understand the molecular mechanisms involved in rhythm reset. Biologically meaningful parameters were obtained through parameter estimation techniques; the model can predict experimental profiles of SOX, establish qualitative relations between interacting species in the system and serves as an important tool to understand the profiles of various species. The model was also able to successfully predict the rhythm reset in MD treated hepatoma cell line, HepG2.
Collapse
Affiliation(s)
- Uma Kizhuveetil
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences building, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Meghana V Palukuri
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Priyanshu Sharma
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences building, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Devarajan Karunagaran
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences building, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Raghunathan Rengaswamy
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai, 600036, India
| | - G K Suraishkumar
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences building, Indian Institute of Technology Madras, Chennai, 600036, India.
| |
Collapse
|
21
|
Affiliation(s)
- Valerie Vaissier Welborn
- Kenneth S. Pitzer Center for Theoretical Chemistry and Department of Chemistry, University of California, Berkeley, California 94720, United States
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Teresa Head-Gordon
- Kenneth S. Pitzer Center for Theoretical Chemistry and Department of Chemistry, University of California, Berkeley, California 94720, United States
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
- Department of Chemical and Biomolecular Engineering and Department of Bioengineering, University of California, Berkeley, California 94720, United States
| |
Collapse
|
22
|
Carballal S, Valez V, Alvarez-Paggi D, Tovmasyan A, Batinic-Haberle I, Ferrer-Sueta G, Murgida DH, Radi R. Manganese porphyrin redox state in endothelial cells: Resonance Raman studies and implications for antioxidant protection towards peroxynitrite. Free Radic Biol Med 2018; 126:379-392. [PMID: 30144631 DOI: 10.1016/j.freeradbiomed.2018.08.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 08/18/2018] [Accepted: 08/20/2018] [Indexed: 10/28/2022]
Abstract
Cationic manganese(III) ortho N-substituted pyridylporphyrins (MnP) act as efficient antioxidants catalyzing superoxide dismutation and accelerating peroxynitrite reduction. Importantly, MnP can reach mitochondria offering protection against reactive species in different animal models of disease. Although an LC-MS/MS-based method for MnP quantitation and subcellular distribution has been reported, a direct method capable of evaluating both the uptake and the redox state of MnP in living cells has not yet been developed. In the present work we applied resonance Raman (RR) spectroscopy to analyze the intracellular accumulation of two potent MnP-based lipophilic SOD mimics, MnTnBuOE-2-PyP5+ and MnTnHex-2-PyP5+ within endothelial cells. RR experiments with isolated mitochondria revealed that the reduction of Mn(III)P was affected by inhibitors of the electron transport chain, supporting the action of MnP as efficient redox active compounds in mitochondria. Indeed, RR spectra confirmed that MnP added in the Mn(III) state can be incorporated into the cells, readily reduced by intracellular components to the Mn(II) state and oxidized by peroxynitrite. To assess the combined impact of reactivity and bioavailability, we studied the kinetics of Mn(III)TnBuOE-2-PyP5+ with peroxynitrite and evaluated the cytoprotective capacity of MnP by exposing the endothelial cells to nitro-oxidative stress induced by peroxynitrite. We observed a preservation of normal mitochondrial function, attenuation of cell damage and prevention of apoptotic cell death. These data introduce a novel application of RR spectroscopy for the direct detection of MnP and their redox states inside living cells, and helps to rationalize their antioxidant capacity in biological systems.
Collapse
Affiliation(s)
- Sebastián Carballal
- Departmento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo, Uruguay
| | - Valeria Valez
- Departmento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo, Uruguay
| | - Damián Alvarez-Paggi
- Departamento de Química Inorgánica, Analítica y Química Física and INQUIMAE-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pab. 2, piso 1, C1428EHA Buenos Aires, Argentina
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Gerardo Ferrer-Sueta
- Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo, Uruguay; Laboratorio de Fisicoquímica Biológica, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Daniel H Murgida
- Departamento de Química Inorgánica, Analítica y Química Física and INQUIMAE-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pab. 2, piso 1, C1428EHA Buenos Aires, Argentina
| | - Rafael Radi
- Departmento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
23
|
Abstract
The concept of cell signaling in the context of nonenzyme-assisted protein modifications by reactive electrophilic and oxidative species, broadly known as redox signaling, is a uniquely complex topic that has been approached from numerous different and multidisciplinary angles. Our Review reflects on five aspects critical for understanding how nature harnesses these noncanonical post-translational modifications to coordinate distinct cellular activities: (1) specific players and their generation, (2) physicochemical properties, (3) mechanisms of action, (4) methods of interrogation, and (5) functional roles in health and disease. Emphasis is primarily placed on the latest progress in the field, but several aspects of classical work likely forgotten/lost are also recollected. For researchers with interests in getting into the field, our Review is anticipated to function as a primer. For the expert, we aim to stimulate thought and discussion about fundamentals of redox signaling mechanisms and nuances of specificity/selectivity and timing in this sophisticated yet fascinating arena at the crossroads of chemistry and biology.
Collapse
Affiliation(s)
- Saba Parvez
- Department of Pharmacology and Toxicology, College of
Pharmacy, University of Utah, Salt Lake City, Utah, 84112, USA
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Marcus J. C. Long
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Jesse R. Poganik
- Ecole Polytechnique Fédérale de Lausanne,
Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Yimon Aye
- Ecole Polytechnique Fédérale de Lausanne,
Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
- Department of Biochemistry, Weill Cornell Medicine, New
York, New York, 10065, USA
| |
Collapse
|
24
|
Erxleben A. Transition metal salen complexes in bioinorganic and medicinal chemistry. Inorganica Chim Acta 2018. [DOI: 10.1016/j.ica.2017.06.060] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
25
|
Ferrer-Sueta G, Campolo N, Trujillo M, Bartesaghi S, Carballal S, Romero N, Alvarez B, Radi R. Biochemistry of Peroxynitrite and Protein Tyrosine Nitration. Chem Rev 2018; 118:1338-1408. [DOI: 10.1021/acs.chemrev.7b00568] [Citation(s) in RCA: 292] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Gerardo Ferrer-Sueta
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Nicolás Campolo
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Madia Trujillo
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Silvina Bartesaghi
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Sebastián Carballal
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Natalia Romero
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Beatriz Alvarez
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
26
|
Demicheli V, Moreno DM, Radi R. Human Mn-superoxide dismutase inactivation by peroxynitrite: a paradigm of metal-catalyzed tyrosine nitration in vitro and in vivo. Metallomics 2018; 10:679-695. [DOI: 10.1039/c7mt00348j] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Nitration of human MnSOD at active site Tyr34 represents a biologically-relevant oxidative post-translational modification that causes enzyme inactivation.
Collapse
Affiliation(s)
- Verónica Demicheli
- Departmento de Bioquimica
- Facultad de Medicina
- Center for Free Radical and Biomedical Research
- Universidad de la República
- Montevideo
| | - Diego M. Moreno
- Instituto de Química Rosario (IQUIR, CONICET-UNR)
- Área Química General e Inorgánica
- Facultad de Ciencias Bioquímicas y Farmacéuticas
- Universidad Nacional de Rosario
- Argentina
| | - Rafael Radi
- Departmento de Bioquimica
- Facultad de Medicina
- Center for Free Radical and Biomedical Research
- Universidad de la República
- Montevideo
| |
Collapse
|
27
|
Yamada T, Steinz MM, Kenne E, Lanner JT. Muscle Weakness in Rheumatoid Arthritis: The Role of Ca 2+ and Free Radical Signaling. EBioMedicine 2017; 23:12-19. [PMID: 28781131 PMCID: PMC5605300 DOI: 10.1016/j.ebiom.2017.07.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/13/2017] [Accepted: 07/24/2017] [Indexed: 02/07/2023] Open
Abstract
In addition to the primary symptoms arising from inflammatory processes in the joints, muscle weakness is commonly reported by patients with rheumatoid arthritis (RA). Muscle weakness not only reduces the quality of life for the affected patients, but also dramatically increases the burden on society since patients' work ability decreases. A 25–70% reduction in muscular strength has been observed in pateints with RA when compared with age-matched healthy controls. The reduction in muscle strength is often larger than what could be explained by the reduction in muscle size in patients with RA, which indicates that intracellular (intrinsic) muscle dysfunction plays an important role in the underlying mechanism of muscle weakness associated with RA. In this review, we highlight the present understanding of RA-associated muscle weakness with special focus on how enhanced Ca2 + release from the ryanodine receptor and free radicals (reactive oxygen/nitrogen species) contributes to muscle weakness, and recent developments of novel therapeutic interventions. Muscle weakness is commonly reported by patients with rheumatoid arthritis (RA). Intrinsic muscle weakness is important in the underlying mechanisms of muscle weakness associated with rheumatoid arthritis. Enhanced Ca2 + release and peroxynitrite-induced stress contributes to RA-induced muscle weakness.
Collapse
Affiliation(s)
- Takashi Yamada
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Maarten M Steinz
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ellinor Kenne
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
28
|
Miller AF, Wang T. A Single Outer-Sphere Mutation Stabilizes apo-Mn Superoxide Dismutase by 35 °C and Disfavors Mn Binding. Biochemistry 2017; 56:3787-3799. [PMID: 28704037 PMCID: PMC6010041 DOI: 10.1021/acs.biochem.7b00175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The catalytic active site of Mn-specific superoxide dismutase (MnSOD) is organized around a redox-active Mn ion. The most highly conserved difference between MnSODs and the homologous FeSODs is the origin of a Gln in the second coordination sphere. In MnSODs it derives from the C-terminal domain whereas in FeSODs it derives from the N-terminal domain, yet its side chain occupies almost superimposable positions in the active sites of these two types of SODs. Mutation of this Gln69 to Glu in Escherichia coli FeSOD increased the Fe3+/2+ reduction midpoint potential by >0.6 V without disrupting the structure or Fe binding [ Yikilmaz, E., Rodgers, D. W., and Miller, A.-F. ( 2006 ) Biochemistry 45 ( 4 ), 1151 - 1161 ]. We now describe the analogous Q146E mutant of MnSOD, explaining its low Mn content in terms increased stability of the apo-Mn protein. In 0.8 M guanidinium HCl, Q146E-apoMnSOD displays an apparent melting midpoint temperature (Tm) 35 °C higher that of wild-type (WT) apoMnSOD, whereas the Tm of WT-holoMnSOD is only 20 °C higher than that of WT-apoMnSOD. In contrast, the Tm attributed to Q146E-holoMnSOD is 40 °C lower than that of Q146E-apoMnSOD. Thus, our data refute the notion that the WT residues optimize the structural stability of the protein and instead are consistent with conservation on the basis of enzyme function and therefore ability to bind metal ion. We propose that the WT-MnSOD protein conserves a destabilizing amino acid at position 146 as part of a strategy to favor metal ion binding.
Collapse
Affiliation(s)
- Anne-Frances Miller
- Department of Chemistry, University of Kentucky, Lexington Kentucky, 40506, United States
- Department of Molecular and Cellular Biochemistry and Center for Structural Biology, University of Kentucky, 741 S. Limestone Street, Lexington, KY. 40536-0509
| | - Ting Wang
- Department of Chemistry, University of Kentucky, Lexington Kentucky, 40506, United States
| |
Collapse
|
29
|
Biological functions controlled by manganese redox changes in mononuclear Mn-dependent enzymes. Essays Biochem 2017; 61:259-270. [PMID: 28487402 DOI: 10.1042/ebc20160070] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/05/2017] [Accepted: 03/17/2017] [Indexed: 02/06/2023]
Abstract
Remarkably few enzymes are known to employ a mononuclear manganese ion that undergoes changes in redox state during catalysis. Many questions remain to be answered about the role of substrate binding and/or protein environment in modulating the redox properties of enzyme-bound Mn(II), the nature of the dioxygen species involved in the catalytic mechanism, and how these enzymes acquire Mn(II) given that many other metal ions in the cell form more stable protein complexes. Here, we summarize current knowledge concerning the structure and mechanism of five mononuclear manganese-dependent enzymes: superoxide dismutase, oxalate oxidase (OxOx), oxalate decarboxylase (OxDC), homoprotocatechuate 3,4-dioxygenase, and lipoxygenase (LOX). Spectroscopic measurements and/or computational studies suggest that Mn(III)/Mn(II) are the catalytically active oxidation states of the metal, and the importance of 'second-shell' hydrogen bonding interactions with metal ligands has been demonstrated for a number of examples. The ability of these enzymes to modulate the redox properties of the Mn(III)/Mn(II) couple, thereby allowing them to generate substrate-based radicals, appears essential for accessing diverse chemistries of fundamental importance to organisms in all branches of life.
Collapse
|
30
|
Sensing the active site properties of enzymes as a function of the size of an effective peptidic environment using DFT reactivity parameters. Theor Chem Acc 2016. [DOI: 10.1007/s00214-016-1980-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
31
|
Demicheli V, Moreno DM, Jara GE, Lima A, Carballal S, Ríos N, Batthyany C, Ferrer-Sueta G, Quijano C, Estrı́n DA, Martí MA, Radi R. Mechanism of the Reaction of Human Manganese Superoxide Dismutase with Peroxynitrite: Nitration of Critical Tyrosine 34. Biochemistry 2016; 55:3403-17. [PMID: 27227512 DOI: 10.1021/acs.biochem.6b00045] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Human Mn-containing superoxide dismutase (hMnSOD) is a mitochondrial enzyme that metabolizes superoxide radical (O2(•-)). O2(•-) reacts at diffusional rates with nitric oxide to yield a potent nitrating species, peroxynitrite anion (ONOO(-)). MnSOD is nitrated and inactivated in vivo, with active site Tyr34 as the key oxidatively modified residue. We previously reported a k of ∼1.0 × 10(5) M(-1) s(-1) for the reaction of hMnSOD with ONOO(-) by direct stopped-flow spectroscopy and the critical role of Mn in the nitration process. In this study, we further established the mechanism of the reaction of hMnSOD with ONOO(-), including the necessary re-examination of the second-order rate constant by an independent method and the delineation of the microscopic steps that lead to the regio-specific nitration of Tyr34. The redetermination of k was performed by competition kinetics utilizing coumarin boronic acid, which reacts with ONOO(-) at a rate of ∼1 × 10(6) M(-1) s(-1) to yield the fluorescence product, 7-hydroxycoumarin. Time-resolved fluorescence studies in the presence of increasing concentrations of hMnSOD provided a k of ∼1.0 × 10(5) M(-1) s(-1), fully consistent with the direct method. Proteomic analysis indicated that ONOO(-), but not other nitrating agents, mediates the selective modification of active site Tyr34. Hybrid quantum-classical (quantum mechanics/molecular mechanics) simulations supported a series of steps that involve the initial reaction of ONOO(-) with Mn(III) to yield Mn(IV) and intermediates that ultimately culminate in 3-nitroTyr34. The data reported herein provide a kinetic and mechanistic basis for rationalizing how MnSOD constitutes an intramitochondrial target for ONOO(-) and the microscopic events, with atomic level resolution, that lead to selective and efficient nitration of critical Tyr34.
Collapse
Affiliation(s)
- Verónica Demicheli
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República , Avda. General Flores 2125, Montevideo, Uruguay.,Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República , Avda. General Flores 2125, Montevideo, Uruguay
| | - Diego M Moreno
- Instituto de Química de Rosario (IQUIR-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario , Suipacha 531, S2002LRK Rosario, Argentina
| | - Gabriel E Jara
- Departamento de Química Inorgánica, Analítica y Química-Física (INQUIMAE-CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria , Intendente Güiraldes 2160, C1428EGA Ciudad Autónoma de Buenos Aires, Argentina
| | - Analía Lima
- Unidad Bioquímica y Proteómica Analíticas, Institut Pasteur de Montevideo , Montevideo, Uruguay
| | - Sebastián Carballal
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República , Avda. General Flores 2125, Montevideo, Uruguay.,Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República , Avda. General Flores 2125, Montevideo, Uruguay
| | - Natalia Ríos
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República , Avda. General Flores 2125, Montevideo, Uruguay.,Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República , Avda. General Flores 2125, Montevideo, Uruguay.,Departamento de Química Orgánica, Facultad de Química, Universidad de la República , Avda. General Flores 2124, Montevideo, Uruguay
| | - Carlos Batthyany
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República , Avda. General Flores 2125, Montevideo, Uruguay.,Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República , Avda. General Flores 2125, Montevideo, Uruguay.,Unidad Bioquímica y Proteómica Analíticas, Institut Pasteur de Montevideo , Montevideo, Uruguay
| | - Gerardo Ferrer-Sueta
- Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República , Avda. General Flores 2125, Montevideo, Uruguay.,Laboratorio de Fisicoquímica Biológica, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la Repúbica , Igua 4225, Montevideo, Uruguay
| | - Celia Quijano
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República , Avda. General Flores 2125, Montevideo, Uruguay.,Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República , Avda. General Flores 2125, Montevideo, Uruguay
| | - Darío A Estrı́n
- Departamento de Química Inorgánica, Analítica y Química-Física (INQUIMAE-CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria , Intendente Güiraldes 2160, C1428EGA Ciudad Autónoma de Buenos Aires, Argentina
| | - Marcelo A Martí
- Departamento de Química Biológica e IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria , Intendente Güiraldes 2160, C1428EGA Ciudad Autónoma de Buenos Aires, Argentina
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República , Avda. General Flores 2125, Montevideo, Uruguay.,Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República , Avda. General Flores 2125, Montevideo, Uruguay
| |
Collapse
|
32
|
Porporato PE, Payen VL, Baselet B, Sonveaux P. Metabolic changes associated with tumor metastasis, part 2: Mitochondria, lipid and amino acid metabolism. Cell Mol Life Sci 2016; 73:1349-63. [PMID: 26646069 PMCID: PMC11108268 DOI: 10.1007/s00018-015-2100-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/16/2015] [Accepted: 11/23/2015] [Indexed: 12/13/2022]
Abstract
Metabolic alterations are a hallmark of cancer controlling tumor progression and metastasis. Among the various metabolic phenotypes encountered in tumors, this review focuses on the contributions of mitochondria, lipid and amino acid metabolism to the metastatic process. Tumor cells require functional mitochondria to grow, proliferate and metastasize, but shifts in mitochondrial activities confer pro-metastatic traits encompassing increased production of mitochondrial reactive oxygen species (mtROS), enhanced resistance to apoptosis and the increased or de novo production of metabolic intermediates of the TCA cycle behaving as oncometabolites, including succinate, fumarate, and D-2-hydroxyglutarate that control energy production, biosynthesis and the redox state. Lipid metabolism and the metabolism of amino acids, such as glutamine, glutamate and proline are also currently emerging as focal control points of cancer metastasis.
Collapse
Affiliation(s)
- Paolo E Porporato
- Pole of Pharmacology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 52, box B1.53.09, 1200, Brussels, Belgium
| | - Valéry L Payen
- Pole of Pharmacology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 52, box B1.53.09, 1200, Brussels, Belgium
| | - Bjorn Baselet
- Pole of Pharmacology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 52, box B1.53.09, 1200, Brussels, Belgium
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK·CEN, 2400 Mol, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 52, box B1.53.09, 1200, Brussels, Belgium.
| |
Collapse
|
33
|
Cheng AJ, Yamada T, Rassier DE, Andersson DC, Westerblad H, Lanner JT. Reactive oxygen/nitrogen species and contractile function in skeletal muscle during fatigue and recovery. J Physiol 2016; 594:5149-60. [PMID: 26857536 DOI: 10.1113/jp270650] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/23/2015] [Indexed: 01/17/2023] Open
Abstract
The production of reactive oxygen/nitrogen species (ROS/RNS) is generally considered to increase during physical exercise. Nevertheless, direct measurements of ROS/RNS often show modest increases in ROS/RNS in muscle fibres even during intensive fatiguing stimulation, and the major source(s) of ROS/RNS during exercise is still being debated. In rested muscle fibres, mild and acute exposure to exogenous ROS/RNS generally increases myofibrillar submaximal force, whereas stronger or prolonged exposure has the opposite effect. Endogenous production of ROS/RNS seems to preferentially decrease submaximal force and positive effects of antioxidants are mainly observed during fatigue induced by submaximal contractions. Fatigued muscle fibres frequently enter a prolonged state of reduced submaximal force, which is caused by a ROS/RNS-dependent decrease in sarcoplasmic reticulum Ca(2+) release and/or myofibrillar Ca(2+) sensitivity. Increased ROS/RNS production during exercise can also be beneficial and recent human and animal studies show that antioxidant supplementation can hamper the beneficial effects of endurance training. In conclusion, increased ROS/RNS production have both beneficial and detrimental effects on skeletal muscle function and the outcome depends on a combination of factors: the type of ROS/RNS; the magnitude, duration and location of ROS/RNS production; and the defence systems, including both endogenous and exogenous antioxidants.
Collapse
Affiliation(s)
| | | | - Dilson E Rassier
- McGill University, 475 Pine Avenue West, Montreal, QC, Canada, H2W1S4
| | | | | | | |
Collapse
|
34
|
Interplay between oxidant species and energy metabolism. Redox Biol 2015; 8:28-42. [PMID: 26741399 PMCID: PMC4710798 DOI: 10.1016/j.redox.2015.11.010] [Citation(s) in RCA: 217] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/20/2015] [Accepted: 11/25/2015] [Indexed: 02/07/2023] Open
Abstract
It has long been recognized that energy metabolism is linked to the production of reactive oxygen species (ROS) and critical enzymes allied to metabolic pathways can be affected by redox reactions. This interplay between energy metabolism and ROS becomes most apparent during the aging process and in the onset and progression of many age-related diseases (i.e. diabetes, metabolic syndrome, atherosclerosis, neurodegenerative diseases). As such, the capacity to identify metabolic pathways involved in ROS formation, as well as specific targets and oxidative modifications is crucial to our understanding of the molecular basis of age-related diseases and for the design of novel therapeutic strategies. Herein we review oxidant formation associated with the cell's energetic metabolism, key antioxidants involved in ROS detoxification, and the principal targets of oxidant species in metabolic routes and discuss their relevance in cell signaling and age-related diseases. Energy metabolism is both a source and target of oxidant species. Reactive oxygen species are formed in redox reactions in catabolic pathways. Sensitive targets of oxidant species regulate the flux of metabolic pathways. Metabolic pathways and antioxidant systems are regulated coordinately.
Collapse
|
35
|
Hunter GJ, Trinh CH, Bonetta R, Stewart EE, Cabelli DE, Hunter T. The structure of the Caenorhabditis elegans manganese superoxide dismutase MnSOD-3-azide complex. Protein Sci 2015; 24:1777-88. [PMID: 26257399 PMCID: PMC4622211 DOI: 10.1002/pro.2768] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 08/03/2015] [Indexed: 01/18/2023]
Abstract
C. elegans MnSOD-3 has been implicated in the longevity pathway and its mechanism of catalysis is relevant to the aging process and carcinogenesis. The structures of MnSOD-3 provide unique crystallographic evidence of a dynamic region of the tetrameric interface (residues 41-54). We have determined the structure of the MnSOD-3-azide complex to 1.77-Å resolution. Analysis of this complex shows that the substrate analog, azide, binds end-on to the manganese center as a sixth ligand and that it ligates directly to a third and new solvent molecule also positioned within interacting distance to the His30 and Tyr34 residues of the substrate access funnel. This is the first structure of a eukaryotic MnSOD-azide complex that demonstrates the extended, uninterrupted hydrogen-bonded network that forms a proton relay incorporating three outer sphere solvent molecules, the substrate analog, the gateway residues, Gln142, and the solvent ligand. This configuration supports the formation and release of the hydrogen peroxide product in agreement with the 5-6-5 catalytic mechanism for MnSOD. The high product dissociation constant k4 of MnSOD-3 reflects low product inhibition making this enzyme efficient even at high levels of superoxide.
Collapse
Affiliation(s)
- Gary J Hunter
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of MaltaMsida, Malta
| | - Chi H Trinh
- Astbury Centre for Structural Molecular Biology, Institute of Molecular and Cellular Biology, University of LeedsLeeds, United Kingdom
| | - Rosalin Bonetta
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of MaltaMsida, Malta
| | - Emma E Stewart
- Astbury Centre for Structural Molecular Biology, Institute of Molecular and Cellular Biology, University of LeedsLeeds, United Kingdom
| | - Diane E Cabelli
- Chemistry Department, Brookhaven National LaboratoryUpton, New York
| | - Therese Hunter
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of MaltaMsida, Malta
| |
Collapse
|
36
|
Rashid GMM, Taylor CR, Liu Y, Zhang X, Rea D, Fülöp V, Bugg TDH. Identification of Manganese Superoxide Dismutase from Sphingobacterium sp. T2 as a Novel Bacterial Enzyme for Lignin Oxidation. ACS Chem Biol 2015. [PMID: 26198187 DOI: 10.1021/acschembio.5b00298] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The valorization of aromatic heteropolymer lignin is an important unsolved problem in the development of a biomass-based biorefinery, for which novel high-activity biocatalysts are needed. Sequencing of the genomic DNA of lignin-degrading bacterial strain Sphingobacterium sp. T2 revealed no matches to known lignin-degrading genes. Proteomic matches for two manganese superoxide dismutase proteins were found in partially purified extracellular fractions. Recombinant MnSOD1 and MnSOD2 were both found to show high activity for oxidation of Organosolv and Kraft lignin, and lignin model compounds, generating multiple oxidation products. Structure determination revealed that the products result from aryl-Cα and Cα-Cβ bond oxidative cleavage and O-demethylation. The crystal structure of MnSOD1 was determined to 1.35 Å resolution, revealing a typical MnSOD homodimer harboring a five-coordinate trigonal bipyramidal Mn(II) center ligated by three His, one Asp, and a water/hydroxide in each active site. We propose that the lignin oxidation reactivity of these enzymes is due to the production of a hydroxyl radical, a highly reactive oxidant. This is the first demonstration that MnSOD is a microbial lignin-oxidizing enzyme.
Collapse
Affiliation(s)
- Goran M. M. Rashid
- Department of Chemistry and ‡School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Charles R. Taylor
- Department of Chemistry and ‡School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Yangqingxue Liu
- Department of Chemistry and ‡School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Xiaoyang Zhang
- Department of Chemistry and ‡School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Dean Rea
- Department of Chemistry and ‡School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Vilmos Fülöp
- Department of Chemistry and ‡School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Timothy D. H. Bugg
- Department of Chemistry and ‡School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| |
Collapse
|
37
|
Markevich NI, Hoek JB. Computational modeling analysis of mitochondrial superoxide production under varying substrate conditions and upon inhibition of different segments of the electron transport chain. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1847:656-79. [PMID: 25868872 PMCID: PMC4426091 DOI: 10.1016/j.bbabio.2015.04.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 03/24/2015] [Accepted: 04/06/2015] [Indexed: 12/13/2022]
Abstract
A computational mechanistic model of superoxide (O2•-) formation in the mitochondrial electron transport chain (ETC) was developed to facilitate the quantitative analysis of factors controlling mitochondrial O2•- production and assist in the interpretation of experimental studies. The model takes into account all individual electron transfer reactions in Complexes I and III. The model accounts for multiple, often seemingly contradictory observations on the effects of ΔΨ and ΔpH, and for the effects of multiple substrate and inhibitor conditions, including differential effects of Complex III inhibitors antimycin A, myxothiazol and stigmatellin. Simulation results confirm that, in addition to O2•- formation in Complex III and at the flavin site of Complex I, the quinone binding site of Complex I is an additional superoxide generating site that accounts for experimental observations on O2•- production during reverse electron transfer. However, our simulation results predict that, when cytochrome c oxidase is inhibited during oxidation of succinate, ROS production at this site is eliminated and almost all superoxide in Complex I is generated by reduced FMN, even when the redox pressure for reverse electron transfer from succinate is strong. In addition, the model indicates that conflicting literature data on the kinetics of electron transfer in Complex III involving the iron-sulfur protein-cytochrome bL complex can be resolved in favor of a dissociation of the protein only after electron transfer to cytochrome bH. The model predictions can be helpful in understanding factors driving mitochondrial superoxide formation in intact cells and tissues.
Collapse
Affiliation(s)
- Nikolai I Markevich
- MitoCare Center for Mitochondrial Research, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 14290, Russia.
| | - Jan B Hoek
- MitoCare Center for Mitochondrial Research, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
38
|
Sakai T, Sato B, Hara K, Hara Y, Naritomi Y, Koyanagi S, Hara H, Nagao T, Ishibashi T. Consumption of water containing over 3.5 mg of dissolved hydrogen could improve vascular endothelial function. Vasc Health Risk Manag 2014; 10:591-7. [PMID: 25378931 PMCID: PMC4207582 DOI: 10.2147/vhrm.s68844] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The redox imbalance between nitric oxide and superoxide generated in the endothelium is thought to play a pivotal role in the development of endothelial dysfunction. A third reactive oxygen species (ROS), H2O2, is known to have both beneficial and detrimental effects on the vasculature. Nonetheless, the influence of the hydroxyl radical, a byproduct of H2O2 decay, is unclear, and there is no direct evidence that the hydroxyl radical impairs endothelial function in conduit arteries. Molecular hydrogen (H2) neutralizes detrimental ROS, especially the hydroxyl radical. OBJECTIVES To assess the influence of the hydroxyl radical on the endothelium and to confirm that a gaseous antioxidant, H2, can be a useful modulator of blood vessel function. METHODS The efficacy of water containing a high concentration of H2 was tested by measuring flow-mediated dilation (FMD) of the brachial artery (BA). The subjects were randomly divided into two groups: the high-H2 group, who drank high-H2 water containing 7 ppm H2 (3.5 mg H2 in 500 mL water); and the placebo group. Endothelial function was evaluated by measuring the FMD of the BA. After measurement of diameter of the BA and FMD at baseline, volunteers drank the high-H2 water or placebo water immediately and with a 30-minute interval; FMD was compared to baseline. RESULTS FMD increased in the high-H2 group (eight males; eight females) from 6.80%±1.96% to 7.64%±1.68% (mean ± standard deviation) and decreased from 8.07%±2.41% to 6.87%±2.94% in the placebo group (ten males; eight females). The ratio to the baseline in the changes of FMD showed significant improvement (P<0.05) in the high-H2 group compared to the placebo group. CONCLUSION H2 may protect the vasculature from shear stress-derived detrimental ROS, such as the hydroxyl radical, by maintaining the nitric oxide-mediated vasomotor response.
Collapse
Affiliation(s)
- Takaaki Sakai
- Department of Cardiology, Haradoi Hospital, Fukuoka, Japan
| | - Bunpei Sato
- MiZ Company Limited, Fujisawa, Kanagawa, Japan
| | - Koji Hara
- Department of Internal Medicine, Haradoi Hospital, Fukuoka, Japan
| | - Yuichi Hara
- Department of Internal Medicine, Haradoi Hospital, Fukuoka, Japan
| | - Yuji Naritomi
- Department of Internal Medicine, Haradoi Hospital, Fukuoka, Japan
| | - Samon Koyanagi
- Department of Cardiology, Haradoi Hospital, Fukuoka, Japan
| | - Hiroshi Hara
- Department of Internal Medicine, Haradoi Hospital, Fukuoka, Japan
| | | | - Toru Ishibashi
- Department of Rheumatology and Orthopedic Surgery, Haradoi Hospital, Fukuoka, Japan
| |
Collapse
|
39
|
Kembro JM, Cortassa S, Aon MA. Complex oscillatory redox dynamics with signaling potential at the edge between normal and pathological mitochondrial function. Front Physiol 2014; 5:257. [PMID: 25071602 PMCID: PMC4085651 DOI: 10.3389/fphys.2014.00257] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 06/17/2014] [Indexed: 12/31/2022] Open
Abstract
The time-keeping properties bestowed by oscillatory behavior on functional rhythms represent an evolutionarily conserved trait in living systems. Mitochondrial networks function as timekeepers maximizing energetic output while tuning reactive oxygen species (ROS) within physiological levels compatible with signaling. In this work, we explore the potential for timekeeping functions dependent on mitochondrial dynamics with the validated two-compartment mitochondrial energetic-redox (ME-R) computational model, that takes into account (a) four main redox couples [NADH, NADPH, GSH, Trx(SH)2], (b) scavenging systems (glutathione, thioredoxin, SOD, catalase) distributed in matrix and extra-matrix compartments, and (c) transport of ROS species between them. Herein, we describe that the ME-R model can exhibit highly complex oscillatory dynamics in energetic/redox variables and ROS species, consisting of at least five frequencies with modulated amplitudes and period according to power spectral analysis. By stability analysis we describe that the extent of steady state—as against complex oscillatory behavior—was dependent upon the abundance of Mn and Cu, Zn SODs, and their interplay with ROS production in the respiratory chain. Large parametric regions corresponding to oscillatory dynamics of increasingly complex waveforms were obtained at low Cu, Zn SOD concentration as a function of Mn SOD. This oscillatory domain was greatly reduced at higher levels of Cu, Zn SOD. Interestingly, the realm of complex oscillations was located at the edge between normal and pathological mitochondrial energetic behavior, and was characterized by oxidative stress. We conclude that complex oscillatory dynamics could represent a frequency- and amplitude-modulated H2O2 signaling mechanism that arises under intense oxidative stress. By modulating SOD, cells could have evolved an adaptive compromise between relative constancy and the flexibility required under stressful redox/energetic conditions.
Collapse
Affiliation(s)
- Jackelyn M Kembro
- Facultad de Ciencias Exactas, Físicas y Naturales, Instituto de Investigaciones Biológicas y Tecnológicas (Consejo Nacional de Investigaciones Científicas y Técnicas-UNC) and Instituto de Ciencia y Tecnología de los Alimentos, Universidad Nacional de Córdoba Córdoba, Argentina
| | - Sonia Cortassa
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Miguel A Aon
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine Baltimore, MD, USA
| |
Collapse
|
40
|
Sheng Y, Abreu IA, Cabelli DE, Maroney MJ, Miller AF, Teixeira M, Valentine JS. Superoxide dismutases and superoxide reductases. Chem Rev 2014; 114:3854-918. [PMID: 24684599 PMCID: PMC4317059 DOI: 10.1021/cr4005296] [Citation(s) in RCA: 659] [Impact Index Per Article: 59.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Indexed: 11/30/2022]
Affiliation(s)
- Yuewei Sheng
- Department
of Chemistry and Biochemistry, University
of California Los Angeles, Los
Angeles, California 90095, United States
| | - Isabel A. Abreu
- Instituto
de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
- Instituto
de Biologia Experimental e Tecnológica, Av. da República,
Qta. do Marquês, Estação Agronómica Nacional,
Edificio IBET/ITQB, 2780-157, Oeiras, Portugal
| | - Diane E. Cabelli
- Chemistry
Department, Brookhaven National Laboratory, Upton, New York 11973, United States
| | - Michael J. Maroney
- Department
of Chemistry, University of Massachusetts
Amherst, Amherst, Massachusetts 01003, United States
| | - Anne-Frances Miller
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506-0055, United States
| | - Miguel Teixeira
- Instituto
de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - Joan Selverstone Valentine
- Department
of Chemistry and Biochemistry, University
of California Los Angeles, Los
Angeles, California 90095, United States
- Department
of Bioinspired Sciences, Ewha Womans University, Seoul 120-750, Republic of Korea
| |
Collapse
|
41
|
|
42
|
Boone CD, Pinard M, McKenna R, Silverman D. Catalytic mechanism of α-class carbonic anhydrases: CO2 hydration and proton transfer. Subcell Biochem 2014; 75:31-52. [PMID: 24146373 DOI: 10.1007/978-94-007-7359-2_3] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The carbonic anhydrases (CAs; EC 4.2.1.1) are a family of metalloenzymes that catalyze the reversible hydration of carbon dioxide (CO2) and dehydration of bicarbonate (HCO3 (-)) in a two-step ping-pong mechanism: [Formula: see text] CAs are ubiquitous enzymes and are categorized into five distinct classes (α, β, γ, δ and ζ). The α-class is found primarily in vertebrates (and the only class of CA in mammals), β is observed in higher plants and some prokaryotes, γ is present only in archaebacteria whereas the δ and ζ classes have only been observed in diatoms.The focus of this chapter is on α-CAs as the structure-function relationship is best understood for this class, in particular for humans. The reader is referred to other reviews for an overview of the structure and catalytic mechanism of the other CA classes. The overall catalytic site structure and geometry of α-CAs are described in the first section of this chapter followed by the kinetic studies, binding of CO2, and the proton shuttle network.
Collapse
Affiliation(s)
- Christopher D Boone
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA,
| | | | | | | |
Collapse
|
43
|
Peroxomanganese complexes as an aid to understanding redox-active manganese enzymes. J Biol Inorg Chem 2013; 19:1-15. [DOI: 10.1007/s00775-013-1067-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 11/07/2013] [Indexed: 02/07/2023]
|
44
|
Ji G, Lv K, Chen H, Wang T, Wang Y, Zhao D, Qu L, Li Y. MiR-146a regulates SOD2 expression in H2O2 stimulated PC12 cells. PLoS One 2013; 8:e69351. [PMID: 23935993 PMCID: PMC3720842 DOI: 10.1371/journal.pone.0069351] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 06/07/2013] [Indexed: 11/19/2022] Open
Abstract
SOD2 (superoxide dismutase 2) is one of the endogenous antioxidant enzymes that protect against reactive oxygen species. While explorations of SOD2 expression regulation are mainly focused on transcriptional and post-translational activation, there are few reports about the post-transcriptional regulation of SOD2. MicroRNAs (miRNAs) are 21nt-25nt (nucleotide) small noncoding RNAs that have emerged as indispensable regulators of gene expression. Here we show that miR-146a, a widely expressed miRNA, is up-regulated by H2O2-induced stress. By sequence analysis we found a binding site for miR-146a in the sod2 mRNA 3'UTR, and a luciferase reporter assay confirmed that miR-146a can interact with this sod2 regulatory region. Our results further show that miR-146a could down-regulate the SOD2 protein expression, and antisense-miR-146a could reverse the decrease of both the SOD2 level and cell viability in H2O2 treated PC12 cells. In conclusion, here we have identified a novel function of miR-146a in the post-transcriptional regulation of SOD2 expression.
Collapse
Affiliation(s)
- Guohua Ji
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Ke Lv
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Hailong Chen
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Tingmei Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yanli Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Dingsheng Zhao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Lina Qu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yinghui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
45
|
Carballal S, Bartesaghi S, Radi R. Kinetic and mechanistic considerations to assess the biological fate of peroxynitrite. Biochim Biophys Acta Gen Subj 2013; 1840:768-80. [PMID: 23872352 DOI: 10.1016/j.bbagen.2013.07.005] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 06/25/2013] [Accepted: 07/04/2013] [Indexed: 01/21/2023]
Abstract
BACKGROUND Peroxynitrite, the product of the reaction between superoxide radicals and nitric oxide, is an elusive oxidant with a short half-life and a low steady-state concentration in biological systems; it promotes nitroxidative damage. SCOPE OF REVIEW We will consider kinetic and mechanistic aspects that allow rationalizing the biological fate of peroxynitrite from data obtained by a combination of methods that include fast kinetic techniques, electron paramagnetic resonance and kinetic simulations. In addition, we provide a quantitative analysis of peroxynitrite production rates and conceivable steady-state levels in living systems. MAJOR CONCLUSIONS The preferential reactions of peroxynitrite in vivo include those with carbon dioxide, thiols and metalloproteins; its homolysis represents only <1% of its fate. To note, carbon dioxide accounts for a significant fraction of peroxynitrite consumption leading to the formation of strong one-electron oxidants, carbonate radicals and nitrogen dioxide. On the other hand, peroxynitrite is rapidly reduced by peroxiredoxins, which represent efficient thiol-based peroxynitrite detoxification systems. Glutathione, present at mM concentration in cells and frequently considered a direct scavenger of peroxynitrite, does not react sufficiently fast with it in vivo; glutathione mainly inhibits peroxynitrite-dependent processes by reactions with secondary radicals. The detection of protein 3-nitrotyrosine, a molecular footprint, can demonstrate peroxynitrite formation in vivo. Basal peroxynitrite formation rates in cells can be estimated in the order of 0.1 to 0.5μMs(-1) and its steady-state concentration at ~1nM. GENERAL SIGNIFICANCE The analysis provides a handle to predict the preferential fate and steady-state levels of peroxynitrite in living systems. This is useful to understand pathophysiological aspects and pharmacological prospects connected to peroxynitrite. This article is part of a Special Issue entitled Current methods to study reactive oxygen species - pros and cons and biophysics of membrane proteins. Guest Editor: Christine Winterbourn.
Collapse
Affiliation(s)
- Sebastián Carballal
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | | | | |
Collapse
|
46
|
Paulsen C, Carroll KS. Cysteine-mediated redox signaling: chemistry, biology, and tools for discovery. Chem Rev 2013; 113:4633-79. [PMID: 23514336 PMCID: PMC4303468 DOI: 10.1021/cr300163e] [Citation(s) in RCA: 864] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Indexed: 02/06/2023]
Affiliation(s)
- Candice
E. Paulsen
- Department of Chemistry, The Scripps Research
Institute, Jupiter, Florida, 33458, United States
| | - Kate S. Carroll
- Department of Chemistry, The Scripps Research
Institute, Jupiter, Florida, 33458, United States
| |
Collapse
|
47
|
Abstract
Protein kinases represent one of the largest families of genes found in eukaryotes. Kinases mediate distinct cellular processes ranging from proliferation, differentiation, survival, and apoptosis. Ligand-mediated activation of receptor kinases can lead to the production of endogenous hydrogen peroxide (H₂O₂) by membrane-bound NADPH oxidases. In turn, H₂O₂ can be utilized as a secondary messenger in signal transduction pathways. This review presents an overview of the molecular mechanisms involved in redox regulation of protein kinases and its effects on signaling cascades. In the first half, we will focus primarily on receptor tyrosine kinases (RTKs), whereas the latter will concentrate on downstream non-receptor kinases involved in relaying stimulant response. Select examples from the literature are used to highlight the functional role of H₂O₂ regarding kinase activity, as well as the components involved in H₂O₂ production and regulation during cellular signaling. In addition, studies demonstrating direct modulation of protein kinases by H₂O₂ through cysteine oxidation will be emphasized. Identification of these redox-sensitive residues may help uncover signaling mechanisms conserved within kinase subfamilies. In some cases, these residues can even be exploited as targets for the development of new therapeutics. Continued efforts in this field will further basic understanding of kinase redox regulation, and delineate the mechanisms involved in physiological and pathological H₂O₂ responses.
Collapse
Affiliation(s)
- Thu H Truong
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
48
|
Jackson TA, Gutman CT, Maliekal J, Miller AF, Brunold TC. Geometric and electronic structures of manganese-substituted iron superoxide dismutase. Inorg Chem 2013; 52:3356-67. [PMID: 23461587 PMCID: PMC3974275 DOI: 10.1021/ic302867y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The active-site structures of the oxidized and reduced forms of manganese-substituted iron superoxide dismutase (Mn(Fe)SOD) are examined, for the first time, using a combination of spectroscopic and computational methods. On the basis of electronic absorption, circular dichroism (CD), magnetic CD (MCD), and variable-temperature variable-field MCD data obtained for oxidized Mn(Fe)SOD, we propose that the active site of this species is virtually identical to that of wild-type manganese SOD (MnSOD), with both containing a metal ion that resides in a trigonal bipyramidal ligand environment. This proposal is corroborated by quantum mechanical/molecular mechanical (QM/MM) computations performed on complete protein models of Mn(Fe)SOD in both its oxidized and reduced states and, for comparison, wild-type (WT) MnSOD. The major differences between the QM/MM optimized active sites of WT MnSOD and Mn(Fe)SOD are a smaller (His)N-Mn-N(His) equatorial angle and a longer (Gln146(69))NH···O(sol) H-bond distance in the metal-substituted protein. Importantly, these modest geometric differences are consistent with our spectroscopic data obtained for the oxidized proteins and high-field electron paramagnetic resonance spectra reported previously for reduced Mn(Fe)SOD and MnSOD. As Mn(Fe)SOD exhibits a reduction midpoint potential (E(m)) almost 700 mV higher than that of MnSOD, which has been shown to be sufficient for explaining the lack of SOD activity displayed by the metal-subtituted species (Vance, C. K.; Miller, A. F. Biochemistry 2001, 40, 13079-13087), E(m)'s were computed for our experimentally validated QM/MM optimized models of Mn(Fe)SOD and MnSOD. These computations properly reproduce the experimental trend and reveal that the drastically elevated E(m) of the metal substituted protein stems from a larger separation between the second-sphere Gln residue and the coordinated solvent in Mn(Fe)SOD relative to MnSOD, which causes a weakening of the corresponding H-bond interaction in the oxidized state and alleviates steric crowding in the reduced state.
Collapse
Affiliation(s)
| | | | - James Maliekal
- Department of Chemistry, University of Kentucky, Lexington, KY 40506
| | | | - Thomas C. Brunold
- To whom correspondence should be addressed: 1101 University Ave., Madison, WI 53706, phone: (608) 265-9056, fax: (608) 262-6143,
| |
Collapse
|
49
|
Rawat SJ, Creasy CL, Peterson JR, Chernoff J. The tumor suppressor Mst1 promotes changes in the cellular redox state by phosphorylation and inactivation of peroxiredoxin-1 protein. J Biol Chem 2013; 288:8762-8771. [PMID: 23386615 DOI: 10.1074/jbc.m112.414524] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The serine/threonine protein kinases Mst1 and Mst2 can be activated by cellular stressors including hydrogen peroxide. Using two independent protein interaction screens, we show that these kinases associate, in an oxidation-dependent manner, with Prdx1, an enzyme that regulates the cellular redox state by reducing hydrogen peroxide to water and oxygen. Mst1 inactivates Prdx1 by phosphorylating it at Thr-90 and Thr-183, leading to accumulation of hydrogen peroxide in cells. These results suggest that hydrogen peroxide-stimulated Mst1 activates a positive feedback loop to sustain an oxidizing cellular state.
Collapse
Affiliation(s)
- Sonali Jalan Rawat
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111; Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| | - Caretha L Creasy
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| | - Jeffrey R Peterson
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| | - Jonathan Chernoff
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111.
| |
Collapse
|
50
|
Macholl S, Tietze D, Buntkowsky G. NMR crystallography of amides, peptides and protein–ligand complexes. CrystEngComm 2013. [DOI: 10.1039/c3ce40908b] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|