1
|
Devarakonda SS, Basha S, Pithakumar A, L B T, Mukunda DC, Rodrigues J, K A, Biswas S, Pai AR, Belurkar S, Mahato KK. Molecular mechanisms of neurofilament alterations and its application in assessing neurodegenerative disorders. Ageing Res Rev 2024; 102:102566. [PMID: 39481763 DOI: 10.1016/j.arr.2024.102566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/04/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Neurofilaments are intermediate filaments present in neurons. These provide structural support and maintain the size and shape of the neurons. Dysregulation, mutation, and aggregation of neurofilaments raise the levels of these proteins in the blood and cerebrospinal fluid (CSF), which are characteristic features of axonal damage and certain rare neurological diseases, such as Giant Axonal Neuropathy and Charcot-Mare-Tooth disease. Understanding the structure, dynamics, and function of neurofilaments has been greatly enhanced by a diverse range of biochemical and preclinical investigations conducted over more than four decades. Recently, there has been a resurgence of interest in post-translational modifications of neurofilaments, such as phosphorylation, aggregation, mutation, oxidation, etc. Over the past twenty years, several rare disorders have been studied from structural alterations of neurofilaments. These disorders are monitored by fluid biomarkers such as neurofilament light chains. Currently, there are many tools, such as Enzyme-Linked Immunosorbent Assay, Electrochemiluminescence Assay, Single-Molecule Array, Western/immunoblotting, etc., in use to assess the neurofilament proteins in Blood and CSF. However, all these techniques utilize expensive, non-specific, or antibody-based methods, which make them unsuitable for routine screening of neurodegenerative disorders. This provides room to search for newer sensitive, cost-effective, point-of-care tools for rapid screening of the disease. For a long time, the molecular mechanisms of neurofilaments have been poorly understood due to insufficient research attempts, and a deeper understanding of them remains elusive. Therefore, this review aims to highlight the available literature on molecular mechanisms of neurofilaments and the function of neurofilaments in axonal transport, axonal conduction, axonal growth, and neurofilament aggregation, respectively. Further, this review discusses the role of neurofilaments as potential biomarkers for the identification of several neurodegenerative diseases in clinical laboratory practice.
Collapse
Affiliation(s)
| | - Shaik Basha
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Anjana Pithakumar
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Thoshna L B
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | | | - Jackson Rodrigues
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Ameera K
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Shimul Biswas
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Aparna Ramakrishna Pai
- Department of Neurology, Kasturba Medical College-Manipal, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Sushma Belurkar
- Department of Pathology, Kasturba Medical College-Manipal, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Krishna Kishore Mahato
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India.
| |
Collapse
|
2
|
Lv J, Chen F, Zhang C, Kang Y, Yang Y, Zhang C. Role of Peroxynitrite in the Pathogenesis of Parkinson's Disease and Its Fluorescence Imaging-Based Detection. BIOSENSORS 2024; 14:506. [PMID: 39451719 PMCID: PMC11506598 DOI: 10.3390/bios14100506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder, affecting the lives of millions of people worldwide. Although the mechanism underlying PD pathogenesis is largely undefined, increasing evidence indicates that oxidative and nitrosative stresses play a crucial role in PD occurrence and development. Among them, the role of oxidative stress has been widely acknowledged, but there is relatively less attention given to nitrosative stress, which is mainly derived from peroxynitrite. In the present review, after briefly introducing the background of PD, we discuss the physiopathological function of peroxynitrite and especially highlight how overloaded peroxynitrite is involved in PD pathogenesis. Then, we summarize the currently reported fluorescence imaging-based peroxynitrite detection probes. Moreover, we specifically emphasize the probes that have been applied in PD research. Finally, we propose perspectives on how to develop a more applicable peroxynitrite probe and leverage it for PD theranostics. Conclusively, the present review broadens the knowledge on the pathological role of peroxynitrite in the context of PD and sheds light on how to develop and utilize fluorescence imaging-based strategies for peroxynitrite detection.
Collapse
Affiliation(s)
- Jiye Lv
- School of Basic Medical Sciences, Shanxi Medical University, 56 Xinjiannan Road, Taiyuan 030001, China
| | - Feiyu Chen
- School of Chinese Medicine, Tianjin University of Traditional Medicine, 10 Poyanghu Road, Tianjin 301617, China
| | - Changchan Zhang
- School of Basic Medical Sciences, Shanxi Medical University, 56 Xinjiannan Road, Taiyuan 030001, China
| | - Yubing Kang
- School of Basic Medical Sciences, Shanxi Medical University, 56 Xinjiannan Road, Taiyuan 030001, China
| | - Yan Yang
- School of Basic Medical Sciences, Shanxi Medical University, 56 Xinjiannan Road, Taiyuan 030001, China
| | - Chengwu Zhang
- School of Basic Medical Sciences, Shanxi Medical University, 56 Xinjiannan Road, Taiyuan 030001, China
| |
Collapse
|
3
|
Lescouzères L, Patten SA. Promising animal models for amyotrophic lateral sclerosis drug discovery: a comprehensive update. Expert Opin Drug Discov 2024; 19:1213-1233. [PMID: 39115327 DOI: 10.1080/17460441.2024.2387791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/30/2024] [Indexed: 10/12/2024]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of motor neurons. Several animal models have been generated to understand ALS pathogenesis. They have provided valuable insight into disease mechanisms and the development of therapeutic strategies. AREAS COVERED In this review, the authors provide a concise overview of simple genetic model organisms, including C. elegans, Drosophila, zebrafish, and mouse genetic models that have been generated to study ALS. They emphasize the benefits of each model and their application in translational research for discovering new chemicals, gene therapy approaches, and antibody-based strategies for treating ALS. EXPERT OPINION Significant progress is being made in identifying new therapeutic targets for ALS. This progress is being enabled by promising animal models of the disease using increasingly effective genetic and pharmacological strategies. There are still challenges to be overcome in order to achieve improved success rates for translating drugs from animal models to clinics for treating ALS. Several promising future directions include the establishment of novel preclinical protocol standards, as well as the combination of animal models with human induced pluripotent stem cells (iPSCs).
Collapse
Affiliation(s)
- Léa Lescouzères
- INRS - Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada
| | - Shunmoogum A Patten
- INRS - Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
- Departement de Neurosciences, Université de Montréal, Montreal, Canada
| |
Collapse
|
4
|
Jia W, Gong X, Ye Z, Li N, Zhan X. Nitroproteomics is instrumental for stratification and targeted treatments of astrocytoma patients: expert recommendations for advanced 3PM approach with improved individual outcomes. EPMA J 2023; 14:673-696. [PMID: 38094577 PMCID: PMC10713973 DOI: 10.1007/s13167-023-00348-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/11/2023] [Indexed: 12/05/2024]
Abstract
Protein tyrosine nitration is a selectively and reversible important post-translational modification, which is closely related to oxidative stress. Astrocytoma is the most common neuroepithelial tumor with heterogeneity and complexity. In the past, the diagnosis of astrocytoma was based on the histological and clinical features, and the treatment methods were nothing more than surgery-assisted radiotherapy and chemotherapy. Obviously, traditional methods short falls an effective treatment for astrocytoma. In late 2021, the World Health Organization (WHO) adopted molecular biomarkers in the comprehensive diagnosis of astrocytoma, such as IDH-mutant and DNA methylation, which enabled the risk stratification, classification, and clinical prognosis prediction of astrocytoma to be more correct. Protein tyrosine nitration is closely related to the pathogenesis of astrocytoma. We hypothesize that nitroproteome is significantly different in astrocytoma relative to controls, which leads to establishment of nitroprotein biomarkers for patient stratification, diagnostics, and prediction of disease stages and severity grade, targeted prevention in secondary care, treatment algorithms tailored to individualized patient profile in the framework of predictive, preventive, and personalized medicine (PPPM; 3P medicine). Nitroproteomics based on gel electrophoresis and tandem mass spectrometry is an effective tool to identify the nitroproteins and effective biomarkers in human astrocytomas, clarifying the biological roles of oxidative/nitrative stress in the pathophysiology of astrocytomas, functional characteristics of nitroproteins in astrocytomas, nitration-mediated signal pathway network, and early diagnosis and treatment of astrocytomas. The results finds that these nitroproteins are enriched in mitotic cell components, which are related to transcription regulation, signal transduction, controlling subcellular organelle events, cell perception, maintaining cell homeostasis, and immune activity. Eleven statistically significant signal pathways are identified in astrocytoma, including remodeling of epithelial adherens junctions, germ cell-sertoli cell junction signaling, 14-3-3-mediated signaling, phagosome maturation, gap junction signaling, axonal guidance signaling, assembly of RNA polymerase III complex, and TREM1 signaling. Furthermore, protein tyrosine nitration is closely associated with the therapeutic effects of protein drugs, and molecular mechanism and drug targets of cancer. It provides valuable data for studying the protein nitration biomarkers, molecular mechanisms, and therapeutic targets of astrocytoma towards PPPM (3P medicine) practice. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-023-00348-y.
Collapse
Affiliation(s)
- Wenshuang Jia
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xiaoxia Gong
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Zhen Ye
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Na Li
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
5
|
Griswold-Prenner I, Kashyap AK, Mazhar S, Hall ZW, Fazelinia H, Ischiropoulos H. Unveiling the human nitroproteome: Protein tyrosine nitration in cell signaling and cancer. J Biol Chem 2023; 299:105038. [PMID: 37442231 PMCID: PMC10413360 DOI: 10.1016/j.jbc.2023.105038] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/28/2023] [Accepted: 07/06/2023] [Indexed: 07/15/2023] Open
Abstract
Covalent amino acid modification significantly expands protein functional capability in regulating biological processes. Tyrosine residues can undergo phosphorylation, sulfation, adenylation, halogenation, and nitration. These posttranslational modifications (PTMs) result from the actions of specific enzymes: tyrosine kinases, tyrosyl-protein sulfotransferase(s), adenylate transferase(s), oxidoreductases, peroxidases, and metal-heme containing proteins. Whereas phosphorylation, sulfation, and adenylation modify the hydroxyl group of tyrosine, tyrosine halogenation and nitration target the adjacent carbon residues. Because aberrant tyrosine nitration has been associated with human disorders and with animal models of disease, we have created an updated and curated database of 908 human nitrated proteins. We have also analyzed this new resource to provide insight into the role of tyrosine nitration in cancer biology, an area that has not previously been considered in detail. Unexpectedly, we have found that 879 of the 1971 known sites of tyrosine nitration are also sites of phosphorylation suggesting an extensive role for nitration in cell signaling. Overall, the review offers several forward-looking opportunities for future research and new perspectives for understanding the role of tyrosine nitration in cancer biology.
Collapse
Affiliation(s)
| | | | | | - Zach W Hall
- Nitrase Therapeutics, Brisbane, California, USA
| | - Hossein Fazelinia
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Harry Ischiropoulos
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
6
|
Smail SW, Babaei E, Amin K. Hematological, Inflammatory, Coagulation, and Oxidative/Antioxidant Biomarkers as Predictors for Severity and Mortality in COVID-19: A Prospective Cohort-Study. Int J Gen Med 2023; 16:565-580. [PMID: 36824986 PMCID: PMC9942608 DOI: 10.2147/ijgm.s402206] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/10/2023] [Indexed: 02/19/2023] Open
Abstract
Purpose Oxidative stress (OS) and inflammation are pivotal points in the pathophysiology of coronavirus disease-2019 (COVID-19). This study aims to use routine laboratory and oxidative stress/antioxidative biomarkers as predictors for the mortality of the disease. Patients and Methods This prospective cohort study, made up of 120 COVID-19 patients from emergency units in Erbil, Duhok, Kirkuk, and Sulaymaniyah cities in Iraq, from May the 1st to May the 30th, 2021, and 60 healthy controls (HCs) (n = 60). The patients were re-categorized into mild (n = 54), severe (n = 40), and critical (n = 26) groups based on the clinical criteria. Following admission to the hospital, blood was directly collected for measuring routine laboratory biomarkers. Results Neutrophils and neutrophil/lymphocyte ratio (NLR) were higher in the critical group, while lymphocytes were lower in the severe and critical groups compared to the mild group. The CRP, ferritin, and D-dimer values were more elevated in severe and critical cases than in mild COVID-19 cases. The levels of malondialdehyde (MDA), nitric oxide (NO), and copper were elevated, while the superoxide dismutase (SOD) activity level and total antioxidant capacity (TAC) level were lower. However, vitamin C, glutathione peroxidase (GPx), and catalase activity levels were not changed in the COVID-19 groups compared to the HCs. NO and ferritin were predictors of ICU hospitalization; D-dimer, MDA, and NLR were predictors of mortality. NO, and NLR were predictors of SpO2 depression. Moreover, NO, and copper have both good diagnostic values, their cutoffs were 39.01 and 11.93, respectively. Conclusion There is an association between immune dysregulation and oxidative imbalance. The biomarkers, that could be considered as predictors for the severity and mortality of COVID-19, are the NLR, NO, ferritin, and D-dimer. The age equal to and older than 50 has a poor prognosis in the Kurdish population.
Collapse
Affiliation(s)
- Shukur Wasman Smail
- Department of Biology, College of Science, Salahaddin University, Erbil, Iraq
| | - Esmaeil Babaei
- Department of Biology, School of Natural Sciences, University of Tabriz, Tabriz, Iran
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq
| | - Kawa Amin
- College of Medicine, University of Sulaimani, Sulaymaniyah, Iraq
| |
Collapse
|
7
|
Franklin R, Hare M, Beckman JS. Determining Copper and Zinc Content in Superoxide Dismutase Using Electron Capture Dissociation Under Native Spray Conditions. Methods Mol Biol 2022; 2500:201-210. [PMID: 35657595 DOI: 10.1007/978-1-0716-2325-1_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Localizing metal binding to specific sites in proteins remains a challenging analytical problem in vitro and in vivo. Although metal binding can be maintained by "native" electrospray ionization with intact proteins for quantitation by mass spectrometry, subsequent fragmentation of proteins with slow-heating methods like collision-induced dissociation (CID) can scramble and detach metals. In contrast, electron capture dissociation (ECD) fragmentation produces highly localized bond cleavage that is well known to preserve posttranslational modifications. We show how a newly available ECD tool that can be retrofitted on standard QTOF mass spectrometers allows the sites of copper and zinc binding to be localized in the antioxidant enzyme Cu, Zn superoxide dismutase (SOD1). The loss of zinc from Cu, Zn SOD1 has been shown to induce motor neuron death and could have a causal role in the fatal neurodegenerative disease, amyotrophic lateral sclerosis (ALS). The methods described enable copper loss to be distinguished from zinc using distinct ECD fragments of SOD1 and are broadly applicable to other metalloproteins.
Collapse
Affiliation(s)
- Rachel Franklin
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, USA
| | | | - Joseph S Beckman
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, USA.
- e-MSion Inc., Corvallis, OR, USA.
| |
Collapse
|
8
|
Hydrogen Peroxide and Amyotrophic Lateral Sclerosis: From Biochemistry to Pathophysiology. Antioxidants (Basel) 2021; 11:antiox11010052. [PMID: 35052556 PMCID: PMC8773294 DOI: 10.3390/antiox11010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/18/2021] [Accepted: 12/24/2021] [Indexed: 11/19/2022] Open
Abstract
Free radicals are unstable chemical reactive species produced during Redox dyshomeostasis (RDH) inside living cells and are implicated in the pathogenesis of various neurodegenerative diseases. One of the most complicated and life-threatening motor neurodegenerative diseases (MND) is amyotrophic lateral sclerosis (ALS) because of the poor understanding of its pathophysiology and absence of an effective treatment for its cure. During the last 25 years, researchers around the globe have focused their interest on copper/zinc superoxide dismutase (Cu/Zn SOD, SOD1) protein after the landmark discovery of mutant SOD1 (mSOD1) gene as a risk factor for ALS. Substantial evidence suggests that toxic gain of function due to redox disturbance caused by reactive oxygen species (ROS) changes the biophysical properties of native SOD1 protein thus, instigating its fibrillization and misfolding. These abnormal misfolding aggregates or inclusions of SOD1 play a role in the pathogenesis of both forms of ALS, i.e., Sporadic ALS (sALS) and familial ALS (fALS). However, what leads to a decrease in the stability and misfolding of SOD1 is still in question and our scientific knowledge is scarce. A large number of studies have been conducted in this area to explore the biochemical mechanistic pathway of SOD1 aggregation. Several studies, over the past two decades, have shown that the SOD1-catalyzed biochemical reaction product hydrogen peroxide (H2O2) at a pathological concentration act as a substrate to trigger the misfolding trajectories and toxicity of SOD1 in the pathogenesis of ALS. These toxic aggregates of SOD1 also cause aberrant localization of TAR-DNA binding protein 43 (TDP-43), which is characteristic of neuronal cytoplasmic inclusions (NCI) found in ALS. Here in this review, we present the evidence implicating the pivotal role of H2O2 in modulating the toxicity of SOD1 in the pathophysiology of the incurable and highly complex disease ALS. Also, highlighting the role of H2O2 in ALS, we believe will encourage scientists to target pathological concentrations of H2O2 thereby halting the misfolding of SOD1.
Collapse
|
9
|
Keon M, Musrie B, Dinger M, Brennan SE, Santos J, Saksena NK. Destination Amyotrophic Lateral Sclerosis. Front Neurol 2021; 12:596006. [PMID: 33854469 PMCID: PMC8039771 DOI: 10.3389/fneur.2021.596006] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 03/02/2021] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a prototypical neurodegenerative disease characterized by progressive degeneration of motor neurons both in the brain and spinal cord. The constantly evolving nature of ALS represents a fundamental dimension of individual differences that underlie this disorder, yet it involves multiple levels of functional entities that alternate in different directions and finally converge functionally to define ALS disease progression. ALS may start from a single entity and gradually becomes multifactorial. However, the functional convergence of these diverse entities in eventually defining ALS progression is poorly understood. Various hypotheses have been proposed without any consensus between the for-and-against schools of thought. The present review aims to capture explanatory hierarchy both in terms of hypotheses and mechanisms to provide better insights on how they functionally connect. We can then integrate them within a common functional frame of reference for a better understanding of ALS and defining future treatments and possible therapeutic strategies. Here, we provide a philosophical understanding of how early leads are crucial to understanding the endpoints in ALS, because invariably, all early symptomatic leads are underpinned by neurodegeneration at the cellular, molecular and genomic levels. Consolidation of these ideas could be applied to other neurodegenerative diseases (NDs) and guide further critical thinking to unveil their roadmap of destination ALS.
Collapse
Affiliation(s)
- Matt Keon
- GenieUs Genomics Pty Ltd., Sydney, NSW, Australia
| | | | - Marcel Dinger
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | | | - Jerran Santos
- Advanced Tissue Engineering and Stem Cell Biology Group, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
10
|
Role of Oxidative Stress in the Pathogenesis of Amyotrophic Lateral Sclerosis: Antioxidant Metalloenzymes and Therapeutic Strategies. Biomolecules 2021; 11:biom11030437. [PMID: 33809730 PMCID: PMC8002298 DOI: 10.3390/biom11030437] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/12/2021] [Accepted: 03/12/2021] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) affects motor neurons in the cerebral cortex, brainstem and spinal cord and leads to death due to respiratory failure within three to five years. Although the clinical symptoms of this disease were first described in 1869 and it is the most common motor neuron disease and the most common neurodegenerative disease in middle-aged individuals, the exact etiopathogenesis of ALS remains unclear and it remains incurable. However, free oxygen radicals (i.e., molecules containing one or more free electrons) are known to contribute to the pathogenesis of this disease as they very readily bind intracellular structures, leading to functional impairment. Antioxidant enzymes, which are often metalloenzymes, inactivate free oxygen radicals by converting them into a less harmful substance. One of the most important antioxidant enzymes is Cu2+Zn2+ superoxide dismutase (SOD1), which is mutated in 20% of cases of the familial form of ALS (fALS) and up to 7% of sporadic ALS (sALS) cases. In addition, the proper functioning of catalase and glutathione peroxidase (GPx) is essential for antioxidant protection. In this review article, we focus on the mechanisms through which these enzymes are involved in the antioxidant response to oxidative stress and thus the pathogenesis of ALS and their potential as therapeutic targets.
Collapse
|
11
|
Onyango IG, Bennett JP, Stokin GB. Regulation of neuronal bioenergetics as a therapeutic strategy in neurodegenerative diseases. Neural Regen Res 2021; 16:1467-1482. [PMID: 33433460 PMCID: PMC8323696 DOI: 10.4103/1673-5374.303007] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis are a heterogeneous group of debilitating disorders with multifactorial etiologies and pathogeneses that manifest distinct molecular mechanisms and clinical manifestations with abnormal protein dynamics and impaired bioenergetics. Mitochondrial dysfunction is emerging as an important feature in the etiopathogenesis of these age-related neurodegenerative diseases. The prevalence and incidence of these diseases is on the rise with the increasing global population and average lifespan. Although many therapeutic approaches have been tested, there are currently no effective treatment routes for the prevention or cure of these diseases. We present the current status of our knowledge and understanding of the involvement of mitochondrial dysfunction in these diseases and highlight recent advances in novel therapeutic strategies targeting neuronal bioenergetics as potential approach for treating these diseases.
Collapse
Affiliation(s)
- Isaac G Onyango
- Center for Translational Medicine, International Clinical Research Centre (ICRC), St. Anne's University Hospital, Brno, Czech Republic
| | - James P Bennett
- Neurodegeneration Therapeutics, 3050A Berkmar Drive, Charlottesville, VA, USA
| | - Gorazd B Stokin
- Center for Translational Medicine, International Clinical Research Centre (ICRC), St. Anne's University Hospital, Brno, Czech Republic
| |
Collapse
|
12
|
3-Nitrotyrosine and related derivatives in proteins: precursors, radical intermediates and impact in function. Essays Biochem 2020; 64:111-133. [PMID: 32016371 DOI: 10.1042/ebc20190052] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/30/2019] [Accepted: 01/03/2020] [Indexed: 12/22/2022]
Abstract
Oxidative post-translational modification of proteins by molecular oxygen (O2)- and nitric oxide (•NO)-derived reactive species is a usual process that occurs in mammalian tissues under both physiological and pathological conditions and can exert either regulatory or cytotoxic effects. Although the side chain of several amino acids is prone to experience oxidative modifications, tyrosine residues are one of the preferred targets of one-electron oxidants, given the ability of their phenolic side chain to undergo reversible one-electron oxidation to the relatively stable tyrosyl radical. Naturally occurring as reversible catalytic intermediates at the active site of a variety of enzymes, tyrosyl radicals can also lead to the formation of several stable oxidative products through radical-radical reactions, as is the case of 3-nitrotyrosine (NO2Tyr). The formation of NO2Tyr mainly occurs through the fast reaction between the tyrosyl radical and nitrogen dioxide (•NO2). One of the key endogenous nitrating agents is peroxynitrite (ONOO-), the product of the reaction of superoxide radical (O2•-) with •NO, but ONOO--independent mechanisms of nitration have been also disclosed. This chemical modification notably affects the physicochemical properties of tyrosine residues and because of this, it can have a remarkable impact on protein structure and function, both in vitro and in vivo. Although low amounts of NO2Tyr are detected under basal conditions, significantly increased levels are found at pathological states related with an overproduction of reactive species, such as cardiovascular and neurodegenerative diseases, inflammation and aging. While NO2Tyr is a well-established stable oxidative stress biomarker and a good predictor of disease progression, its role as a pathogenic mediator has been laboriously defined for just a small number of nitrated proteins and awaits further studies.
Collapse
|
13
|
Vernazza S, Tirendi S, Bassi AM, Traverso CE, Saccà SC. Neuroinflammation in Primary Open-Angle Glaucoma. J Clin Med 2020; 9:E3172. [PMID: 33007927 PMCID: PMC7601106 DOI: 10.3390/jcm9103172] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 12/18/2022] Open
Abstract
Primary open-angle glaucoma (POAG) is the second leading cause of irreversible blindness worldwide. Increasing evidence suggests oxidative damage and immune response defects are key factors contributing to glaucoma onset. Indeed, both the failure of the trabecular meshwork tissue in the conventional outflow pathway and the neuroinflammation process, which drives the neurodegeneration, seem to be linked to the age-related over-production of free radicals (i.e., mitochondrial dysfunction) and to oxidative stress-linked immunostimulatory signaling. Several previous studies have described a wide range of oxidative stress-related makers which are found in glaucomatous patients, including low levels of antioxidant defences, dysfunction/activation of glial cells, the activation of the NF-κB pathway and the up-regulation of pro-inflammatory cytokines, and so on. However, the intraocular pressure is still currently the only risk factor modifiable by medication or glaucoma surgery. This present review aims to summarize the multiple cellular processes, which promote different risk factors in glaucoma including aging, oxidative stress, trabecular meshwork defects, glial activation response, neurodegenerative insults, and the altered regulation of immune response.
Collapse
Affiliation(s)
| | - Sara Tirendi
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (S.T.); (A.M.B.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), Italy
| | - Anna Maria Bassi
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (S.T.); (A.M.B.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), Italy
| | - Carlo Enrico Traverso
- Clinica Oculistica, DiNOGMI, University of Genoa, 16132 Genoa, Italy;
- Ophthalmology Unit, IRCCS-Polyclinic San Martino Hospital, 16132 Genoa, Italy;
| | | |
Collapse
|
14
|
Roy P, Panda A, Hati S, Dasgupta S. pH-Dependent Nitrotyrosine Formation in Ribonuclease A is Enhanced in the Presence of Polyethylene Glycol (PEG). Chem Asian J 2019; 14:4780-4792. [PMID: 31591811 DOI: 10.1002/asia.201901225] [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: 09/02/2019] [Revised: 10/03/2019] [Indexed: 11/08/2022]
Abstract
Protein nitration can occur as a result of peroxynitrite-mediated oxidative stress. Excess production of peroxynitrite (PN) within the cellular medium can cause oxidative damage to biomolecules. The in vitro nitration of Ribonuclease A (RNase A) results in nitrotyrosine (NT) formation with a strong dependence on the pH of the medium. In order to mimic the cellular environment in this study, PN-mediated RNase A nitration has been carried out in a crowded medium. The degree of nitration is higher at pH 7.4 (physiological pH) compared to pH 6.0 (tumor cell pH). The extent of nitration increases significantly when PN is added to RNase A in the presence of crowding agents PEG 400 and PEG 6000. PEG has been found to stabilize PN over a prolonged period, thereby increasing the degree of nitration. NT formation in RNase A also results in a significant loss in enzymatic activity.
Collapse
Affiliation(s)
- Pritam Roy
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Atashi Panda
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Sumon Hati
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Swagata Dasgupta
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| |
Collapse
|
15
|
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
|
16
|
Abstract
Nitric oxide (NO) is a cellular signalling molecule widely conserved among organisms, including microorganisms such as bacteria, yeasts, and fungi, and higher eukaryotes such as plants and mammals. NO is mainly produced by the activities of NO synthase (NOS) or nitrite reductase (NIR). There are several NO detoxification systems, including NO dioxygenase (NOD) and S-nitrosoglutathione reductase (GSNOR). NO homeostasis, based on the balance between NO synthesis and degradation, is important for regulating its physiological functions, since an excess of NO causes nitrosative stress due to the high reactivity of NO and NO-derived compounds. In yeast, NO may be involved in stress responses, but the role of NO and the mechanism underlying NO signalling are poorly understood due to the lack of mammalian NOS orthologs in the yeast genome. NOS and NIR activities have been observed in yeast cells, but the gene-encoding NOS and the mechanism by which NO production is catalysed by NIR remain unclear. On the other hand, yeast cells employ NOD and GSNOR to maintain intracellular redox balance following endogenous NO production, treatment with exogenous NO, or exposure to environmental stresses. This article reviews NO metabolism (synthesis, degradation) and its regulation in yeast. The physiological roles of NO in yeast, including the oxidative stress response, are also discussed. Such investigations into NO signalling are essential for understanding how NO modulates the genetics and physiology of yeast. In addition to being responsible for the pathology and pharmacology of various degenerative diseases, NO signalling may be a potential target for the construction and engineering of industrial yeast strains.
Collapse
|
17
|
Ramdial K, Franco MC, Estevez AG. Cellular mechanisms of peroxynitrite-induced neuronal death. Brain Res Bull 2017; 133:4-11. [PMID: 28655600 DOI: 10.1016/j.brainresbull.2017.05.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 04/21/2017] [Accepted: 05/12/2017] [Indexed: 12/13/2022]
|
18
|
Pal A, Das S. Morphine causes persistent induction of nitrated neurofilaments in cortex and subcortex even during abstinence. Neuroscience 2015; 291:177-88. [DOI: 10.1016/j.neuroscience.2015.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 02/03/2015] [Accepted: 02/04/2015] [Indexed: 01/14/2023]
|
19
|
Hilton JB, White AR, Crouch PJ. Metal-deficient SOD1 in amyotrophic lateral sclerosis. J Mol Med (Berl) 2015; 93:481-7. [PMID: 25754173 PMCID: PMC4408375 DOI: 10.1007/s00109-015-1273-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/24/2015] [Accepted: 02/25/2015] [Indexed: 12/13/2022]
Abstract
Mutations to the ubiquitous antioxidant enzyme Cu/Zn superoxide dismutase (SOD1) were the first established genetic cause of the fatal, adult-onset neurodegenerative disease amyotrophic lateral sclerosis (ALS). It is widely accepted that these mutations do not cause ALS via a loss of antioxidant function, but elucidating the alternate toxic gain of function has proven to be elusive. Under physiological conditions, SOD1 binds one copper ion and one zinc ion per monomer to form a highly stable and functional homodimer, but there is now ample evidence to indicate aberrant persistence of SOD1 in an intermediate metal-deficient state may contribute to the protein’s involvement in ALS. This review briefly discusses some of the data to support a role for metal-deficient SOD1 in the development of ALS and some of the outcomes from drug development studies that have aimed to modify the symptoms of ALS by targeting the metal state of SOD1. The implications for the metal state of SOD1 in cases of sporadic ALS that do not involve mutant SOD1 are also discussed.
Collapse
Affiliation(s)
- James B Hilton
- Department of Pathology, The University of Melbourne, Melbourne, Victoria, 3010, Australia
| | | | | |
Collapse
|
20
|
Yeo WS, Kim YJ, Kabir MH, Kang JW, Ahsan-Ul-Bari M, Kim KP. Mass spectrometric analysis of protein tyrosine nitration in aging and neurodegenerative diseases. MASS SPECTROMETRY REVIEWS 2015; 34:166-183. [PMID: 24889964 DOI: 10.1002/mas.21429] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
This review highlights the significance of protein tyrosine nitration (PTN) in signal transduction pathways, the progress achieved in analytical methods, and the implication of nitration in the cellular pathophysiology of aging and age-related neurodegenerative diseases. Although mass spectrometry of nitrated peptides has become a powerful tool for the characterization of nitrated peptides, the low stoichiometry of this modification clearly necessitates the use of affinity chromatography to enrich modified peptides. Analysis of nitropeptides involves identification of endogenous, intact modification as well as chemical conversion of the nitro group to a chemically reactive amine group and further modifications that enable affinity capture and enhance detectability by altering molecular properties. In this review, we focus on the recent progress in chemical derivatization of nitropeptides for enrichment and mass analysis, and for detection and quantification using various analytical tools. PTN participates in physiological processes, such as aging and neurodegenerative diseases. Accumulation of 3-nitrotyrosine has been found to occur during the aging process; this was identified through mass spectrometry. Further, there are several studies implicating the presence of nitrated tyrosine in age-related diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Woon-Seok Yeo
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, 143-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
21
|
O'Hare Doig RL, Bartlett CA, Maghzal GJ, Lam M, Archer M, Stocker R, Fitzgerald M. Reactive species and oxidative stress in optic nerve vulnerable to secondary degeneration. Exp Neurol 2014; 261:136-46. [PMID: 24931225 DOI: 10.1016/j.expneurol.2014.06.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 05/24/2014] [Accepted: 06/05/2014] [Indexed: 12/20/2022]
Abstract
Secondary degeneration contributes substantially to structural and functional deficits following traumatic injury to the CNS. While it has been proposed that oxidative stress is a feature of secondary degeneration, contributing reactive species and resultant oxidized products have not been clearly identified in vivo. The study is designed to identify contributors to, and consequences of, oxidative stress in a white matter tract vulnerable to secondary degeneration. Partial dorsal transection of the optic nerve (ON) was used to model secondary degeneration in ventral nerve unaffected by the primary injury. Reactive species were assessed using fluorescent labelling and liquid chromatography/tandem mass spectroscopy (LC/MS/MS). Antioxidant enzymes and oxidized products were semi-quantified immunohistochemically. Mitophagy was assessed by electron microscopy. Fluorescent indicators of reactive oxygen and/or nitrogen species increased at 1, 3 and 7days after injury, in ventral ON. LC/MS/MS confirmed increases in reactive species linked to infiltrating microglia/macrophages in dorsal ON. Similarly, immunoreactivity for glutathione peroxidase and haem oxygenase-1 increased in ventral ON at 3 and 7days after injury, respectively. Despite increased antioxidant immunoreactivity, DNA oxidation was evident from 1day, lipid oxidation at 3days, and protein nitration at 7days after injury. Nitrosative and oxidative damage was particularly evident in CC1-positive oligodendrocytes, at times after injury at which structural abnormalities of the Node of Ranvier/paranode complex have been reported. The incidence of mitochondrial autophagic profiles was also significantly increased from 3days. Despite modest increases in antioxidant enzymes, increased reactive species are accompanied by oxidative and nitrosative damage to DNA, lipid and protein, associated with increasing abnormal mitochondria, which together may contribute to the deficits of secondary degeneration.
Collapse
Affiliation(s)
- Ryan L O'Hare Doig
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Carole A Bartlett
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Ghassan J Maghzal
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia; University of New South Wales, NSW, Australia
| | - Magdalena Lam
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Michael Archer
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Roland Stocker
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia; University of New South Wales, NSW, Australia
| | - Melinda Fitzgerald
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia.
| |
Collapse
|
22
|
Pollari E, Goldsteins G, Bart G, Koistinaho J, Giniatullin R. The role of oxidative stress in degeneration of the neuromuscular junction in amyotrophic lateral sclerosis. Front Cell Neurosci 2014; 8:131. [PMID: 24860432 PMCID: PMC4026683 DOI: 10.3389/fncel.2014.00131] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/27/2014] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by the progressive loss of motoneurons and degradation of the neuromuscular junctions (NMJ). Consistent with the dying-back hypothesis of motoneuron degeneration the decline in synaptic function initiates from the presynaptic terminals in ALS. Oxidative stress is a major contributory factor to ALS pathology and affects the presynaptic transmitter releasing machinery. Indeed, in ALS mouse models nerve terminals are sensitive to reactive oxygen species (ROS) suggesting that oxidative stress, along with compromised mitochondria and increased intracellular Ca(2+) amplifies the presynaptic decline in NMJ. This initial dysfunction is followed by a neurodegeneration induced by inflammatory agents and loss of trophic support. To develop effective therapeutic approaches against ALS, it is important to identify the mechanisms underlying the initial pathological events. Given the role of oxidative stress in ALS, targeted antioxidant treatments could be a promising therapeutic approach. However, the complex nature of ALS and failure of monotherapies suggest that an antioxidant therapy should be accompanied by anti-inflammatory interventions to enhance the restoration of the redox balance.
Collapse
Affiliation(s)
- Eveliina Pollari
- Molecular Brain Research Laboratory, Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland Kuopio, Finland ; Experimental Neurology - Laboratory of Neurobiology, Department of Neurosciences, Vesalius Research Center, KULeuven - University of Leuven Leuven, Belgium
| | - Gundars Goldsteins
- Molecular Brain Research Laboratory, Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland Kuopio, Finland
| | - Geneviève Bart
- Cell Biology Laboratory, Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland Kuopio, Finland
| | - Jari Koistinaho
- Molecular Brain Research Laboratory, Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland Kuopio, Finland
| | - Rashid Giniatullin
- Cell Biology Laboratory, Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland Kuopio, Finland ; Laboratory of Neurobiology, Department of Physiology, Kazan Federal University Kazan, Russia
| |
Collapse
|
23
|
Hitchler MJ, Domann FE. Regulation of CuZnSOD and its redox signaling potential: implications for amyotrophic lateral sclerosis. Antioxid Redox Signal 2014; 20:1590-8. [PMID: 23795822 PMCID: PMC3960847 DOI: 10.1089/ars.2013.5385] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Molecular oxygen is a Janus-faced electron acceptor for biological systems, serving as a reductant for respiration, or as the genesis for oxygen-derived free radicals that damage macromolecules. Superoxide is well known to perturb nonheme iron proteins, including Fe/S proteins such as aconitase and succinate dehydrogenase, as well as other enzymes containing labile iron such as the prolyl hydroxylase domain-containing family of enzymes; whereas hydrogen peroxide is more specific for two-electron reactions with thiols on glutathione, glutaredoxin, thioredoxin, and the peroxiredoxins. RECENT ADVANCES Over the past two decades, familial cases of amyotrophic lateral sclerosis (ALS) have been shown to have an association with commonly altered superoxide dismutase 1 (SOD1) activity, expression, and protein structure. This has led to speculation that an altered redox balance may have a role in creating the ALS phenotype. CRITICAL ISSUES While SOD1 alterations in familial ALS are manifold, they generally create perturbations in the flux of electrons. The nexus of SOD1 between one- and two-electron signaling processes places it at a key signaling regulatory checkpoint for governing cellular responses to physiological and environmental cues. FUTURE DIRECTIONS The manner in which ALS-associated mutations adjust SOD1's role in controlling the flow of electrons between one- and two-electron signaling processes remains obscure. Here, we discuss the ways in which SOD1 mutations influence the form and function of copper zinc SOD, the consequences of these alterations on free radical biology, and how these alterations might influence cell signaling during the onset of ALS.
Collapse
Affiliation(s)
- Michael J Hitchler
- 1 Department of Radiation Oncology, Kaiser Permanente Los Angeles Medical Center , Los Angeles, California
| | | |
Collapse
|
24
|
Capacity of HSYA to inhibit nitrotyrosine formation induced by focal ischemic brain injury. Nitric Oxide 2013; 35:144-51. [PMID: 24126016 DOI: 10.1016/j.niox.2013.10.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 09/25/2013] [Accepted: 10/01/2013] [Indexed: 12/21/2022]
Abstract
Peroxynitrite-mediated protein tyrosine nitration represents a crucial pathogenic mechanism of stroke. Hydroxysafflor yellow A (HSYA) is the most important active component of the safflower plant. Here we assess the neuroprotective efficacy of HSYA and investigate the mechanism through anti-nitrative pathway. Rats were subjected to 60-min ischemia followed by reperfusion. HSYA (2.5-10mg/kg) was injected at 1h after ischemia onset. Other groups received HSYA (10mg/kg) treatment at 3-9h after onset. Infarct volume, brain edema, and neurological score were evaluated at 24h after ischemia. Nitrotyrosine and inducible NO synthase (iNOS) expression, as well as NO level (nitrate/nitrite) in ischemic cortex was examined within 24h after ischemia. The ability of HSYA to scavenge peroxynitrite was evaluated in vitro. Infarct volume was significantly decreased by HSYA (P<0.05), with a therapeutic window of 3h after ischemia at dose of 10mg/kg. HSYA treatment also reduced brain edema and improved neurological score (P<0.05). Nitrotyrosine formation was dose- and time-dependently inhibited by HSYA. The time window of HSYA in decreasing protein tyrosine nitration paralleled its action in infarct volume. HSYA also greatly reduced iNOS expression and NO content at 24h after ischemia, suggesting prevention of peroxynitrite generation from iNOS. In vitro, HSYA blocked authentic peroxynitrite-induced tyrosine nitration in bovine serum albumin and primary cortical neurons. Collectively, our results indicated that post-ischemic HSYA treatment attenuates brain ischemic injury which is at least partially due to reducing nitrotyrosine formation, possibly by the combined mechanism of its peroxynitrite scavenging ability and its reduction in iNOS production.
Collapse
|
25
|
Deeb RS, Nuriel T, Cheung C, Summers B, Lamon BD, Gross SS, Hajjar DP. Characterization of a cellular denitrase activity that reverses nitration of cyclooxygenase. Am J Physiol Heart Circ Physiol 2013; 305:H687-98. [PMID: 23792683 PMCID: PMC3761327 DOI: 10.1152/ajpheart.00876.2012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 06/20/2013] [Indexed: 12/11/2022]
Abstract
Protein 3-nitrotyrosine (3-NT) formation is frequently regarded as a simple biomarker of disease, an irreversible posttranslational modification that can disrupt protein structure and function. Nevertheless, evidence that protein 3-NT modifications may be site selective and reversible, thus allowing for physiological regulation of protein activity, has begun to emerge. We have previously reported that cyclooxygenase (COX)-1 undergoes heme-dependent nitration of Tyr(385), an internal and catalytically essential residue. In the present study, we demonstrate that nitrated COX-1 undergoes a rapid reversal of nitration by substrate-selective and biologically regulated denitrase activity. Using nitrated COX-1 as a substrate, denitrase activity was validated and quantified by analytic HPLC with electrochemical detection and determined to be constitutively active in murine and human endothelial cells, macrophages, and a variety of tissue samples. Smooth muscle cells, however, contained little denitrase activity. Further characterizing this denitrase activity, we found that it was inhibited by free 3-NT and may be enhanced by endogenous nitric oxide and exogenously administered carbon monoxide. Finally, we describe a purification protocol that results in significant enrichment of a discrete denitrase-containing fraction, which maintains activity throughout the purification process. These findings reveal that nitrated COX-1 is a substrate for a denitrase in cells and tissues, implying that the reciprocal processes of nitration and denitration may modulate bioactive lipid synthesis in the setting of inflammation. In addition, our data reveal that denitration is a controlled process that may have broad importance for regulating cell signaling events in nitric oxide-generating systems during oxidative/nitrosative stress.
Collapse
MESH Headings
- Adaptation, Physiological/physiology
- Animals
- Cell Line
- Cells, Cultured
- Cyclooxygenase 1/metabolism
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- Humans
- Macrophages/cytology
- Macrophages/metabolism
- Mice
- Mice, Inbred C57BL
- Models, Animal
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Nitrates/metabolism
- Nitric Oxide/metabolism
- Nitric Oxide Synthase/metabolism
- Oxidative Stress/physiology
- Oxidoreductases/metabolism
- Rats
- Tyrosine/analogs & derivatives
- Tyrosine/metabolism
Collapse
Affiliation(s)
- Ruba S Deeb
- Department of Pathology, Weill Cornell Medical College, Cornell University, New York, New York
| | | | | | | | | | | | | |
Collapse
|
26
|
Mead RJ, Higginbottom A, Allen SP, Kirby J, Bennett E, Barber SC, Heath PR, Coluccia A, Patel N, Gardner I, Brancale A, Grierson AJ, Shaw PJ. S[+] Apomorphine is a CNS penetrating activator of the Nrf2-ARE pathway with activity in mouse and patient fibroblast models of amyotrophic lateral sclerosis. Free Radic Biol Med 2013; 61:438-52. [PMID: 23608463 PMCID: PMC3684770 DOI: 10.1016/j.freeradbiomed.2013.04.018] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 03/22/2013] [Accepted: 04/12/2013] [Indexed: 12/13/2022]
Abstract
Compelling evidence indicates that oxidative stress contributes to motor neuron injury in amyotrophic lateral sclerosis (ALS), but antioxidant therapies have not yet achieved therapeutic benefit in the clinic. The nuclear erythroid 2-related-factor 2 (Nrf2) transcription factor is a key regulator of an important neuroprotective response by driving the expression of multiple cytoprotective genes via its interaction with the antioxidant response element (ARE). Dysregulation of the Nrf2-ARE system has been identified in ALS models and human disease. Taking the Nrf2-ARE pathway as an attractive therapeutic target for neuroprotection in ALS, we aimed to identify CNS penetrating, small molecule activators of Nrf2-mediated transcription in a library of 2000 drugs and natural products. Compounds were screened extensively for Nrf2 activation, and antioxidant and neuroprotective properties in vitro. S[+]-Apomorphine, a receptor-inactive enantiomer of the clinically approved dopamine-receptor agonist (R[-]-apomorphine), was identified as a nontoxic Nrf2 activating molecule. In vivo S[+]-apomorphine demonstrated CNS penetrance, Nrf2 induction, and significant attenuation of motor dysfunction in the SOD1(G93A) transgenic mouse model of ALS. S[+]-apomorphine also reduced pathological oxidative stress and improved survival following an oxidative insult in fibroblasts from ALS patients. This molecule emerges as a promising candidate for evaluation as a potential neuroprotective agent in ALS patients in the clinic.
Collapse
Affiliation(s)
- Richard J Mead
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, School of Medicine and Biomedical Sciences, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Adrian Higginbottom
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, School of Medicine and Biomedical Sciences, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Scott P Allen
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, School of Medicine and Biomedical Sciences, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Janine Kirby
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, School of Medicine and Biomedical Sciences, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Ellen Bennett
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, School of Medicine and Biomedical Sciences, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Siân C Barber
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, School of Medicine and Biomedical Sciences, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, School of Medicine and Biomedical Sciences, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Antonio Coluccia
- Welsh School of Pharmacy, King Edward VII Avenue, Cardiff, CF10 3NB Wales, UK
| | - Neelam Patel
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, School of Medicine and Biomedical Sciences, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Iain Gardner
- SimCyp, Blades Enterprise Centre, John Street, Sheffield, S2 4SU, UK
| | - Andrea Brancale
- Welsh School of Pharmacy, King Edward VII Avenue, Cardiff, CF10 3NB Wales, UK
| | - Andrew J Grierson
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, School of Medicine and Biomedical Sciences, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, School of Medicine and Biomedical Sciences, University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK.
| |
Collapse
|
27
|
|
28
|
Lin HL, Kenaan C, Zhang H, Hollenberg PF. Reaction of human cytochrome P450 3A4 with peroxynitrite: nitrotyrosine formation on the proximal side impairs its interaction with NADPH-cytochrome P450 reductase. Chem Res Toxicol 2012; 25:2642-53. [PMID: 23016756 DOI: 10.1021/tx3002753] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The reaction of peroxynitrite (PN) with purified human cytochrome P450 3A4 (CYP3A4) resulted in the loss of the reduced-CO difference spectrum, but the absolute absorption spectrum of the heme was not significantly altered. The loss of 7-benzyloxy-4-(trifluoromethyl)coumarin (BFC) O-debenzylation activity of CYP3A4 was concentration-dependent with respect to PN, and the loss of BFC activity supported by NADPH-cytochrome P450 reductase (CPR) was much greater than that supported by tert-butyl hydroperoxide. Moreover, the PN-treated CYP3A4 exhibited a reduced-CO spectrum when reduced by CPR that was much smaller than when it was reduced by dithionite. These results suggest that modification of CYP3A4 by PN may impair its interaction with CPR, leading to the loss of catalytic activity. Tyrosine nitration, as measured by an increase in mass of 45 Da due to the addition of a nitro group, was used as a biomarker for protein modification by PN. PN-treated CYP3A4 was digested by trypsin and endoproteinase Glu C, and nitrotyrosine formation was then determined by using electrospray ionization-liquid chromatography-tandem mass spectrometry. Tyr residues 99, 307, 347, 430, and 432 were found to be nitrated. Using the GRAMM-X docking program, the structure for the CYP3A4-CPR complex shows that Tyr99, Tyr347, and Tyr430 are on the proximal side of CYP3A4 and are in close contact with three acidic residues in the FMN domain of CPR, suggesting that modification of one or more of these tyrosine residues by PN may influence CPR binding or the transfer of electrons to CYP3A4. Mutagenesis of Tyr430 to Phe or Val revealed that both the aromatic and the hydroxyl groups of Tyr are required for CPR-dependent catalytic activity and thus support the idea that the proximal side Tyr participates in the 3A4-CPR interaction. In conclusion, modification of tyrosine residues by PN and their subsequent identification can be used to enhance our knowledge of the structure/function relationships of the P450s with respect to the electron transfer steps, which are critical for P450 activity.
Collapse
Affiliation(s)
- Hsia-lien Lin
- Department of Pharmacology, University of Michigan, 2301 MSRB III, 1150 West Medical Center Drive, Ann Arbor, Michigan 48109-5632, United States
| | | | | | | |
Collapse
|
29
|
Nitric oxide-mediated oxidative damage and the progressive demise of motor neurons in ALS. Neurotox Res 2012; 22:251-64. [PMID: 22488161 DOI: 10.1007/s12640-012-9322-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 03/15/2012] [Accepted: 03/24/2012] [Indexed: 10/28/2022]
Abstract
Oxidative damage is a common and early feature of Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis (ALS), and other neurodegenerative disorders. Dr. Mark Smith and his colleagues have built the case for oxidative stress being a primary progenitor rather than a secondary end-stage epiphenomenon of neurodegeneration. They proposed that reactive oxygen species contribute to the "age-related cascade of neurodegeneration," whereby accumulative oxidative damage with age promotes other characteristic pathological changes in afflicted brain regions, including protein aggregation, metabolic deficiencies, and inflammation. Nitric oxide (NO) likely plays a critical role in this age-related cascade. NO is a major signaling molecule produced in the central nervous system to modulate neurological activity through stimulating cyclic GMP synthesis. However, the same physiological concentrations of NO, relevant in cellular signaling, may also initiate and amplify oxidative damage by diffusion-limited reactions with superoxide (O(2)(•-)) to produce peroxynitrite (ONOO(-)). This is perhaps best illustrated in ALS where physiological levels of NO promote survival of motor neurons, but the same concentrations can stimulate motor neuron apoptosis and glial cell activation under pathological conditions. While these changes represent a complex mechanism involving multiple cell types in the pathogenesis of ALS, they also reveal general processes underlying neurodegeneration.
Collapse
|
30
|
Morita M, Nakajima A, Takemura A, Okuno M. Involvement of redox- and phosphorylation-dependent pathways in osmotic adaptation in sperm cells of euryhaline tilapia. ACTA ACUST UNITED AC 2011; 214:2096-104. [PMID: 21613527 DOI: 10.1242/jeb.053405] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Sperm cells involved in fertilisation must tolerate hypo-osmotic and hyper-osmotic environments. Euryhaline tilapia (Oreochromis mossambicus) can acclimatise to and reproduce in freshwater and seawater because its sperm are able to adapt to these differing osmotic environments. In this study, we found that the dephosphorylation of sperm proteins in O. mossambicus correlated with the activation of flagellar motility when sperm were exposed to hypotonic or hypertonic conditions, and that differences in phosphorylation may reflect adaptations to a given osmotic environment. Of the sperm proteins that were dephosphorylated, the phosphorylation pattern of an 18 kDa protein, identified as the superoxide anion scavenger Cu/Zn superoxide dismutase (Cu/Zn SOD), was different in freshwater- and seawater-acclimatised tilapia sperm. Cu/Zn SOD was distributed from the sperm head to the flagellum. Additionally, differences were observed between freshwater and seawater tilapia in the nitration of tyrosine residues (which might be mediated by SOD) in sperm flagellar proteins in response to osmotic shock. These results demonstrate that reactive-oxygen-species-dependent mechanisms contribute to both osmotic tolerance and the activation of flagellar motility.
Collapse
Affiliation(s)
- Masaya Morita
- Sesoko Station, Tropical Biosphere Research Center, University of the Ryukyus, Sesoko, Motobu, Okinawa 907-0227, Japan.
| | | | | | | |
Collapse
|
31
|
Vana L, Kanaan NM, Hakala K, Weintraub ST, Binder LI. Peroxynitrite-induced nitrative and oxidative modifications alter tau filament formation. Biochemistry 2011; 50:1203-12. [PMID: 21210655 PMCID: PMC3040256 DOI: 10.1021/bi101735m] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Tau undergoes numerous posttranslational modifications during the progression of Alzheimer's disease (AD). Some of these changes accelerate tau aggregation, while others are inhibitory. AD-associated inflammation is thought to create oxygen and nitrogen radicals such as peroxynitrite (PN). In vitro, PN can nitrate many proteins, including tau. We have previously demonstrated that tau's ability to form filaments is profoundly affected by treatment with PN and have attributed this inhibition to tyrosine nitration. However, PN is highly reactive and unstable leading to oxidative amino acid modifications through its free radical byproducts. To test whether PN can modify other amino acids in tau via oxidative modifications, a mutant form of the tau protein lacking all tyrosines (5XY → F) was constructed. 5XY → F tau readily forms filaments; however, like wild-type tau the extent of polymerization was greatly reduced following PN treatment. Since 5XY → F tau cannot be nitrated, it was clear that nonnitrative modifications are generated by PN treatment and that these modifications change tau filament formation. Mass spectrometry was used to identify these oxidative alterations in wild-type tau and 5XY → F tau. PN-treated wild-type tau and 5XY → F tau consistently displayed lysine formylation throughout tau in a nonsequence-specific distribution. Lysine formylation likely results from reactive free radical exposure caused by PN treatment. Therefore, our results indicate that PN treatment of proteins in vitro cannot be used to study protein nitration as it likely induces numerous other random oxidative modifications clouding the interpretations of any functional consequences of tyrosine nitration.
Collapse
Affiliation(s)
- Laurel Vana
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States.
| | | | | | | | | |
Collapse
|
32
|
Onufriev MV. Nitrosative stress in the brain: Autoantibodies to nitrotyrosine in the liquor as a potential marker. NEUROCHEM J+ 2010. [DOI: 10.1134/s1819712410030116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
Abstract
Cellular damage occurring under oxidative conditions has been ascribed mainly to the formation of peroxynitrite (ONOOH/ONOO(-)) that originates from the reaction of NO(*) with O(2) (*-). The detrimental effects of peroxynitrite are exacerbated by the reaction with CO(2) that leads to ONOOC(O)O(-), which further decays to the strong oxidant radicals NO(2) (*) and CO(3) (*-). The reaction with CO(2), however, may redirect peroxynitrite specificity. An excessive formation of peroxynitrite represents an important mechanism contributing to the DNA damage, the inactivation of metabolic enzymes, ionic pumps, and structural proteins, and the disruption of cell membranes. Because of its ability to oxidize biomolecules, peroxynitrite is implicated in an increasing list of diseases, including neurodegenerative and cardiovascular disorders, inflammation, pain, autoimmunity, cancer, and aging. However, peroxynitrite displays also protective activities: (i) at high concentrations, it shows anti-viral, anti-microbial, and anti-parasitic actions; and (ii) at low concentrations, it stimulates protective mechanisms in the cardiovascular, nervous, and respiratory systems. The detrimental effects of peroxynitrite and related reactive species are impaired by (pseudo-) enzymatic systems, mainly represented by heme-proteins (e.g., hemoglobin and myoglobin). Here, we report biochemical aspects of peroxynitrite actions being at the root of its biomedical effects.
Collapse
|
34
|
Barber SC, Shaw PJ. Oxidative stress in ALS: key role in motor neuron injury and therapeutic target. Free Radic Biol Med 2010; 48:629-41. [PMID: 19969067 DOI: 10.1016/j.freeradbiomed.2009.11.018] [Citation(s) in RCA: 443] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Revised: 10/16/2009] [Accepted: 11/29/2009] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder characterized by death of motor neurons leading to muscle wasting, paralysis, and death, usually within 2-3 years of symptom onset. The causes of ALS are not completely understood, and the neurodegenerative processes involved in disease progression are diverse and complex. There is substantial evidence implicating oxidative stress as a central mechanism by which motor neuron death occurs, including elevated markers of oxidative damage in ALS patient spinal cord and cerebrospinal fluid and mutations in the antioxidant enzyme superoxide dismutase 1 (SOD1) causing approximately 20% of familial ALS cases. However, the precise mechanism(s) by which mutant SOD1 leads to motor neuron degeneration has not been defined with certainty, and the ultimate trigger for increased oxidative stress in non-SOD1 cases remains unclear. Although some antioxidants have shown potential beneficial effects in animal models, human clinical trials of antioxidant therapies have so far been disappointing. Here, the evidence implicating oxidative stress in ALS pathogenesis is reviewed, along with how oxidative damage triggers or exacerbates other neurodegenerative processes, and we review the trials of a variety of antioxidants as potential therapies for ALS.
Collapse
Affiliation(s)
- Siân C Barber
- Academic Neurology Unit and Sheffield Care & Research Centre for Motor Neuron Disorders, Department of Neuroscience, University of Sheffield, Sheffield S10 2RX, UK
| | | |
Collapse
|
35
|
Perry J, Shin D, Getzoff E, Tainer J. The structural biochemistry of the superoxide dismutases. BIOCHIMICA ET BIOPHYSICA ACTA 2010; 1804:245-62. [PMID: 19914407 PMCID: PMC3098211 DOI: 10.1016/j.bbapap.2009.11.004] [Citation(s) in RCA: 350] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2009] [Revised: 11/04/2009] [Accepted: 11/05/2009] [Indexed: 01/11/2023]
Abstract
The discovery of superoxide dismutases (SODs), which convert superoxide radicals to molecular oxygen and hydrogen peroxide, has been termed the most important discovery of modern biology never to win a Nobel Prize. Here, we review the reasons this discovery has been underappreciated, as well as discuss the robust results supporting its premier biological importance and utility for current research. We highlight our understanding of SOD function gained through structural biology analyses, which reveal important hydrogen-bonding schemes and metal-binding motifs. These structural features create remarkable enzymes that promote catalysis at faster than diffusion-limited rates by using electrostatic guidance. These architectures additionally alter the redox potential of the active site metal center to a range suitable for the superoxide disproportionation reaction and protect against inhibition of catalysis by molecules such as phosphate. SOD structures may also control their enzymatic activity through product inhibition; manipulation of these product inhibition levels has the potential to generate therapeutic forms of SOD. Markedly, structural destabilization of the SOD architecture can lead to disease, as mutations in Cu,ZnSOD may result in familial amyotrophic lateral sclerosis, a relatively common, rapidly progressing and fatal neurodegenerative disorder. We describe our current understanding of how these Cu,ZnSOD mutations may lead to aggregation/fibril formation, as a detailed understanding of these mechanisms provides new avenues for the development of therapeutics against this so far untreatable neurodegenerative pathology.
Collapse
Affiliation(s)
- J.J.P. Perry
- Skaggs Institute for Chemical Biology and Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- The School of Biotechnology, Amrita University, Kollam, Kerala 690525, India
| | - D.S. Shin
- Skaggs Institute for Chemical Biology and Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - E.D. Getzoff
- Skaggs Institute for Chemical Biology and Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - J.A. Tainer
- Skaggs Institute for Chemical Biology and Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Life Sciences Division, Department of Molecular Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| |
Collapse
|
36
|
Aliev G, Palacios HH, Gasimov E, Obrenovich ME, Morales L, Leszek J, Bragin V, Solís Herrera A, Gokhman D. Oxidative Stress Induced Mitochondrial Failure and Vascular Hypoperfusion as a Key Initiator for the Development of Alzheimer Disease. Pharmaceuticals (Basel) 2010; 3:158-187. [PMID: 27713247 PMCID: PMC3991025 DOI: 10.3390/ph3010158] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2009] [Revised: 01/11/2010] [Accepted: 01/14/2010] [Indexed: 11/17/2022] Open
Abstract
Mitochondrial dysfunction may be a principal underlying event in aging, including age-associated brain degeneration. Mitochondria provide energy for basic metabolic processes. Their decay with age impairs cellular metabolism and leads to a decline of cellular function. Alzheimer disease (AD) and cerebrovascular accidents (CVAs) are two leading causes of age-related dementia. Increasing evidence strongly supports the theory that oxidative stress, largely due to reactive oxygen species (ROS), induces mitochondrial damage, which arises from chronic hypoperfusion and is primarily responsible for the pathogenesis that underlies both disease processes. Mitochondrial membrane potential, respiratory control ratios and cellular oxygen consumption decline with age and correlate with increased oxidant production. The sustained hypoperfusion and oxidative stress in brain tissues can stimulate the expression of nitric oxide synthases (NOSs) and brain endothelium probably increase the accumulation of oxidative stress products, which therefore contributes to blood brain barrier (BBB) breakdown and brain parenchymal cell damage. Determining the mechanisms behind these imbalances may provide crucial information in the development of new, more effective therapies for stroke and AD patients in the near future.
Collapse
Affiliation(s)
- Gjumrakch Aliev
- School of Health Science and Healthcare Administration, University of Atlanta, 6685 Peachtree Industrial Blvd., Atlanta, Georgia, 30360, USA.
- Department of Nutrition and Biochemistry, Faculty of Sciences, Javeriana University, Bogotà D.C., Colombia.
- Stress Relief and Memory Training Center, Brooklyn, New York, NY 11235, USA.
| | - Hector H Palacios
- Department of Biology, College of Sciences, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249-1664, USA
| | - Eldar Gasimov
- Department of Cytology, Histology and Embryology, Azerbaijan Medical University, 25 Street Bakhikhanov, Baku AZ10 25, Azerbaijan
| | - Mark E Obrenovich
- Department of Pathology, School of Medicine, Case Western Reserve University, WRB 5301, Cleveland, Ohio, 44106, USA
| | - Ludis Morales
- Department of Nutrition and Biochemistry, Faculty of Sciences, Javeriana University, Bogotà D.C., Colombia
| | - Jerzy Leszek
- Department of Psychiatry, Wroclaw Medical University, 25 St. Kraszewskiego, Wroclaw, 50-229, Poland
| | - Valentin Bragin
- Stress Relief and Memory Training Center, Brooklyn, New York, NY 11235, USA
| | - Arturo Solís Herrera
- Dirección de Investigación y desarrollo, Centro de Estudios de la Fotosíntesis Humana, S.C. López Velarde 108 y 109, Centro, Aguascalientes, Aguascalientes, 20000, México
| | - Dmitry Gokhman
- Department of Mathematics, College of Sciences, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| |
Collapse
|
37
|
Trujillo M, Alvarez B, Souza JM, Romero N, Castro L, Thomson L, Radi R. Mechanisms and Biological Consequences of Peroxynitrite-Dependent Protein Oxidation and Nitration. Nitric Oxide 2010. [DOI: 10.1016/b978-0-12-373866-0.00003-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
38
|
Basso M, Samengo G, Nardo G, Massignan T, D'Alessandro G, Tartari S, Cantoni L, Marino M, Cheroni C, De Biasi S, Giordana MT, Strong MJ, Estevez AG, Salmona M, Bendotti C, Bonetto V. Characterization of detergent-insoluble proteins in ALS indicates a causal link between nitrative stress and aggregation in pathogenesis. PLoS One 2009; 4:e8130. [PMID: 19956584 PMCID: PMC2780298 DOI: 10.1371/journal.pone.0008130] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Accepted: 11/10/2009] [Indexed: 01/12/2023] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is a progressive and fatal motor neuron disease, and protein aggregation has been proposed as a possible pathogenetic mechanism. However, the aggregate protein constituents are poorly characterized so knowledge on the role of aggregation in pathogenesis is limited. Methodology/Principal Findings We carried out a proteomic analysis of the protein composition of the insoluble fraction, as a model of protein aggregates, from familial ALS (fALS) mouse model at different disease stages. We identified several proteins enriched in the detergent-insoluble fraction already at a preclinical stage, including intermediate filaments, chaperones and mitochondrial proteins. Aconitase, HSC70 and cyclophilin A were also significantly enriched in the insoluble fraction of spinal cords of ALS patients. Moreover, we found that the majority of proteins in mice and HSP90 in patients were tyrosine-nitrated. We therefore investigated the role of nitrative stress in aggregate formation in fALS-like murine motor neuron-neuroblastoma (NSC-34) cell lines. By inhibiting nitric oxide synthesis the amount of insoluble proteins, particularly aconitase, HSC70, cyclophilin A and SOD1 can be substantially reduced. Conclusion/Significance Analysis of the insoluble fractions from cellular/mouse models and human tissues revealed novel aggregation-prone proteins and suggests that nitrative stress contribute to protein aggregate formation in ALS.
Collapse
Affiliation(s)
- Manuela Basso
- Dulbecco Telethon Institute, Milan, Italy
- Department of Molecular Biochemistry and Pharmacology, “Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Giuseppina Samengo
- Dulbecco Telethon Institute, Milan, Italy
- Department of Molecular Biochemistry and Pharmacology, “Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Giovanni Nardo
- Dulbecco Telethon Institute, Milan, Italy
- Department of Molecular Biochemistry and Pharmacology, “Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Tania Massignan
- Dulbecco Telethon Institute, Milan, Italy
- Department of Molecular Biochemistry and Pharmacology, “Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Giuseppina D'Alessandro
- Department of Molecular Biochemistry and Pharmacology, “Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Silvia Tartari
- Department of Molecular Biochemistry and Pharmacology, “Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Lavinia Cantoni
- Department of Molecular Biochemistry and Pharmacology, “Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Marianna Marino
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Cristina Cheroni
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Silvia De Biasi
- Department of Biomolecular Sciences and Biotechnology, University of Milan, Milan, Italy
| | | | - Michael J. Strong
- Robarts Research Institute and Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Alvaro G. Estevez
- Burke Medical Research Institute, White Plains, New York, United States of America
| | - Mario Salmona
- Department of Molecular Biochemistry and Pharmacology, “Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Caterina Bendotti
- Department of Neuroscience, “Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Valentina Bonetto
- Dulbecco Telethon Institute, Milan, Italy
- Department of Molecular Biochemistry and Pharmacology, “Mario Negri” Institute for Pharmacological Research, Milan, Italy
- * E-mail:
| |
Collapse
|
39
|
Calabrese V, Cornelius C, Rizzarelli E, Owen JB, Dinkova-Kostova AT, Butterfield DA. Nitric oxide in cell survival: a janus molecule. Antioxid Redox Signal 2009; 11:2717-39. [PMID: 19558211 DOI: 10.1089/ars.2009.2721] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Nitric oxide (NO), plays multiple roles in the nervous system. In addition to regulating proliferation, survival and differentiation of neurons, NO is involved in synaptic activity, neural plasticity, and memory function. Nitric oxide promotes survival and differentiation of neural cells and exerts long-lasting effects through regulation of transcription factors and modulation of gene expression. Signaling by reactive nitrogen species is carried out mainly by targeted modifications of critical cysteine residues in proteins, including S-nitrosylation and S-oxidation, as well as by lipid nitration. NO and other reactive nitrogen species are also involved in neuroinflammation and neurodegeneration, such as in Alzheimer disease, amyotrophic lateral sclerosis, Parkinson disease, multiple sclerosis, Friedreich ataxia, and Huntington disease. Susceptibility to NO and peroxynitrite exposure may depend on factors such as the intracellular reduced glutathione and cellular stress resistance signaling pathways. Thus, neurons, in contrast to astrocytes, appear particularly vulnerable to the effects of nitrosative stress. This article reviews the current understanding of the cytotoxic versus cytoprotective effects of NO in the central nervous system, highlighting the Janus-faced properties of this small molecule. The significance of NO in redox signaling and modulation of the adaptive cellular stress responses and its exciting future perspectives also are discussed.
Collapse
Affiliation(s)
- Vittorio Calabrese
- Department of Chemistry, Biochemistry and Molecular Biology Section, Faculty of Medicine, University of Catania , Catania, Italy.
| | | | | | | | | | | |
Collapse
|
40
|
Lee JR, Kim JK, Lee SJ, Kim KP. Role of protein tyrosine nitration in neurodegenerative diseases and atherosclerosis. Arch Pharm Res 2009; 32:1109-18. [PMID: 19727603 DOI: 10.1007/s12272-009-1802-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Revised: 04/01/2009] [Accepted: 06/25/2009] [Indexed: 02/07/2023]
Abstract
Nitric oxide generates reactive nitrosative species, such as peroxynitrite (ONOO(-)) that may be involved in a number of diseases. ONOO(-) can mediate protein tyrosine nitration which causes structural changes of affected proteins and leads to their inactivation. Various proteomics and immunological methods including mass spectrometry combined with both liquid and 2-D PAGE, and immunodetection have been employed to identify and characterize nitrated proteins from pathological samples. This review presents the pahtobiological roles of the pathogenic posttranslational modification in neurodegenerative diseases and atherosclerosis.
Collapse
Affiliation(s)
- Jung Rok Lee
- Department of Molecular Biotechnology, Konkuk University, Seoul, Korea
| | | | | | | |
Collapse
|
41
|
Molnar KS, Karabacak NM, Johnson JL, Wang Q, Tiwari A, Hayward LJ, Coales SJ, Hamuro Y, Agar JN. A common property of amyotrophic lateral sclerosis-associated variants: destabilization of the copper/zinc superoxide dismutase electrostatic loop. J Biol Chem 2009; 284:30965-73. [PMID: 19635794 DOI: 10.1074/jbc.m109.023945] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
At least 119 mutations in the gene encoding copper/zinc superoxide dismutase (SOD1) cause amyotrophic lateral sclerosis by an unidentified toxic gain of function. We compared the dynamic properties of 13 as-isolated, partially metallated, SOD1 variant enzymes using hydrogen-deuterium exchange. We identified a shared property of these familial amyotrophic lateral sclerosis-related SOD1 variants, namely structural and dynamic change affecting the electrostatic loop (loop VII) of SOD1. Furthermore, SOD1 variants that have severely compromised metal binding affinities demonstrated additional structural and dynamic changes to the zinc-binding loop (loop IV) of SOD1. Although the biological consequences of increased loop VII mobility are not fully understood, this common property is consistent with the hypotheses that SOD1 mutations exert toxicity via aggregation or aberrant association with other cellular constituents.
Collapse
Affiliation(s)
- Kathleen S Molnar
- Department of Chemistry, Volen Center, Brandeis University, Waltham, Massachusetts 02454, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Gunaydin H, Houk KN. Mechanisms of peroxynitrite-mediated nitration of tyrosine. Chem Res Toxicol 2009; 22:894-8. [PMID: 19374346 DOI: 10.1021/tx800463y] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The mechanisms of tyrosine nitration by peroxynitrous acid or nitrosoperoxycarbonate were investigated with the CBS-QB3 method. Either the protonation of peroxynitrite or a reaction with carbon dioxide gives a reactive peroxide intermediate. Peroxynitrous acid-mediated nitration of phenol occurs via unimolecular decomposition to give nitrogen dioxide and hydroxyl radicals. Nitrosoperoxycarbonate also undergoes unimolecular decomposition to give carbonate and nitrogen dioxide radicals. The reactions of tyrosine with the hydroxyl or carbonate radicals give a phenoxy radical intermediate. The reaction of the nitrogen dioxide with this radical intermediate followed by tautomerization gives nitrated tyrosine in both cases. According to CBS-QB3 calculations, the rate-limiting step for the nitration of phenol is the decomposition of peroxynitrous acid or nitrosoperoxycarbonate.
Collapse
Affiliation(s)
- Hakan Gunaydin
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, USA
| | | |
Collapse
|
43
|
Trumbull KA, Beckman JS. A role for copper in the toxicity of zinc-deficient superoxide dismutase to motor neurons in amyotrophic lateral sclerosis. Antioxid Redox Signal 2009; 11:1627-39. [PMID: 19309264 PMCID: PMC2842582 DOI: 10.1089/ars.2009.2574] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Accepted: 03/22/2009] [Indexed: 10/21/2022]
Abstract
In the 16 years since mutations to copper, zinc superoxide dismutase (SOD1) were first linked to familial amyotrophic lateral sclerosis (ALS), a multitude of apparently contradictory results have prevented any general consensus to emerge about the mechanism of toxicity. A decade ago, we showed that the loss of zinc from SOD1 results in the remaining copper in SOD1 to become extremely toxic to motor neurons in culture by a mechanism requiring nitric oxide. The loss of zinc causes SOD1 to become more accessible, more redox reactive, and a better catalyst of tyrosine nitration. Although SOD1 mutant proteins have a modestly reduced affinity for zinc, wild-type SOD1 can be induced to lose zinc by dialysis at slightly acidic pH. Our zinc-deficient hypothesis offers a compelling explanation for how mutant SOD1s have an increased propensity to become selectively toxic to motor neurons and also explains how wild-type SOD1 can be toxic in nonfamilial ALS patients. One critical prediction is that a therapeutic agent directed at zinc-deficient mutant SOD1 could be even more effective in treating sporadic ALS patients. Although transgenic mice experiments have yielded contradictory evidence to the zinc-deficient hypothesis, we will review more recent studies that support a role for copper in ALS. A more careful examination of the role of copper and zinc binding to SOD1 may help counter the growing disillusion in the ALS field about understanding the pathological role of SOD1.
Collapse
Affiliation(s)
- Kari A. Trumbull
- Linus Pauling Institute, Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon
| | - Joseph S. Beckman
- Linus Pauling Institute, Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon
- Environmental Health Science Center, Oregon State University, Corvallis, Oregon
| |
Collapse
|
44
|
Perrot R, Eyer J. Neuronal intermediate filaments and neurodegenerative disorders. Brain Res Bull 2009; 80:282-95. [PMID: 19539727 DOI: 10.1016/j.brainresbull.2009.06.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Revised: 06/05/2009] [Accepted: 06/05/2009] [Indexed: 12/12/2022]
Abstract
Intermediate filaments represent the most abundant cytoskeletal element in mature neurons. Mutations and/or accumulations of neuronal intermediate filament proteins are frequently observed in several human neurodegenerative disorders. Although it is now admitted that disorganization of the neurofilament network may be directly involved in neurodegeneration, certain type of perikaryal intermediate filament aggregates confer protection in motor neuron disease. The use of various mouse models provided a better knowledge of the role played by the disorganization of intermediate filaments in the pathogenesis of neurodegenerative disorders, but the mechanisms leading to the formation of these aggregates remain elusive. Here, we will review some neurodegenerative diseases involving intermediate filaments abnormalities and possible mechanisms susceptible to provoke them.
Collapse
Affiliation(s)
- Rodolphe Perrot
- Department of Anatomy and Physiology of Laval University, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Quebec, Canada
| | | |
Collapse
|
45
|
Nitric Oxide as an Initiator of Brain Lesions During the Development of Alzheimer Disease. Neurotox Res 2009; 16:293-305. [DOI: 10.1007/s12640-009-9066-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 05/16/2009] [Accepted: 05/16/2009] [Indexed: 01/11/2023]
|
46
|
Understanding peroxynitrite biochemistry and its potential for treating human diseases. Arch Biochem Biophys 2009; 484:114-6. [PMID: 19393377 DOI: 10.1016/j.abb.2009.03.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 03/17/2009] [Indexed: 11/20/2022]
|
47
|
D'Orazio M, Cervoni L, Giartosio A, Rotilio G, Battistoni A. Thermal stability and redox properties of M. tuberculosis CuSOD. Arch Biochem Biophys 2009; 486:119-24. [PMID: 19383490 DOI: 10.1016/j.abb.2009.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Revised: 04/07/2009] [Accepted: 04/11/2009] [Indexed: 11/19/2022]
Abstract
The superoxide dismutase from Mycobacterium tuberculosis is the only Cu-containing superoxide dismutase that lacks zinc in the active site. To explore the structural properties of this unusual enzyme, we have investigated its stability by differential scanning calorimetry. We have found that the holo-enzyme is significantly more stable than the apo-protein or the partially metallated enzyme, but that its melting temperature is markedly lower than that of all the other characterized eukaryotic and prokaryotic Cu,Zn superoxide dismutases. We have also observed that, unlike the zinc-free eukaryotic or bacterial enzymes, the active site copper of the mycobacterial enzyme is not reduced by ascorbate, confirming that its redox properties are comparable to those typical of the enzymes containing zinc in the active site. Our findings highlight the role of zinc in conferring stability to Cu,Zn superoxide dismutases and indicate that the structural rearrangements observed in M. tuberculosis Cu,SOD compensate for the absence of zinc in achieving a fully active enzyme.
Collapse
|
48
|
Tavori H, Aviram M, Khatib S, Musa R, Nitecki S, Hoffman A, Vaya J. Human carotid atherosclerotic plaque increases oxidative state of macrophages and low-density lipoproteins, whereas paraoxonase 1 (PON1) decreases such atherogenic effects. Free Radic Biol Med 2009; 46:607-15. [PMID: 19103284 DOI: 10.1016/j.freeradbiomed.2008.11.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Revised: 11/13/2008] [Accepted: 11/16/2008] [Indexed: 02/06/2023]
Abstract
Human atherosclerotic plaque contains a variety of oxidized lipids, which can facilitate further oxidation. Paraoxonase 1 (PON1) is a high-density lipoprotein (HDL)-associated esterase (lipolactonase), exhibiting antiatherogenic properties. The aims of the present study were to examine the oxidizing potency of the human carotid plaque lipid extract (LE), and the antiatherogenic role of PON1 on LE oxidation competence. Human carotid plaques were extracted by organic solvent, and the extract was incubated with lipoprotein particles, with macrophages, or with probes sensitive to oxidative stress, with or without preincubation with PON1, followed by oxidative-stress assessment. Our findings imply that the LE oxidized LDL, macrophages, and exogenous probes and decreases HDL-mediated cholesterol efflux from macrophages, in a dose-dependent manner. Incubation of PON1 with LE significantly affects LE composition, reduces LE atherogenic properties, and decreases the extract's total peroxide concentration by 44%, macrophage oxidation by 25%, and probe oxidation by up to 52%. We conclude that these results expand our understanding of how the plaque itself accelerates atherogenesis and provides an important mechanism for attenuation of atherosclerosis development by the antioxidant action of PON1 on the atherosclerotic plaque.
Collapse
Affiliation(s)
- Hagai Tavori
- MIGAL - Galilee Technology Center, Kiryat Shmona, Tel Hai College, Israel
| | | | | | | | | | | | | |
Collapse
|
49
|
Soulère L, Bernard J. Design, solid phase synthesis and evaluation of cationic ferrocenoyl peptide bioconjugates as potential antioxidant enzyme mimics. Bioorg Med Chem Lett 2009; 19:1173-6. [DOI: 10.1016/j.bmcl.2008.12.084] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Revised: 12/16/2008] [Accepted: 12/19/2008] [Indexed: 11/16/2022]
|
50
|
Age related and hypothyroidism related changes on the stoichiometry of neurofilament subunits in the developing rat brain. Int J Dev Neurosci 2009; 27:257-61. [DOI: 10.1016/j.ijdevneu.2008.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Accepted: 12/23/2008] [Indexed: 11/19/2022] Open
|