1
|
Üremiş N, Üremiş MM. Oxidative/Nitrosative Stress, Apoptosis, and Redox Signaling: Key Players in Neurodegenerative Diseases. J Biochem Mol Toxicol 2025; 39:e70133. [PMID: 39799559 PMCID: PMC11725306 DOI: 10.1002/jbt.70133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 01/15/2025]
Abstract
Neurodegenerative diseases are significant health concerns that have a profound impact on the quality and duration of life for millions of individuals. These diseases are characterized by pathological changes in various brain regions, specific genetic mutations associated with the disease, deposits of abnormal proteins, and the degeneration of neurological cells. As neurodegenerative disorders vary in their epidemiological characteristics and vulnerability of neurons, treatment of these diseases is usually aimed at slowing disease progression. The heterogeneity of genetic and environmental factors involved in the process of neurodegeneration makes current treatment methods inadequate. However, the existence of common molecular mechanisms in the pathogenesis of these diseases may allow the development of new targeted therapeutic strategies. Oxidative and nitrosative stress damages membrane components by accumulating ROS and RNS and disrupting redox balance. This process results in the induction of apoptosis, which is important in the pathogenesis of neurodegenerative diseases through oxidative stress. Studies conducted using postmortem human samples, animal models, and cell cultures have demonstrated that oxidative stress, nitrosative stress, and apoptosis are crucial factors in the development of diseases such as Alzheimer's, Parkinson's, Multiple Sclerosis, amyotrophic lateral sclerosis, and Huntington's disease. The excessive production of reactive oxygen and nitrogen species, elevated levels of free radicals, heightened mitochondrial stress, disturbances in energy metabolism, and the oxidation and nitrosylation of cellular macromolecules are recognized as triggers for neuronal cell death. Challenges in managing and treating neurodegenerative diseases require a better understanding of this field at the molecular level. Therefore, this review elaborates on the molecular mechanisms by which oxidative and nitrosative stress are involved in neuronal apoptosis.
Collapse
Affiliation(s)
- Nuray Üremiş
- Department of Medical BiochemistryFaculty of Medicine, Kahramanmaraş Sütçü İmam UniversityKahramanmaraşTurkey
| | - Muhammed Mehdi Üremiş
- Department of Medical BiochemistryFaculty of Medicine, Kahramanmaraş Sütçü İmam UniversityKahramanmaraşTurkey
| |
Collapse
|
2
|
Martin LJ, Koh SJ, Price A, Park D, Kim BW. Nuclear Localization of Human SOD1 in Motor Neurons in Mouse Model and Patient Amyotrophic Lateral Sclerosis: Possible Links to Cholinergic Phenotype, NADPH Oxidase, Oxidative Stress, and DNA Damage. Int J Mol Sci 2024; 25:9106. [PMID: 39201793 PMCID: PMC11354607 DOI: 10.3390/ijms25169106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 09/03/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal disease that causes degeneration of motor neurons (MNs) and paralysis. ALS can be caused by mutations in the gene that encodes copper/zinc superoxide dismutase (SOD1). SOD1 is known mostly as a cytosolic antioxidant protein, but SOD1 is also in the nucleus of non-transgenic (tg) and human SOD1 (hSOD1) tg mouse MNs. SOD1's nuclear presence in different cell types and subnuclear compartmentations are unknown, as are the nuclear functions of SOD1. We examined hSOD1 nuclear localization and DNA damage in tg mice expressing mutated and wildtype variants of hSOD1 (hSOD1-G93A and hSOD1-wildtype). We also studied ALS patient-derived induced pluripotent stem (iPS) cells to determine the nuclear presence of SOD1 in undifferentiated and differentiated MNs. In hSOD1-G93A and hSOD1-wildtype tg mice, choline acetyltransferase (ChAT)-positive MNs had nuclear hSOD1, but while hSOD1-wildtype mouse MNs also had nuclear ChAT, hSOD1-G93A mouse MNs showed symptom-related loss of nuclear ChAT. The interneurons had preserved parvalbumin nuclear positivity in hSOD1-G93A mice. hSOD1-G93A was seen less commonly in spinal cord astrocytes and, notably, oligodendrocytes, but as the disease emerged, the oligodendrocytes had increased mutant hSOD1 nuclear presence. Brain and spinal cord subcellular fractionation identified mutant hSOD1 in soluble nuclear extracts of the brain and spinal cord, but mutant hSOD1 was concentrated in the chromatin nuclear extract only in the spinal cord. Nuclear extracts from mutant hSOD1 tg mouse spinal cords had altered protein nitration, footprinting peroxynitrite presence, and the intact nuclear extracts had strongly increased superoxide production as well as the active NADPH oxidase marker, p47phox. The comet assay showed that MNs from hSOD1-G93A mice progressively (6-14 weeks of age) accumulated DNA single-strand breaks. Ablation of the NCF1 gene, encoding p47phox, and pharmacological inhibition of NADPH oxidase with systemic treatment of apocynin (10 mg/kg, ip) extended the mean lifespan of hSOD1-G93A mice by about 25% and mitigated genomic DNA damage progression. In human postmortem CNS, SOD1 was found in the nucleus of neurons and glia; nuclear SOD1 was increased in degenerating neurons in ALS cases and formed inclusions. Human iPS cells had nuclear SOD1 during directed differentiation to MNs, but mutant SOD1-expressing cells failed to establish wildtype MN nuclear SOD1 levels. We conclude that SOD1 has a prominent nuclear presence in the central nervous system, perhaps adopting aberrant contexts to participate in ALS pathobiology.
Collapse
Affiliation(s)
- Lee J. Martin
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA (D.P.)
- Pathobiology Graduate Program, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA
- Department of Anesthesiology & Critical Care Medicine, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA
| | - Shannon J. Koh
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA (D.P.)
- Texas Health Presbyterian Hospital, Dallas, TX 75231, USA
| | - Antionette Price
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA (D.P.)
| | - Dongseok Park
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA (D.P.)
| | - Byung Woo Kim
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA (D.P.)
- Pathobiology Graduate Program, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD 21205-2196, USA
| |
Collapse
|
3
|
Sharma S, Deep S. Inhibition of fibril formation by polyphenols: molecular mechanisms, challenges, and prospective solutions. Chem Commun (Camb) 2024; 60:6717-6727. [PMID: 38835221 DOI: 10.1039/d4cc00822g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Fibril formation is a key feature in neurodegenerative diseases like Alzheimer's, Parkinson's, and systemic amyloidosis. Polyphenols, found in plant-based foods, show promise in inhibiting fibril formation and disrupting disease progression. The ability of polyphenols to break the amyloid fibrils of many disease-linked proteins has been tested in numerous studies. Polyphenols have their distinctive mechanism of action. They behave differently on various events in the aggregation pathway. Their action also differs for different proteins. Some polyphenols only inhibit the formation of fibrils whereas others break the preformed fibrils. Some break the fibrils into smaller species, and some change them to other morphologies. This article delves into the intricate molecular mechanisms underlying the inhibitory effects of polyphenols on fibrillogenesis, shedding light on their interactions with amyloidogenic proteins and the disruption of fibril assembly pathways. However, addressing the challenges associated with solubility, stability, and bioavailability of polyphenols is crucial. The current strategies involve nanotechnology to improve the solubility and bioavailability, thus showing the potential to enhance the efficacy of polyphenols as therapeutics. Advancements in structural biology, computational modeling, and biophysics have provided insights into polyphenol-fibril interactions, offering hope for novel therapies for neurodegenerative diseases and amyloidosis.
Collapse
Affiliation(s)
- Shilpa Sharma
- Department of Chemistry and Biochemistry, University of Wisconsin, Milwaukee, Wisconsin, USA
| | - Shashank Deep
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi, India.
| |
Collapse
|
4
|
Wang Z, Yan M, Yu M, Zhang G, Fang W, Yu F. A Fluorescent Probe with Zwitterionic ESIPT Feature for Ratiometric Monitoring of Peroxynitrite In Vitro and In Vivo. Anal Chem 2024; 96:3600-3608. [PMID: 38372498 DOI: 10.1021/acs.analchem.3c05718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Peroxynitrite (ONOO-), as a short-term reactive biological oxidant, could lead to a series of effects in various physiological and pathological processes due to its subtle concentration changes. In vivo monitoring of ONOO- and relevant physiological processes is urgently required. Herein, we describe a novel fluorescent probe termed HBT-Fl-BnB for the ratiometric detection of ONOO- in vitro and in vivo. The probe consists of an HBT core with Fl groups at the ortho and para positions responding to the zwitterionic excited-state intramolecular proton-transfer (zwitterionic ESIPT) process and a boronic acid pinacol ester with dual roles that block the zwitterionic ESIPT and recognize ONOO-. Thanks to the specificity as well as low cytotoxicity, success in imaging of endogenous and exogenous ONOO- in living cells by HBT-Fl-BnB was obtained. Additionally, the applicability of HBT-Fl-BnB to tracking the abnormal expression of ONOO- in vivo induced by inactivated Escherichia coli was also explored. This is the first report of a fluorescent probe for ONOO- sensing via a zwitterionic ESIPT mechanism.
Collapse
Affiliation(s)
- Zhenkai Wang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, 159 Longpan Road, Nanjing 210037,China
- Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Miao Yan
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, 159 Longpan Road, Nanjing 210037,China
| | - Miaomiao Yu
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, 159 Longpan Road, Nanjing 210037,China
| | - Gang Zhang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, 159 Longpan Road, Nanjing 210037,China
| | - Weiwei Fang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, 159 Longpan Road, Nanjing 210037,China
| | - Fabiao Yu
- Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
5
|
Cunha-Oliveira T, Montezinho L, Simões RF, Carvalho M, Ferreiro E, Silva FSG. Mitochondria: A Promising Convergent Target for the Treatment of Amyotrophic Lateral Sclerosis. Cells 2024; 13:248. [PMID: 38334639 PMCID: PMC10854804 DOI: 10.3390/cells13030248] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 02/10/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by the progressive loss of motor neurons, for which current treatment options are limited. Recent studies have shed light on the role of mitochondria in ALS pathogenesis, making them an attractive therapeutic intervention target. This review contains a very comprehensive critical description of the involvement of mitochondria and mitochondria-mediated mechanisms in ALS. The review covers several key areas related to mitochondria in ALS, including impaired mitochondrial function, mitochondrial bioenergetics, reactive oxygen species, metabolic processes and energy metabolism, mitochondrial dynamics, turnover, autophagy and mitophagy, impaired mitochondrial transport, and apoptosis. This review also highlights preclinical and clinical studies that have investigated various mitochondria-targeted therapies for ALS treatment. These include strategies to improve mitochondrial function, such as the use of dichloroacetate, ketogenic and high-fat diets, acetyl-carnitine, and mitochondria-targeted antioxidants. Additionally, antiapoptotic agents, like the mPTP-targeting agents minocycline and rasagiline, are discussed. The paper aims to contribute to the identification of effective mitochondria-targeted therapies for ALS treatment by synthesizing the current understanding of the role of mitochondria in ALS pathogenesis and reviewing potential convergent therapeutic interventions. The complex interplay between mitochondria and the pathogenic mechanisms of ALS holds promise for the development of novel treatment strategies to combat this devastating disease.
Collapse
Affiliation(s)
- Teresa Cunha-Oliveira
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Liliana Montezinho
- Center for Investigation Vasco da Gama (CIVG), Escola Universitária Vasco da Gama, 3020-210 Coimbra, Portugal;
| | - Rui F. Simões
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Marcelo Carvalho
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Elisabete Ferreiro
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Filomena S. G. Silva
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Mitotag Lda, Biocant Park, 3060-197 Cantanhede, Portugal
| |
Collapse
|
6
|
Huang YN, Greig NH, Huang PS, Chiang YH, Hoffer A, Yang CH, Tweedie D, Chen Y, Ou JC, Wang JY. Pomalidomide Improves Motor Behavioral Deficits and Protects Cerebral Cortex and Striatum Against Neurodegeneration Through a Reduction of Oxidative/Nitrosative Damages and Neuroinflammation After Traumatic Brain Injury. Cell Transplant 2024; 33:9636897241237049. [PMID: 38483119 PMCID: PMC10943757 DOI: 10.1177/09636897241237049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 03/18/2024] Open
Abstract
Neuronal damage resulting from traumatic brain injury (TBI) causes disruption of neuronal projections and neurotransmission that contribute to behavioral deficits. Cellular generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) is an early event following TBI. ROS often damage DNA, lipids, proteins, and carbohydrates while RNS attack proteins. The products of lipid peroxidation 4-hydroxynonenal (4-HNE) and protein nitration 3-nitrotyrosine (3-NT) are often used as indicators of oxidative and nitrosative damages, respectively. Increasing evidence has shown that striatum is vulnerable to damage from TBI with a disturbed dopamine neurotransmission. TBI results in neurodegeneration, oxidative stress, neuroinflammation, neuronal apoptosis, and autophagy in the striatum and contribute to motor or behavioral deficits. Pomalidomide (Pom) is a Food and Drug Administration (FDA)-approved immunomodulatory drug clinically used in treating multiple myeloma. We previously showed that Pom reduces neuroinflammation and neuronal death induced by TBI in rat cerebral cortex. Here, we further compared the effects of Pom in cortex and striatum focusing on neurodegeneration, oxidative and nitrosative damages, as well as neuroinflammation following TBI. Sprague-Dawley rats subjected to a controlled cortical impact were used as the animal model of TBI. Systemic administration of Pom (0.5 mg/kg, intravenous [i.v.]) at 5 h post-injury alleviated motor behavioral deficits, contusion volume at 24 h after TBI. Pom alleviated TBI-induced neurodegeneration stained by Fluoro-Jade C in both cortex and striatum. Notably, Pom treatment reduces oxidative and nitrosative damages in cortex and striatum and is more efficacious in striatum (93% reduction in 4-HNE-positive and 84% reduction in 3-NT-positive neurons) than in cerebral cortex (42% reduction in 4-HNE-positive and 55% reduction in 3-NT-positive neurons). In addition, Pom attenuated microgliosis, astrogliosis, and elevations of proinflammatory cytokines in cortical and striatal tissue. We conclude that Pom may contribute to improved motor behavioral outcomes after TBI through targeting oxidative/nitrosative damages and neuroinflammation.
Collapse
Affiliation(s)
- Ya-Ni Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei
- Department of Nursing, Hsin Sheng Junior College of Medical Care and Management, Taoyuan City
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Pen-Sen Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei
| | - Yung-Hsiao Chiang
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei
- Neuroscience Research Center, Taipei Medical University, Taipei
| | - Alan Hoffer
- Department of Neurosurgery, University Hospitals of Cleveland, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Chih-Hao Yang
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ying Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei
| | - Ju-Chi Ou
- Neuroscience Research Center, Taipei Medical University, Taipei
| | - Jia-Yi Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei
- Neuroscience Research Center, Taipei Medical University, Taipei
| |
Collapse
|
7
|
Weemering DN, Midei M, Milner P, Gopalakrishnan V, Kumar A, Dannenberg AJ, Bunte TM, Foucher J, Ingre C, Ķēniņa V, Rallmann K, van den Berg LH, van Eijk RPA. A randomized, double-blind, placebo-controlled phase 2 study to assess safety, tolerability, and efficacy of RT001 in patients with amyotrophic lateral sclerosis. Eur J Neurol 2023; 30:3722-3731. [PMID: 37550954 DOI: 10.1111/ene.16020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/20/2023] [Accepted: 07/30/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND AND PURPOSE RT001 is a deuterated synthetic homologue of linoleic acid, which makes membrane polyunsaturated fatty acids resistant to lipid peroxidation, a process involved in motor neuron degeneration in amyotrophic lateral sclerosis (ALS). METHODS We conducted a randomized, multicenter, placebo-controlled clinical trial. Patients with ALS were randomly allocated to receive either RT001 or placebo for 24 weeks. After the double-blind period, all patients received RT001 during an open-label phase for 24 weeks. The primary outcome measures were safety and tolerability. Key efficacy outcomes included the ALS Functional Rating Scale (ALSFRS-R), percent predicted slow vital capacity, and plasma neurofilament light chain concentration. RESULTS In total, 43 patients (RT001 = 21; placebo = 22) were randomized. RT001 was well tolerated; one patient required dose reduction due to adverse events (AEs). Numerically, there were more AEs in the RT001 group compared to the placebo group (71% versus 55%, p = 0.35), with gastrointestinal symptoms being the most common (43% in RT001, 27% in placebo, p = 0.35). Two patients in the RT001 group experienced a serious AE, though unrelated to treatment. The least-squares mean difference in ALSFRS-R total score at week 24 of treatment was 1.90 (95% confidence interval = -1.39 to 5.19) in favor of RT001 (p = 0.25). The directions of other efficacy outcomes favored RT001 compared to placebo, although no inferential statistics were performed. CONCLUSIONS Initial data indicate that RT001 is safe and well tolerated. Given the exploratory nature of the study, a larger clinical trial is required to evaluate its efficacy.
Collapse
Affiliation(s)
- Daphne N Weemering
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Mark Midei
- BioJiva, LLC, Los Altos, California, USA
| | | | | | - Anil Kumar
- BioJiva, LLC, Los Altos, California, USA
| | | | - Tommy M Bunte
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Juliette Foucher
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, ME Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Ingre
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, ME Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Viktorija Ķēniņa
- Rare Neurological Disease Centre, Riga Stradinš Clinical University Hospital, Riga, Latvia
- Department of Biology and Microbiology, Riga Stradinš University, Riga, Latvia
| | - Karin Rallmann
- Department of Neurology and Neurosurgery, University of Tartu, Tartu, Estonia
| | - Leonard H van den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ruben P A van Eijk
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
- Biostatistics & Research Support, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
8
|
Corcia P, Vourc'h P, Bernard E, Cassereau J, Codron P, Fleury MC, Guy N, Mouzat K, Pradat PF, Soriani MH, Couratier P. French National Protocol for genetic of amyotrophic lateral sclerosis. Rev Neurol (Paris) 2023; 179:1020-1029. [PMID: 37735015 DOI: 10.1016/j.neurol.2023.05.005] [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: 04/06/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 09/23/2023]
Abstract
Relationships between genes and amyotrophic lateral sclerosis (ALS) have been widely accepted since the first studies highlighting pathogenic mutations in the SOD1 gene 30years ago. Over the last three decades, scientific literature has clearly highlighted the central role played by genetic factors in the disease, in both clinics and pathophysiology, as well as in therapeutics. This implies that health professionals who care for patients with ALS are increasingly faced with patients and relatives eager to have answers to questions related to the role of genetic factors in the occurrence of the disease and the risk for their relatives to develop ALS. In order to address these public health issues, the French ALS network FILSLAN proposed to the Haute Autorité de santé (HAS) the drafting of a French National Protocol (PNDS) on ALS genetics. This PNDS was developed according to the "method for developing a national diagnosis and care protocol for rare diseases" published by the HAS in 2012 (methodological guide for PNDS available on the HAS website: http://www.has-sante.fr/). This document aims to provide the most recent data on the role of genes in ALS and to detail the implications for diagnosis and care.
Collapse
Affiliation(s)
- P Corcia
- CRMR SLA, CHRU Bretonneau, Tours, France; UMR 1253 iBrain, Tours, France.
| | - P Vourc'h
- UMR 1253 iBrain, Tours, France; Laboratoire de biochimie et biologie moléculaire, CHRU Bretonneau, Tours, France
| | | | | | - P Codron
- CRMR SLA, CHU d'Angers, Angers, France
| | - M-C Fleury
- CRC SLA, CHU de Strasbourg, Strasbourg, France
| | - N Guy
- CRC SLA, CHU de Clermont-Ferrand, Clermont-Ferrand, France
| | - K Mouzat
- Laboratoire de biochimie et biologie moléculaire, CHU de Nîmes, Nîmes, France
| | - P-F Pradat
- CRMR SLA, CHU Pitié-Salpêtrière, Paris, France
| | | | | |
Collapse
|
9
|
Geethika M, Singh N, Kumar S, Kumar SKN, Mugesh G. A Redox Modulatory SOD Mimetic Nanozyme Prevents the Formation of Cytotoxic Peroxynitrite and Improves Nitric Oxide Bioavailability in Human Endothelial Cells. Adv Healthc Mater 2023; 12:e2300621. [PMID: 37524524 DOI: 10.1002/adhm.202300621] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/22/2023] [Indexed: 08/02/2023]
Abstract
The endothelium-derived signalling molecule nitric oxide (NO) in addition to controlling multifarious servo-regulatory functions, suppresses key processes in vascular lesion formation and prevents atherogenesis and other vascular abnormalities. The conversion of NO into cytotoxic and powerful oxidant peroxynitrite (ONOO- ) in a superoxide (O2 .- )-rich environment has emerged as a major reason for reduced NO levels in vascular walls, leading to endothelial dysfunction and cardiovascular complications. So, designing superoxide dismutase (SOD) mimetics that can selectively catalyze the dismutation of O2 .- in the presence of NO, considering their rapid reaction is challenging and is of therapeutic relevance. Herein, the authors report that SOD mimetic cerium vanadate (CeVO4 ) nanozymes effectively regulate the bioavailability of both NO and O2 .- , the two vital constitutive molecules of vascular endothelium, even in the absence of cellular SOD enzyme. The nanozymes optimally modulate the O2 .- level in endothelial cells under oxidative stress conditions and improve endogenously generated NO levels by preventing the formation of ONOO- . Furthermore, nanoparticles exhibit size- and morphology-dependent uptake into the cells and internalize via the clathrin-mediated endocytosis pathway. Intravenous administration of CeVO4 nanoparticles in mice caused no definite organ toxicity and unaltered haematological and biochemical parameters, indicating their biosafety and potential use in biological applications.
Collapse
Affiliation(s)
- Motika Geethika
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Namrata Singh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Sagar Kumar
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | | | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
10
|
Djordjevic G, Milosevic V, Ljubisavljevic S, Stojanovic I, Stojanov A. Values of Nitric Oxide and Superoxide Dismutase in Cerebrospinal Fluid of Patients with Sporadic Amyotrophic Lateral Sclerosis. Neurol India 2023; 71:742-747. [PMID: 37635508 DOI: 10.4103/0028-3886.383853] [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: 08/29/2023]
Abstract
Introduction Neurons are highly energy-dependent and highly specialized cells, showing great sensitivity to oxidative stress (OS). Nitric oxide (NO) and its oxidation products play a central role in neurodegeneration. This study aimed to contribute to the further elucidation of the role of OS in the pathogenesis of amyotrophic lateral sclerosis (ALS). Methods We assessed NO and superoxide dismutase (SOD) levels in cerebrospinal fluid (CSF) of 24 sporadic ALS (sALS) patients (13 of them presented with spinal form while 11 patients had bulbar form) and 20 controls (CG). Results The obtained SOD levels in sALS patients were lower than those in CG (p < 0.001), while NO showed higher levels compared to CG (p < 0.001). Observed separately, there were no significant differences in the levels of NO and SOD in CSF between patients about their clinical presentations (p > 0.05). There were significant negative correlations between SOD and NO levels in all sALS patients (r = 0.31, P = 0.025). Significant correlation between SOD and functional rating scale as well as disease progression index was recorded in patients with sALS (r = 0.618. r = 0.425, P < 0.01), while NO levels were significantly associated with disease progression only (r = 0.348, P < 0.01). Conclusion The data presented clearly support the role of impaired oxidant/antioxidant balance in the pathogenesis of ALS, where NO overproduction and decreased SOD defense activity seem to be particularly involved. The CSF SOD and NO level might serve as useful biomarkers for functional disorder and progression of the disease.
Collapse
Affiliation(s)
- Gordana Djordjevic
- Faculty of Medicine, University of Nis; Clinic of Neurology, University Clinical Center Nis, Nis, Serbia
| | - Vuk Milosevic
- Faculty of Medicine, University of Nis; Clinic of Neurology, University Clinical Center Nis, Nis, Serbia
| | - Srdjan Ljubisavljevic
- Faculty of Medicine, University of Nis; Clinic of Neurology, University Clinical Center Nis, Nis, Serbia
| | | | | |
Collapse
|
11
|
Valles SL, Singh SK, Campos-Campos J, Colmena C, Campo-Palacio I, Alvarez-Gamez K, Caballero O, Jorda A. Functions of Astrocytes under Normal Conditions and after a Brain Disease. Int J Mol Sci 2023; 24:ijms24098434. [PMID: 37176144 PMCID: PMC10179527 DOI: 10.3390/ijms24098434] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
In the central nervous system (CNS) there are a greater number of glial cells than neurons (between five and ten times more). Furthermore, they have a greater number of functions (more than eight functions). Glia comprises different types of cells, those of neural origin (astrocytes, radial glia, and oligodendroglia) and differentiated blood monocytes (microglia). During ontogeny, neurons develop earlier (at fetal day 15 in the rat) and astrocytes develop later (at fetal day 21 in the rat), which could indicate their important and crucial role in the CNS. Analysis of the phylogeny reveals that reptiles have a lower number of astrocytes compared to neurons and in humans this is reversed, as there have a greater number of astrocytes compared to neurons. These data perhaps imply that astrocytes are important and special cells, involved in many vital functions, including memory, and learning processes. In addition, astrocytes are involved in different mechanisms that protect the CNS through the production of antioxidant and anti-inflammatory proteins and they clean the extracellular environment and help neurons to communicate correctly with each other. The production of inflammatory mediators is important to prevent changes in brain homeostasis. On the contrary, excessive, or continued production appears as a characteristic element in many diseases, such as Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and in neurodevelopmental diseases, such as bipolar disorder, schizophrenia, and autism. Furthermore, different drugs and techniques have been developed to reverse oxidative stress and/or excess of inflammation that occurs in many CNS diseases, but much remains to be investigated. This review attempts to highlight the functional relevance of astrocytes in normal and neuropathological conditions by showing the molecular and cellular mechanisms of their role in the CNS.
Collapse
Affiliation(s)
- Soraya L Valles
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Sandeep Kumar Singh
- Indian Scientific Education and Technology Foundation, Lucknow 226002, India
| | - Juan Campos-Campos
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
- Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain
| | - Carlos Colmena
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Ignacio Campo-Palacio
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Kenia Alvarez-Gamez
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Oscar Caballero
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
- Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain
| | - Adrian Jorda
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
- Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
12
|
Olufunmilayo EO, Gerke-Duncan MB, Holsinger RMD. Oxidative Stress and Antioxidants in Neurodegenerative Disorders. Antioxidants (Basel) 2023; 12:antiox12020517. [PMID: 36830075 PMCID: PMC9952099 DOI: 10.3390/antiox12020517] [Citation(s) in RCA: 179] [Impact Index Per Article: 89.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Neurodegenerative disorders constitute a substantial proportion of neurological diseases with significant public health importance. The pathophysiology of neurodegenerative diseases is characterized by a complex interplay of various general and disease-specific factors that lead to the end point of neuronal degeneration and loss, and the eventual clinical manifestations. Oxidative stress is the result of an imbalance between pro-oxidant species and antioxidant systems, characterized by an elevation in the levels of reactive oxygen and reactive nitrogen species, and a reduction in the levels of endogenous antioxidants. Recent studies have increasingly highlighted oxidative stress and associated mitochondrial dysfunction to be important players in the pathophysiologic processes involved in neurodegenerative conditions. In this article, we review the current knowledge of the general effects of oxidative stress on the central nervous system, the different specific routes by which oxidative stress influences the pathophysiologic processes involved in Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis and Huntington's disease, and how oxidative stress may be therapeutically reversed/mitigated in order to stall the pathological progression of these neurodegenerative disorders to bring about clinical benefits.
Collapse
Affiliation(s)
- Edward O. Olufunmilayo
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Department of Medicine, University College Hospital, Queen Elizabeth Road, Oritamefa, Ibadan 5116, PMB, Nigeria
| | - Michelle B. Gerke-Duncan
- Education Innovation, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - R. M. Damian Holsinger
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Correspondence:
| |
Collapse
|
13
|
Tarannum A, Arif Z, Mustafa M, Abul Qais F, Habib S, Uddin M, Alam K. Studies on the synergistic action of methylglyoxal and peroxynitrite on structure and function of human serum albumin. J Biomol Struct Dyn 2023; 41:67-80. [PMID: 34842044 DOI: 10.1080/07391102.2021.2003865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Albumin, an important serum protein, is continuously exposed to various oxidizing/nitrating and glycating agents. Depending upon the nature/concentration of reactive species present, the protein may be glycated, oxidized/nitroxidized or glyco-nitro-oxidized. Peroxynitrite is a powerful nitroxidant and has been reported to damage a wide array of macromolecules. On the other hand, methylglyoxal is a very strong reactive dicarbonyl and a potent precursor for the formation of advanced glycation end products under pathological conditions. In certain pathological conditions albumin may be modified by peroxynitrite and methylglyoxal simultaneously. There is dearth of literature suggests that structural/conformational and functional alteration in albumin upon glycation and oxidation/nitroxidation, however the alterations produced by glyco-nitro-oxidation has not yet been explored. Therefore, in this study, simultaneous effect of glycation and nitroxidation on the structure and conformation, vis-a-vis function of albumin was explored. Glyco-nitro-oxidized albumin showed decreased free amino acid content together with decreased affinity of albumin towards cobalt. Molecular docking model and molecular dynamic simulations showed close interaction and formation of stable complexes between methylglyoxal, peroxynitrite and albumin. Formation of carboxymethyl lysine and 3-nitrotyrosine in glyco-nitro-oxidized albumin were confirmed by MALDI-TOF MS and UP-LC MS. Aggregate formation in glyco-nitro-oxidized albumin was visualized by transmission electron microscopy. On the basis of these results, it may be speculated that, albumin modified with endogenously generated methylglyoxal and peroxynitrite might be a driving factor in the progression of heightened inflammatory autoimmune responses. The work presents a ground to study the role of glyco-nitro-oxidized albumin in the pathogenesis and progression of various autoimmune diseases including rheumatoid arthritis. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Akhlas Tarannum
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Zarina Arif
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Mohd Mustafa
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Faizan Abul Qais
- Dept of Agricultural Microbiology, Faculty of Agricultural Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Safia Habib
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Moin Uddin
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Khursheed Alam
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| |
Collapse
|
14
|
Steinert JR, Amal H. The contribution of an imbalanced redox signalling to neurological and neurodegenerative conditions. Free Radic Biol Med 2023; 194:71-83. [PMID: 36435368 DOI: 10.1016/j.freeradbiomed.2022.11.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
Nitric oxide and other redox active molecules such as oxygen free radicals provide essential signalling in diverse neuronal functions, but their excess production and insufficient scavenging induces cytotoxic redox stress which is associated with numerous neurodegenerative and neurological conditions. A further component of redox signalling is mediated by a homeostatic regulation of divalent metal ions, the imbalance of which contributes to neuronal dysfunction. Additional antioxidant molecules such as glutathione and enzymes such as super oxide dismutase are involved in maintaining a physiological redox status within neurons. When cellular processes are perturbed and generation of free radicals overwhelms the antioxidants capacity of the neurons, a resulting redox damage leads to neuronal dysfunction and cell death. Cellular sources for production of redox-active molecules may include NADPH oxidases, mitochondria, cytochrome P450 and nitric oxide (NO)-generating enzymes, such as endothelial, neuronal and inducible NO synthases. Several neurodegenerative and developmental neurological conditions are associated with an imbalanced redox state as a result of neuroinflammatory processes leading to nitrosative and oxidative stress. Ongoing research aims at understanding the causes and consequences of such imbalanced redox homeostasis and its role in neuronal dysfunction.
Collapse
Affiliation(s)
- Joern R Steinert
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham, NG7 2NR, UK.
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
15
|
Yerton M, Winter A, Kostov A, Lieberman C, Gelevski D, Weber H, Doyle M, Kane G, Parikh N, Burke KM, Rohrer M, Stirrat T, Bruno M, Hochman A, Luppino S, Scalia J, Skoniecki D, D'Agostino D, Sinani E, Yu H, Sherman AV, Babu S, Berry JD, Midei MG, Milner PG, Cudkowicz ME, Paganoni S. An expanded access protocol of RT001 in amyotrophic lateral sclerosis-Initial experience with a lipid peroxidation inhibitor. Muscle Nerve 2022; 66:421-425. [PMID: 35765222 PMCID: PMC9796343 DOI: 10.1002/mus.27672] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/22/2022] [Accepted: 06/26/2022] [Indexed: 01/01/2023]
Abstract
INTRODUCTION/AIMS Lipid peroxidation is thought to play a biologically important role in motor neuron death in amyotrophic lateral sclerosis (ALS). 11,11 Di-deuterated linoleic ethyl ester (RT001) prevents lipid peroxidation in cellular and mitochondrial membranes. Herein we report on the use of RT001 under expanded access (EA). METHODS We provided RT001 to patients with ALS via EA at a single site. The starting dose was 2.88 g/day, which was increased to to 8.64 g/day as tolerated. Participants were not eligible for alternative clinical trials. Participants were followed for adverse events and pharmacokinetic (PK) parameters were measured approximately 3 months after RT001 initiation. RESULTS Sixteen participants received RT001 (5.6 ± 1.6 g/day; dose range, 1.92 to 8.64 g/day) for a mean period of 10.8 ± 7.1 months. After 3 months of treatment, PK studies showed that RT001 was absorbed, metabolized, and incorporated into red blood cell membranes at concentrations expected to be therapeutic based on in vitro models. The most common adverse events were gastrointestinal, including diarrhea, which occurred in 25% of the participants, and were considered possibly related to RT001. One participant (6%) discontinued due to an adverse event. Ten serious adverse events occurred: these events were recognized complications of ALS and none were attributed to treatment with RT001. DISCUSSION RT001 was administered safely to a small group of people living with ALS in the context of an EA protocol. Currently, there is an ongoing randomized, double-blind, controlled study of RT001 in ALS.
Collapse
Affiliation(s)
- Megan Yerton
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Allison Winter
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Anthony Kostov
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Cassandra Lieberman
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Dario Gelevski
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Harli Weber
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Michael Doyle
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Geli Kane
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Neil Parikh
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Katherine M. Burke
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Margot Rohrer
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Taylor Stirrat
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Margaret Bruno
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Alison Hochman
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Sarah Luppino
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Jennifer Scalia
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Debra Skoniecki
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Derek D'Agostino
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Ervin Sinani
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Hong Yu
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Alexander V. Sherman
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Suma Babu
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - James D. Berry
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | | | | | - Merit E. Cudkowicz
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Sabrina Paganoni
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts,Spaulding Rehabilitation Hospital, Harvard Medical SchoolBostonMassachusetts
| |
Collapse
|
16
|
Wang Y, Yin R, Tang Z, Liu W, He C, Xia D. Reactive Nitrogen Species Mediated Inactivation of Pathogenic Microorganisms during UVA Photolysis of Nitrite at Surface Water Levels. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:12542-12552. [PMID: 35976624 DOI: 10.1021/acs.est.2c01136] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
UVA photolysis of nitrite (NO2-) occurs in a number of natural and engineered aquatic systems. This study reports for the first time that pathogenic microorganisms can be effectively inactivated during the coexposure of UVA irradiation and NO2- under environmentally relevant conditions. The results demonstrated that more than 3 log inactivation of Escherichia coli K-12, Staphylococcus aureus, and Spingopyxis sp. BM1-1 was achieved by UVA photolysis of 2.0 mg-N L-1 of NO2- in synthetic drinking water and real surface water. The inactivation was mainly attributed to the reactive species generated from UVA photolysis of NO2- rather than UVA irradiation or NO2- oxidation alone. The inactivation was predominantly contributed by the reactive nitrogen species (NO2• and ONOO-/HOONO) instead of the reactive oxygen species (HO• or O2•-). A kinetic model to simulate the reactive species generation from UVA photolysis of NO2- was established, validated, and used to predict the contributions of different reactive species to the inactivation under various environmental conditions. Several advanced tools (e.g., D2O - labeling with Raman spectroscopy) were used to demonstrate that the inactivation by the UVA/NO2- treatment was attributed to the DNA destruction by the reactive nitrogen species, which completely suppressed the viable but nonculturable (VBNC) states and the reactivation of bacteria. This study highlights a novel process for the inactivation of pathogenic microorganisms in water and emphasizes the critical role of reactive nitrogen species in water disinfection and purification.
Collapse
Affiliation(s)
- Yongyi Wang
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Ran Yin
- Department of Civil and Environmental Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong, China
| | - Zhuoyun Tang
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Weiqi Liu
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Chun He
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Dehua Xia
- School of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
17
|
Lu X, Su H, Zhang J, Wang N, Wang H, Liu J, Zhao W. Resorufin-based fluorescent probe with elevated water solubility for visualizing fluctuant peroxynitrite in progression of inflammation. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 267:120620. [PMID: 34802934 DOI: 10.1016/j.saa.2021.120620] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 06/13/2023]
Abstract
Inflammation is a significant protective response in biological systems and associated with various diseases. Peroxynitrite (ONOO-) as a highly active oxidant participates in the inflammatory process of organisms. Thus, it is necessary to construct novel fluorescent probes for exploring inflammation-related diseases through detecting endogenous ONOO-. Resorufin-based fluorescent probes for testing ONOO- were rare and suffered from poor water solubility. In this work, we elaborately designed three resorufin-based incorporating isatin derivatives probes RF-ITs and successfully obtained two highly selective probes RF-IT-OC and RF-IT-EG for ONOO-. Comparing the other two probes, RF-IT-EG containing triethylene glycol monomethyl ether on isatin moiety displayed better water solubility (3.2 mg/L), faster response rate (60 s), larger signal-to-noise ratio (103-fold) and lower detection limit (87 nM) for monitoring ONOO-. The cells imaging results manifested that probe RF-IT-EG could be applied to trace endogenous ONOO- with inappreciable cytotoxicity. Moreover, the RF-IT-EG was capable of tracking the fluctuation of endogenous ONOO- in LPS-stimulated inflamed mouse leg models. This work will provide a faithful and promising probe for illustrating the roles of ONOO- in various inflammation-related diseases.
Collapse
Affiliation(s)
- Xiaoyan Lu
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng, 475004, P. R. China
| | - Huihui Su
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng, 475004, P. R. China
| | - Jian Zhang
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng, 475004, P. R. China.
| | - Nannan Wang
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng, 475004, P. R. China
| | - Han Wang
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng, 475004, P. R. China
| | - Jinying Liu
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng, 475004, P. R. China
| | - Weili Zhao
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng, 475004, P. R. China; School of Pharmacy, Institutes of Integrative Medicine, Fudan University, Shanghai, 201203, P. R. China.
| |
Collapse
|
18
|
Abstract
Significance: Vitamin C (ascorbate), in regard to its effectiveness against malignancies, has had a controversial history in cancer treatment. It has been shown that in vitro and in vivo anticancer efficacy of ascorbate relies on its pro-oxidant effect mainly from an increased generation of reactive oxygen species (ROS). A growing understanding of its anticancer activities and pharmacokinetic properties has prompted scientists to re-evaluate the significance of ascorbate in cancer treatment. Recent Advances: A recent resurge in ascorbate research emerged after discovering that, at high doses, ascorbate preferentially kills Kirsten-Ras (K-ras)- and B-raf oncogene (BRAF)-mutant cancer cells. In addition, some of the main hallmarks of cancer cells, such as redox homeostasis and oxygen-sensing regulation (through inhibition of hypoxia-inducible factor-1 alpha [HIF-1α] activity), are affected by vitamin C. Critical Issues: Currently, there is no clear consensus from the literature in regard to the beneficial effects of antioxidants. Results from both human and animal studies provide no clear evidence about the benefit of antioxidant treatment in preventing or suppressing cancer development. Since pro-oxidants may affect both normal and tumor cells, the extremely low toxicity of ascorbate represents a main advantage. This guarantees the safe inclusion of ascorbate in clinical protocols to treat cancer patients. Future Directions: Current research could focus on elucidating the wide array of reactions between ascorbate and reactive species, namely ROS, reactive nitrogen species as well as reactive sulfide species, and their intracellular molecular targets. Unraveling these mechanisms could allow researchers to assess what could be the optimal combination of ascorbate with standard treatments.
Collapse
Affiliation(s)
- Christophe Glorieux
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, P. R. China
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique, Chile.,Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
19
|
Martin LJ, Wong M. Skeletal Muscle-Restricted Expression of Human SOD1 in Transgenic Mice Causes a Fatal ALS-Like Syndrome. Front Neurol 2020; 11:592851. [PMID: 33381076 PMCID: PMC7767933 DOI: 10.3389/fneur.2020.592851] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal heterogeneous neurodegenerative disease that causes motor neuron (MN) loss and skeletal muscle paralysis. It is uncertain whether this degeneration of MNs is triggered intrinsically and is autonomous, or if the disease initiating mechanisms are extrinsic to MNs. We hypothesized that skeletal muscle is a primary site of pathogenesis in ALS that triggers MN degeneration. Some inherited forms of ALS are caused by mutations in the superoxide dismutase-1 (SOD1) gene, that encodes an antioxidant protein, so we created transgenic (tg) mice expressing wild-type-, G37R-, and G93A-human SOD1 gene variants only in skeletal muscle. Presence of human SOD1 (hSOD1) protein in skeletal muscle was verified by western blotting, enzyme activity gels, and immunofluorescence in myofibers and satellite cells. These tg mice developed limb weakness and paresis with motor deficits, limb and chest muscle wasting, diaphragm atrophy, and age-related fatal disease with a lifespan shortening of 10–16%. Brown and white adipose tissue also became wasted. Myofibers of tg mice developed crystalline-like inclusions, individualized sarcomere destruction, mitochondriopathy with vesiculation, DNA damage, and activated p53. Satellite cells became apoptotic. The diaphragm developed severe loss of neuromuscular junction presynaptic and postsynaptic integrity, including decreased innervation, loss of synaptophysin, nitration of synaptophysin, and loss of nicotinic acetylcholine receptor and scaffold protein rapsyn. Co-immunoprecipitation identified hSOD1 interaction with rapsyn. Spinal cords of tg mice developed gross atrophy. Spinal MNs formed cytoplasmic and nuclear inclusions, axonopathy, mitochondriopathy, accumulated DNA damage, activated p53 and cleaved caspase-3, and died. Tg mice had a 40–50% loss of MNs. This work shows that hSOD1 in skeletal muscle is a driver of pathogenesis in ALS, that involves myofiber and satellite cell toxicity, and apparent muscle-adipose tissue disease relationships. It also identifies a non-autonomous mechanism for MN degeneration explaining their selective vulnerability as likely a form of target-deprivation retrograde neurodegeneration.
Collapse
Affiliation(s)
- Lee J Martin
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Margaret Wong
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
20
|
Singh N. Antioxidant metal oxide nanozymes: role in cellular redox homeostasis and therapeutics. PURE APPL CHEM 2020. [DOI: 10.1515/pac-2020-0802] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Abstract
Nanomaterials with enzyme-like activity, generally referred to as ‘nanozymes’, find myriad potential in various biomedical fields. More importantly, the nanoparticles that can functionally mimic the activity of cellular antioxidant enzymes attract tremendous interest owing to their possible therapeutic candidature in oxidative stress-mediated disorders. Oxidative stress culminating due to excess reactive oxygen species (ROS) level and dysregulated cellular antioxidant machinery is implicated in the development and progression of various pathophysiological disorders such as cancer, diabetes, cardiovascular and neurodegenerative diseases. Moreover, the optimum essentiality of ROS due to its pivotal role in cell signaling evokes the requirement of novel artificial antioxidant enzymes that can circumvent the detrimental effects of enhanced ROS levels without perturbing the basal redox status of cells. In recent years, the fast emanating artificial enzymes, i.e. nanozymes with antioxidant enzyme-like activity, has made tremendous progress with their broad applications in therapeutics, diagnostic medicine, bio-sensing, and immunoassay. Among various antioxidant nanoparticles reported till-date, the metal oxide nanozymes have emerged as the most efficient and successful candidates in mimicking the activity of first-line defense antioxidant enzymes, i.e. superoxide dismutase, catalase, and glutathione peroxidase. This review intends to exclusively highlight the development of representative metal oxide-based antioxidant nanozymes capable of maintaining the cellular redox homeostasis and their potential therapeutic significance.
Collapse
Affiliation(s)
- Namrata Singh
- Department of Inorganic and Physical Chemistry , Indian Institute of Science , Bangalore 560012 , India
| |
Collapse
|
21
|
Floare ML, Allen SP. Why TDP-43? Why Not? Mechanisms of Metabolic Dysfunction in Amyotrophic Lateral Sclerosis. Neurosci Insights 2020; 15:2633105520957302. [PMID: 32995749 PMCID: PMC7503004 DOI: 10.1177/2633105520957302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive and fatal neurodegenerative disorder for which there is no effective curative treatment available and minimal palliative care. Mutations in the gene encoding the TAR DNA-binding protein 43 (TDP-43) are a well-recognized genetic cause of ALS, and an imbalance in energy homeostasis correlates closely to disease susceptibility and progression. Considering previous research supporting a plethora of downstream cellular impairments originating in the histopathological signature of TDP-43, and the solid evidence around metabolic dysfunction in ALS, a causal association between TDP-43 pathology and metabolic dysfunction cannot be ruled out. Here we discuss how TDP-43 contributes on a molecular level to these impairments in energy homeostasis, and whether the protein's pathological effects on cellular metabolism differ from those of other genetic risk factors associated with ALS such as superoxide dismutase 1 (SOD1), chromosome 9 open reading frame 72 (C9orf72) and fused in sarcoma (FUS).
Collapse
Affiliation(s)
- Mara-Luciana Floare
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Scott P. Allen
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| |
Collapse
|
22
|
Zhuo J, Gong K, Guo Y, Lu G, Chi H, Duan Y, Zhang Z, Li X. A Silyl Ether Based Fluorescent Probe for Rapid Monitoring of Endogenous Peroxynitrite Concentration and Imaging in Living Cells through Multicolor Emission. Chempluschem 2020; 85:684-688. [PMID: 32253835 DOI: 10.1002/cplu.202000100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/21/2020] [Indexed: 11/08/2022]
Abstract
The peroxynitrite ion (ONOO- ) has important roles in many biological processes. We have developed a multicolor ONOO- -sensing probe (SiONNOH) that undergoes deprotonation and desilylation processes, which result in several changes in the emission wavelengths. In response to different concentrations of ONOO- , the probe exhibits fluorescence changes from pink (595 nm at 2 eq. ONOO- ) to green (540 nm at 6 eq. ONOO- ) via orange (3 eq. ONOO- ) and yellow (4 eq. ONOO- ) under physiological conditions until no fluorescence signal is observed after ONOO- is completely eliminated by lipoic acid. The probe shows the high selectivity for ONOO- and the limit of detection is calculated to be 1.27 μM. Moreover, the probe shows the capacity to monitor the concentration ranges of ONOO- through multicolor fluorescence in living cells, which will greatly facilitate the rapid detection of ONOO- concentration ranges by the naked eye under a UV light without any precision instrumentation.
Collapse
Affiliation(s)
- Jiezhen Zhuo
- School of Chemical Engineering, University of Science and Technology Liaoning, 185 Qianshan Zhong Road, Anshan, 114051, P. R. China
| | - Ke Gong
- College of Food and Biological Engineering, Hefei University of Technology, 420 Feicui Road, Hefei, 230601, P. R. China
| | - Yuxin Guo
- School of Chemical Engineering, University of Science and Technology Liaoning, 185 Qianshan Zhong Road, Anshan, 114051, P. R. China
| | - Gonghao Lu
- School of Chemical Engineering, University of Science and Technology Liaoning, 185 Qianshan Zhong Road, Anshan, 114051, P. R. China
| | - Haijun Chi
- School of Chemical Engineering, University of Science and Technology Liaoning, 185 Qianshan Zhong Road, Anshan, 114051, P. R. China
| | - Yajun Duan
- College of Food and Biological Engineering, Hefei University of Technology, 420 Feicui Road, Hefei, 230601, P. R. China
| | - Zhiqiang Zhang
- School of Chemical Engineering, University of Science and Technology Liaoning, 185 Qianshan Zhong Road, Anshan, 114051, P. R. China
| | - Xue Li
- School of Material and Metallurgy, University of Science and Technology Liaoning, 185 Qianshan Zhong Road, Anshan, 114051, P. R. China
| |
Collapse
|
23
|
Croft T, Venkatakrishnan P, Lin SJ. NAD + Metabolism and Regulation: Lessons From Yeast. Biomolecules 2020; 10:E330. [PMID: 32092906 PMCID: PMC7072712 DOI: 10.3390/biom10020330] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/15/2020] [Accepted: 02/16/2020] [Indexed: 12/13/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential metabolite involved in various cellular processes. The cellular NAD+ pool is maintained by three biosynthesis pathways, which are largely conserved from bacteria to human. NAD+ metabolism is an emerging therapeutic target for several human disorders including diabetes, cancer, and neuron degeneration. Factors regulating NAD+ homeostasis have remained incompletely understood due to the dynamic nature and complexity of NAD+ metabolism. Recent studies using the genetically tractable budding yeast Saccharomyces cerevisiae have identified novel NAD+ homeostasis factors. These findings help provide a molecular basis for how may NAD+ and NAD+ homeostasis factors contribute to the maintenance and regulation of cellular function. Here we summarize major NAD+ biosynthesis pathways, selected cellular processes that closely connect with and contribute to NAD+ homeostasis, and regulation of NAD+ metabolism by nutrient-sensing signaling pathways. We also extend the discussions to include possible implications of NAD+ homeostasis factors in human disorders. Understanding the cross-regulation and interconnections of NAD+ precursors and associated cellular pathways will help elucidate the mechanisms of the complex regulation of NAD+ homeostasis. These studies may also contribute to the development of effective NAD+-based therapeutic strategies specific for different types of NAD+ deficiency related disorders.
Collapse
Affiliation(s)
| | | | - Su-Ju Lin
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, CA 95616, USA; (T.C.); (P.V.)
| |
Collapse
|
24
|
Hou JT, Wang B, Zhang Y, Cui B, Cao X, Zhang M, Ye Y, Wang S. Observation of peroxynitrite overproduction in cells during 5-fluorouracil treatment via a ratiometric fluorescent probe. Chem Commun (Camb) 2020; 56:2759-2762. [PMID: 32022003 DOI: 10.1039/c9cc09652c] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We describe a colorimetric and fluorescent probe 3a to detect cellular peroxynitrite with high selectivity and sensitivity. 3a was successfully applied in the bioimaging of exogenous and endogenous peroxynitrite in living cells. The up-regulation of peroxynitrite in cancer cells and normal cells during 5-fluorouracil treatment was finally monitored.
Collapse
Affiliation(s)
- Ji-Ting Hou
- College of Chemistry and Chemical Engineering, Xinyang Normal University, Xinyang 464000, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Kim BW, Jeong YE, Wong M, Martin LJ. DNA damage accumulates and responses are engaged in human ALS brain and spinal motor neurons and DNA repair is activatable in iPSC-derived motor neurons with SOD1 mutations. Acta Neuropathol Commun 2020; 8:7. [PMID: 32005289 PMCID: PMC6995159 DOI: 10.1186/s40478-019-0874-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022] Open
Abstract
DNA damage is implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS). However, relationships between DNA damage accumulation, DNA damage response (DDR), and upper and lower motor neuron vulnerability in human ALS are unclear; furthermore, it is unknown whether epigenetic silencing of DNA repair pathways contributes to ALS pathogenesis. We tested the hypotheses that DNA damage accumulates in ALS motor neurons along with diminished DDR, and that DNA repair genes undergo hypermethylation. Human postmortem CNS tissue was obtained from ALS cases (N = 34) and age-matched controls without neurologic disease (N = 15). Compared to age-matched controls, abasic sites accumulated in genomic DNA of ALS motor cortex and laser capture microdissection-acquired spinal motor neurons but not in motor neuron mitochondrial DNA. By immunohistochemistry, DNA damage accumulated significantly in upper and lower motor neurons in ALS cases as single-stranded DNA and 8-hydroxy-deoxyguanosine (OHdG) compared to age-matched controls. Significant DDR was engaged in ALS motor neurons as evidenced by accumulation of c-Abl, nuclear BRCA1, and ATM activation. DNA damage and DDR were present in motor neurons at pre-attritional stages and throughout the somatodendritic attritional stages of neurodegeneration. Motor neurons with DNA damage were also positive for activated p53 and cleaved caspase-3. Gene-specific promoter DNA methylation pyrosequencing identified the DNA repair genes Ogg1, Apex1, Pnkp and Aptx as hypomethylated in ALS. In human induced-pluripotent stem cell (iPSC)-derived motor neurons with familial ALS SOD1 mutations, DNA repair capacity was similar to isogenic control motor neurons. Our results show that vulnerable neurons in human ALS accumulate DNA damage, and contrary to our hypothesis, strongly activate and mobilize response effectors and DNA repair genes. This DDR in ALS motor neurons involves recruitment of c-Abl and BRCA1 to the nucleus in vivo, and repair of DNA double-strand breaks in human ALS motor neurons with SOD1 mutations in cell culture.
Collapse
Affiliation(s)
- Byung Woo Kim
- Department of Pathology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD, 21205-2196, USA
- Division of Neuropathology, the Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ye Eun Jeong
- Division of Neuropathology, the Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Margaret Wong
- Department of Pathology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD, 21205-2196, USA
| | - Lee J Martin
- Department of Pathology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD, 21205-2196, USA.
- Division of Neuropathology, the Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
26
|
Shi X, Zhan Q, Li Y, Zhou L, Wei S. Multiple Functions Integrated inside a Single Molecule for Amplification of Photodynamic Therapy Activity. Mol Pharm 2020; 17:190-201. [PMID: 31804837 DOI: 10.1021/acs.molpharmaceut.9b00893] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nitric oxide (NO) can play both prosurvival and prodeath roles in photodynamic therapy (PDT). The generation efficiency of peroxynitrite anions (ONOO-), by NO and superoxide anions (O2•-), significantly influenced the outcome. Reports indicated that such efficiency is closely related to the distance between NO and O2•-. Thus, in this manuscript, l-arginine (Arg) ethyl ester-modified zinc phthalocyanine (Arg-ZnPc) was designed and synthesized as a photosensitizer (PS) and NO donor. Post light irradiation, the guanido of Arg-ZnPc can be effectively oxidized by the generated reactive oxygen species (ROS) in the PDT process to release NO. Such a strategy could ensure O2•- and NO generation in the same place at the same time to guarantee effective ONOO- formation. In addition, NO has other multiple synergistic cancer treatment functions, including tumor tissue vasodilatation for drug extravasation promotion, P-glycoprotein (P-gp) downregulation for drug efflux inhibition, and glutathione depletion for cancer cell endogenous antioxidant defense destruction. In vitro and in vivo results indicated that the effective ONOO- formation and multiple functions of Arg-ZnPc could synergistically enhance its PDT activity and ensure satisfactory cancer treatment outcome.
Collapse
Affiliation(s)
- Xianqing Shi
- College of Chemistry and Materials Science, Jiangsu Key Laboratory of Biofunctional Materials, Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, Key Laboratory of Applied Photochemistry , Nanjing Normal University , Nanjing , Jiangsu 210023 , China
| | - Qichen Zhan
- College of Chemistry and Materials Science, Jiangsu Key Laboratory of Biofunctional Materials, Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, Key Laboratory of Applied Photochemistry , Nanjing Normal University , Nanjing , Jiangsu 210023 , China
| | - Yanqing Li
- College of Chemistry and Materials Science, Jiangsu Key Laboratory of Biofunctional Materials, Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, Key Laboratory of Applied Photochemistry , Nanjing Normal University , Nanjing , Jiangsu 210023 , China
| | - Lin Zhou
- College of Chemistry and Materials Science, Jiangsu Key Laboratory of Biofunctional Materials, Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, Key Laboratory of Applied Photochemistry , Nanjing Normal University , Nanjing , Jiangsu 210023 , China
| | - Shaohua Wei
- College of Chemistry and Materials Science, Jiangsu Key Laboratory of Biofunctional Materials, Jiangsu Collaborative Innovation Centre of Biomedical Functional Materials, Key Laboratory of Applied Photochemistry , Nanjing Normal University , Nanjing , Jiangsu 210023 , China.,School of Chemistry and Chemical Engineering , Yancheng Institute of Technology , Yancheng , Jiangsu 224051 , China
| |
Collapse
|
27
|
Murfin L, Weber M, Park SJ, Kim WT, Lopez-Alled CM, McMullin CL, Pradaux-Caggiano F, Lyall CL, Kociok-Köhn G, Wenk J, Bull SD, Yoon J, Kim HM, James TD, Lewis SE. Azulene-Derived Fluorescent Probe for Bioimaging: Detection of Reactive Oxygen and Nitrogen Species by Two-Photon Microscopy. J Am Chem Soc 2019; 141:19389-19396. [PMID: 31773957 PMCID: PMC6909233 DOI: 10.1021/jacs.9b09813] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Indexed: 12/28/2022]
Abstract
Two-photon fluorescence microscopy has become an indispensable technique for cellular imaging. Whereas most two-photon fluorescent probes rely on well-known fluorophores, here we report a new fluorophore for bioimaging, namely azulene. A chemodosimeter, comprising a boronate ester receptor motif conjugated to an appropriately substituted azulene, is shown to be an effective two-photon fluorescent probe for reactive oxygen species, showing good cell penetration, high selectivity for peroxynitrite, no cytotoxicity, and excellent photostability.
Collapse
Affiliation(s)
- Lloyd
C. Murfin
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Maria Weber
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- Center
for Sustainable Circular Technologies, University
of Bath, Bath BA2 7AY, United Kingdom
| | - Sang Jun Park
- Department
of Energy Systems Research, Ajou University, Suwon 443-749, South Korea
| | - Won Tae Kim
- Department
of Energy Systems Research, Ajou University, Suwon 443-749, South Korea
| | - Carlos M. Lopez-Alled
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- Center
for Sustainable Circular Technologies, University
of Bath, Bath BA2 7AY, United Kingdom
| | - Claire L. McMullin
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | | | - Catherine L. Lyall
- Materials
and Chemical Characterization (MC), University of Bath, Bath BA2 7AY, United Kingdom
| | - Gabriele Kociok-Köhn
- Materials
and Chemical Characterization (MC), University of Bath, Bath BA2 7AY, United Kingdom
| | - Jannis Wenk
- Center
for Sustainable Circular Technologies, University
of Bath, Bath BA2 7AY, United Kingdom
- Department
of Chemical Engineering, University of Bath, Bath BA2 7AY, United Kingdom
| | - Steven D. Bull
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- Center
for Sustainable Circular Technologies, University
of Bath, Bath BA2 7AY, United Kingdom
| | - Juyoung Yoon
- Department
of Chemistry and Nano Science, Ewha Woman’s
University, Seoul 120-750, South Korea
| | - Hwan Myung Kim
- Department
of Energy Systems Research, Ajou University, Suwon 443-749, South Korea
| | - Tony D. James
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- Center
for Sustainable Circular Technologies, University
of Bath, Bath BA2 7AY, United Kingdom
| | - Simon E. Lewis
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- Center
for Sustainable Circular Technologies, University
of Bath, Bath BA2 7AY, United Kingdom
| |
Collapse
|
28
|
Role of Catalase in Oxidative Stress- and Age-Associated Degenerative Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9613090. [PMID: 31827713 PMCID: PMC6885225 DOI: 10.1155/2019/9613090] [Citation(s) in RCA: 527] [Impact Index Per Article: 87.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/18/2019] [Accepted: 08/14/2019] [Indexed: 12/22/2022]
Abstract
Reactive species produced in the cell during normal cellular metabolism can chemically react with cellular biomolecules such as nucleic acids, proteins, and lipids, thereby causing their oxidative modifications leading to alterations in their compositions and potential damage to their cellular activities. Fortunately, cells have evolved several antioxidant defense mechanisms (as metabolites, vitamins, and enzymes) to neutralize or mitigate the harmful effect of reactive species and/or their byproducts. Any perturbation in the balance in the level of antioxidants and the reactive species results in a physiological condition called “oxidative stress.” A catalase is one of the crucial antioxidant enzymes that mitigates oxidative stress to a considerable extent by destroying cellular hydrogen peroxide to produce water and oxygen. Deficiency or malfunction of catalase is postulated to be related to the pathogenesis of many age-associated degenerative diseases like diabetes mellitus, hypertension, anemia, vitiligo, Alzheimer's disease, Parkinson's disease, bipolar disorder, cancer, and schizophrenia. Therefore, efforts are being undertaken in many laboratories to explore its use as a potential drug for the treatment of such diseases. This paper describes the direct and indirect involvement of deficiency and/or modification of catalase in the pathogenesis of some important diseases such as diabetes mellitus, Alzheimer's disease, Parkinson's disease, vitiligo, and acatalasemia. Details on the efforts exploring the potential treatment of these diseases using a catalase as a protein therapeutic agent have also been described.
Collapse
|
29
|
Redox active metals in neurodegenerative diseases. J Biol Inorg Chem 2019; 24:1141-1157. [PMID: 31650248 DOI: 10.1007/s00775-019-01731-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/14/2019] [Indexed: 12/11/2022]
Abstract
Copper (Cu) and iron (Fe) are redox active metals essential for the regulation of cellular pathways that are fundamental for brain function, including neurotransmitter synthesis and release, neurotransmission, and protein turnover. Cu and Fe are tightly regulated by sophisticated homeostatic systems that tune the levels and localization of these redox active metals. The regulation of Cu and Fe necessitates their coordination to small organic molecules and metal chaperone proteins that restrict their reactions to specific protein centres, where Cu and Fe cycle between reduced (Fe2+, Cu+) and oxidised states (Fe3+, Cu2+). Perturbation of this regulation is evident in the brain affected by neurodegeneration. Here we review the evidence that links Cu and Fe dyshomeostasis to neurodegeneration as well as the promising preclinical and clinical studies reporting pharmacological intervention to remedy Cu and Fe abnormalities in the treatment of Alzheimer's disease (AD), Parkinson's disease (PD) and Amyotrophic lateral sclerosis (ALS).
Collapse
|
30
|
Functions and dysfunctions of nitric oxide in brain. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1949-1967. [DOI: 10.1016/j.bbadis.2018.11.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/29/2018] [Accepted: 11/11/2018] [Indexed: 02/06/2023]
|
31
|
Radi R. The origins of nitric oxide and peroxynitrite research in Uruguay: 25 years of contributions to the biochemical and biomedical sciences. Nitric Oxide 2019; 87:83-89. [DOI: 10.1016/j.niox.2019.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 12/12/2022]
|
32
|
Yanagi KS, Wu Z, Amaya J, Chapkis N, Duffy AM, Hajdarovic KH, Held A, Mathur AD, Russo K, Ryan VH, Steinert BL, Whitt JP, Fallon JR, Fawzi NL, Lipscombe D, Reenan RA, Wharton KA, Hart AC. Meta-analysis of Genetic Modifiers Reveals Candidate Dysregulated Pathways in Amyotrophic Lateral Sclerosis. Neuroscience 2019; 396:A3-A20. [PMID: 30594291 PMCID: PMC6549511 DOI: 10.1016/j.neuroscience.2018.10.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/14/2018] [Accepted: 10/16/2018] [Indexed: 12/11/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease that has significant overlap with frontotemporal dementia (FTD). Mutations in specific genes have been identified that can cause and/or predispose patients to ALS. However, the clinical variability seen in ALS patients suggests that additional genes impact pathology, susceptibility, severity, and/or progression of the disease. To identify molecular pathways involved in ALS, we undertook a meta-analysis of published genetic modifiers both in patients and in model organisms, and undertook bioinformatic pathway analysis. From 72 published studies, we generated a list of 946 genes whose perturbation (1) impacted ALS in patient populations, (2) altered defects in laboratory models, or (3) modified defects caused by ALS gene ortholog loss of function. Herein, these are all called modifier genes. We found 727 modifier genes that encode proteins with human orthologs. Of these, 43 modifier genes were identified as modifiers of more than one ALS gene/model, consistent with the hypothesis that shared genes and pathways may underlie ALS. Further, we used a gene ontology-based bioinformatic analysis to identify pathways and associated genes that may be important in ALS. To our knowledge this is the first comprehensive survey of ALS modifier genes. This work suggests that shared molecular mechanisms may underlie pathology caused by different ALS disease genes. Surprisingly, few ALS modifier genes have been tested in more than one disease model. Understanding genes that modify ALS-associated defects will help to elucidate the molecular pathways that underlie ALS and provide additional targets for therapeutic intervention.
Collapse
Affiliation(s)
- Katherine S Yanagi
- Neuroscience Graduate Program, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Zhijin Wu
- Department of Biostatistics, Brown University, Providence, Rhode Island 02912, United States.
| | - Joshua Amaya
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Natalie Chapkis
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Amanda M Duffy
- Neuroscience Graduate Program, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Kaitlyn H Hajdarovic
- Neuroscience Graduate Program, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Aaron Held
- Molecular Biology, Cell Biology, and Biochemistry Graduate Program, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Arjun D Mathur
- Molecular Biology, Cell Biology, and Biochemistry Graduate Program, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Kathryn Russo
- Neuroscience Graduate Program, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Veronica H Ryan
- Neuroscience Graduate Program, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Beatrice L Steinert
- Molecular Biology, Cell Biology, and Biochemistry Department, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Joshua P Whitt
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Justin R Fallon
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Nicolas L Fawzi
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Diane Lipscombe
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Robert A Reenan
- Molecular Biology, Cell Biology, and Biochemistry Department, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Kristi A Wharton
- Molecular Biology, Cell Biology, and Biochemistry Department, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| | - Anne C Hart
- Department of Neuroscience, Brown University, Providence, Rhode Island 02912, United States; Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912, United States.
| |
Collapse
|
33
|
García JC, Bustos RH. The Genetic Diagnosis of Neurodegenerative Diseases and Therapeutic Perspectives. Brain Sci 2018; 8:brainsci8120222. [PMID: 30551598 PMCID: PMC6316116 DOI: 10.3390/brainsci8120222] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/26/2018] [Accepted: 12/07/2018] [Indexed: 12/12/2022] Open
Abstract
Genetics has led to a new focus regarding approaches to the most prevalent diseases today. Ascertaining the molecular secrets of neurodegenerative diseases will lead to developing drugs that will change natural history, thereby affecting the quality of life and mortality of patients. The sequencing of candidate genes in patients suffering neurodegenerative pathologies is faster, more accurate, and has a lower cost, thereby enabling algorithms to be proposed regarding the risk of neurodegeneration onset in healthy persons including the year of onset and neurodegeneration severity. Next generation sequencing has resulted in an explosion of articles regarding the diagnosis of neurodegenerative diseases involving exome sequencing or sequencing a whole gene for correlating phenotypical expression with genetic mutations in proteins having key functions. Many of them occur in neuronal glia, which can trigger a proinflammatory effect leading to defective proteins causing sporadic or familial mutations. This article reviews the genetic diagnosis techniques and the importance of bioinformatics in interpreting results from neurodegenerative diseases. Risk scores must be established in the near future regarding diseases with a high incidence in healthy people for defining prevention strategies or an early start for giving drugs in the absence of symptoms.
Collapse
Affiliation(s)
- Julio-César García
- Evidence-Based Therapeutics Group, Department of Clinical Pharmacology, Universidad de La Sabana, Chía 140013, Colombia.
- Department of Clinical Pharmacology, Clínica Universidad de La Sabana, Chía 140013, Colombia.
| | - Rosa-Helena Bustos
- Evidence-Based Therapeutics Group, Department of Clinical Pharmacology, Universidad de La Sabana, Chía 140013, Colombia.
| |
Collapse
|
34
|
Dong Y, Yang Y, Liu J, Awan F, Lu C, Liu Y. Inhibition of Aeromonas hydrophila-induced intestinal inflammation and mucosal barrier function damage in crucian carp by oral administration of Lactococcus lactis. FISH & SHELLFISH IMMUNOLOGY 2018; 83:359-367. [PMID: 30236608 DOI: 10.1016/j.fsi.2018.09.041] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 09/09/2018] [Accepted: 09/12/2018] [Indexed: 06/08/2023]
Abstract
This study explored the immunomodulatory effect and inhibition effects of the candidate probiotic Lactococcus lactis 16-7, which was isolated from crucian carp, on Aeromonas hydrophila infection in crucian carp. The experimental fish were divided into two groups; one was fed a diet supplemented with L. lactis, while the other was fed the control probiotic-free diet. After feeding for 42 d with the experimental diets, the fish that received the diet supplemented with probiotics exhibited a significantly enhanced serum superoxide dismutase activity, phagocytic activities of innate immune cells, and the expression levels of immune-related genes [interferon-γ (INF-γ), interleukin-1β (IL-1β), interleukin-11 (IL-11), tumour necrosis factor α (TNF-α) and myeloid differentiation factor 88 (MyD88)], indicating that L. lactis 16-7 could activate the non-specific immune system of crucian carp. At the end of the feeding trial, the crucian carps in each group were orally infected with A. hydrophila NJ-35. The results show that L. lactis 16-7 could prevent the increase in d-lactic acid concentration and inflammatory response caused by A. hydrophila in crucian carp. Compared with A. hydrophila group, L. lactis 16-7 preserved the integrity of intestinal villi and mitigated A. hydrophila-induced reduce in the transcriptional levels of tight junction (TJ) proteins zonula occludens-1 (ZO-1) and occludin, indicating that L. lactis 16-7 could reduce intestinal mucosal barrier damage and inflammation induced by A. hydrophila in crucian carp. In addition, L. lactis 16-7 could effectively antagonize the colonization of A. hydrophila in the intestine. Overall, these data clearly indicate that L. lactis 16-7 has the potential to be developed as a probiotic agent against A. hydrophila infection in aquaculture.
Collapse
Affiliation(s)
- Yuhao Dong
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yuanyuan Yang
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jin Liu
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Furqan Awan
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Chengping Lu
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yongjie Liu
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
35
|
Prolo C, Rios N, Piacenza L, Álvarez MN, Radi R. Fluorescence and chemiluminescence approaches for peroxynitrite detection. Free Radic Biol Med 2018; 128:59-68. [PMID: 29454880 DOI: 10.1016/j.freeradbiomed.2018.02.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/07/2018] [Accepted: 02/12/2018] [Indexed: 12/23/2022]
Abstract
In the last two decades, there has been a significant advance in understanding the biochemistry of peroxynitrite, an endogenously-produced oxidant and nucleophile. Its relevance as a mediator in several pathologic states and the aging process together with its transient character and low steady-state concentration, motivated the development of a variety of techniques for its unambiguous detection and estimation. Among these, fluorescence and chemiluminescence approaches have represented important tools with enhanced sensitivity but usual limited specificity. In this review, we analyze selected examples of molecular probes that permit the detection of peroxynitrite by fluorescence and chemiluminescence, disclosing their mechanism of reaction with either peroxynitrite or peroxynitrite-derived radicals. Indeed, probes have been divided into 1) redox probes that yield products by a free radical mechanism, and 2) electrophilic probes that evolve to products secondary to the nucleophilic attack by peroxynitrite. Overall, boronate-based compounds are emerging as preferred probes for the sensitive and specific detection and quantitation. Moreover, novel strategies involving genetically-modified fluorescent proteins with the incorporation of unnatural amino acids have been recently described as peroxynitrite sensors. This review analyzes the most commonly used fluorescence and chemiluminescence approaches for peroxynitrite detection and provides some guidelines for appropriate experimental design and data interpretation, including how to estimate peroxynitrite formation rates in cells.
Collapse
Affiliation(s)
- Carolina Prolo
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Natalia Rios
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Lucia Piacenza
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - María Noel Álvarez
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
36
|
Wang S, Chen L, Jangili P, Sharma A, Li W, Hou JT, Qin C, Yoon J, Kim JS. Design and applications of fluorescent detectors for peroxynitrite. Coord Chem Rev 2018. [DOI: 10.1016/j.ccr.2018.06.013] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
37
|
Rubio V, García-Pérez AI, Herráez A, Diez JC. Different roles of Nrf2 and NFKB in the antioxidant imbalance produced by esculetin or quercetin on NB4 leukemia cells. Chem Biol Interact 2018; 294:158-166. [PMID: 30171828 DOI: 10.1016/j.cbi.2018.08.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/23/2018] [Accepted: 08/17/2018] [Indexed: 01/08/2023]
Abstract
Esculetin (6,7-dihydrocoumarin) and the flavonoid quercetin (3,5,7,3',4' pentahydroxyflavone) are compounds that could change the balance of redox homeostasis. NB4 leukemia cells were treated with 25 μM quercetin for 24 h and with esculetin at either 100 or 500 μM for different times. Quercetin increased the levels of pro-inflammatory NFkB p65 in the nucleus correspondingly reducing them in the cytosol. The levels of NFkB p65 decreased in the nucleus at high esculetin concentration treatments for long times (19 h), concomitantly increasing the levels of anti-inflammatory NFkB p50 in the nucleus. This could suggest formation of inhibitory p50 homodimers possibly related with anti-inflammatory response. Lipoxygenase expression was reduced either by esculetin or quercetin. A significant increase of Nrf2 in the nucleus of NB4 cells treated with 100 μM esculetin for 19 h was observed. Quercetin increased the levels of Nrf2 in the cytosol reducing them in the nucleus. Superoxide dismutase expression increased in NB4 cells treated with esculetin in contrast with quercetin. All these data support a relevant differential role for NFkB and Nrf2 in anti-inflammatory and redox response when apoptosis was induced by esculetin or quercetin in human leukemia NB4 cells.
Collapse
Affiliation(s)
- Virginia Rubio
- Unidad de Bioquímica y Biología Molecular, Departamento de Biología de Sistemas, Facultad de Medicina y Ciencias de la Salud, Campus Universitario, Universidad de Alcalá, 28871, Alcalá de Henares, Madrid, Spain
| | - Ana I García-Pérez
- Unidad de Bioquímica y Biología Molecular, Departamento de Biología de Sistemas, Facultad de Medicina y Ciencias de la Salud, Campus Universitario, Universidad de Alcalá, 28871, Alcalá de Henares, Madrid, Spain
| | - Angel Herráez
- Unidad de Bioquímica y Biología Molecular, Departamento de Biología de Sistemas, Facultad de Medicina y Ciencias de la Salud, Campus Universitario, Universidad de Alcalá, 28871, Alcalá de Henares, Madrid, Spain
| | - José C Diez
- Unidad de Bioquímica y Biología Molecular, Departamento de Biología de Sistemas, Facultad de Medicina y Ciencias de la Salud, Campus Universitario, Universidad de Alcalá, 28871, Alcalá de Henares, Madrid, Spain.
| |
Collapse
|
38
|
SK M, Nandi S, Singh RK, Trivedi V, Biswas S. Selective Sensing of Peroxynitrite by Hf-Based UiO-66-B(OH)2 Metal–Organic Framework: Applicability to Cell Imaging. Inorg Chem 2018; 57:10128-10136. [DOI: 10.1021/acs.inorgchem.8b01310] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
39
|
Islam BU, Habib S, Ali SA, Moinuddin, Ali A. Role of Peroxynitrite-Induced Activation of Poly(ADP-Ribose) Polymerase (PARP) in Circulatory Shock and Related Pathological Conditions. Cardiovasc Toxicol 2018; 17:373-383. [PMID: 27990620 DOI: 10.1007/s12012-016-9394-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Peroxynitrite is a powerful oxidant, formed from the reaction of nitric oxide and superoxide. It is known to interact and modify different biological molecules such as DNA, lipids and proteins leading to alterations in their structure and functions. These events elicit various cellular responses, including cell signaling, causing oxidative damage and committing cells to apoptosis or necrosis. This review discusses nitrosative stress-induced modification in the DNA molecule that results in the formation of 8-nitroguanine and 8-oxoguanine, and its role in disease conditions. Different approaches of cell death, such as necrosis and apoptosis, are modulated by cellular high-energy species, such as ATP and NAD+. High concentrations of peroxynitrite are known to cause necrosis, whereas low concentrations lead to apoptosis. Any damage to DNA activates cellular DNA repair machinery, like poly(ADP-ribose) polymerase (PARP). PARP-1, an isoform of PARP, is a DNA nick-sensing enzyme that becomes activated upon sensing DNA breakage and triggers the cleavage of NAD+ into nicotinamide and ADP-ribose and polymerizes the latter on nuclear acceptor proteins. Peroxynitrite-induced hyperactivation of PARP causes depletion of NAD+ and ATP culminating cell dysfunction, necrosis or apoptosis. This mechanistic pathway is implicated in the pathogenesis of a variety of diseases, including circulatory shock (which is characterized by cellular hypoxia triggered by systemic altered perfusion and tissue oxygen utilization leading end-organ dysfunction), sepsis and inflammation, injuries of the lung and the intestine. The cytotoxic effects of peroxynitrite centering on the participation of PARP-1 and ADP-ribose in previously stated diseases have also been discussed in this review.
Collapse
Affiliation(s)
- Badar Ul Islam
- Department of Biochemistry, J. N. Medical College, Aligarh Muslim University, Aligarh, UP, 202002, India
| | - Safia Habib
- Department of Biochemistry, J. N. Medical College, Aligarh Muslim University, Aligarh, UP, 202002, India
| | - Syed Amaan Ali
- Kothiwal Dental College and Research Center, Moradabad, UP, India
| | - Moinuddin
- Department of Biochemistry, J. N. Medical College, Aligarh Muslim University, Aligarh, UP, 202002, India
| | - Asif Ali
- Department of Biochemistry, J. N. Medical College, Aligarh Muslim University, Aligarh, UP, 202002, India.
| |
Collapse
|
40
|
Martin LJ, Chang Q. DNA Damage Response and Repair, DNA Methylation, and Cell Death in Human Neurons and Experimental Animal Neurons Are Different. J Neuropathol Exp Neurol 2018; 77:636-655. [PMID: 29788379 PMCID: PMC6005106 DOI: 10.1093/jnen/nly040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neurological disorders affecting individuals in infancy to old age elude interventions for meaningful protection against neurodegeneration, and preclinical work has not translated to humans. We studied human neuron responses to injury and death stimuli compared to those of animal neurons in culture under similar settings of insult (excitotoxicity, oxidative stress, and DNA damage). Human neurons were differentiated from a cortical neuron cell line and the embryonic stem cell-derived H9 line. Mouse neurons were differentiated from forebrain neural stem cells and embryonic cerebral cortex; pig neurons were derived from forebrain neural stem cells. Mitochondrial morphology was different in human and mouse neurons. Human and mouse neurons challenged with DNA-damaging agent camptothecin showed different chromatin condensation, cell death, and DNA damage sensor activation. DNA damage accumulation and repair kinetics differed among human, mouse, and pig neurons. Promoter CpG island methylation microarrays showed significant differential DNA methylation in human and mouse neurons after injury. Therefore, DNA damage response, DNA repair, DNA methylation, and autonomous cell death mechanisms in human neurons and experimental animal neurons are different.
Collapse
Affiliation(s)
- Lee J Martin
- Department of Pathology, Division of Neuropathology
- Pathobiology Graduate Training Program
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qing Chang
- Department of Pathology, Division of Neuropathology
| |
Collapse
|
41
|
Oxygen radicals, nitric oxide, and peroxynitrite: Redox pathways in molecular medicine. Proc Natl Acad Sci U S A 2018; 115:5839-5848. [PMID: 29802228 DOI: 10.1073/pnas.1804932115] [Citation(s) in RCA: 684] [Impact Index Per Article: 97.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Oxygen-derived free radicals and related oxidants are ubiquitous and short-lived intermediates formed in aerobic organisms throughout life. These reactive species participate in redox reactions leading to oxidative modifications in biomolecules, among which proteins and lipids are preferential targets. Despite a broad array of enzymatic and nonenzymatic antioxidant systems in mammalian cells and microbes, excess oxidant formation causes accumulation of new products that may compromise cell function and structure leading to cell degeneration and death. Oxidative events are associated with pathological conditions and the process of normal aging. Notably, physiological levels of oxidants also modulate cellular functions via homeostatic redox-sensitive cell signaling cascades. On the other hand, nitric oxide (•NO), a free radical and weak oxidant, represents a master physiological regulator via reversible interactions with heme proteins. The bioavailability and actions of •NO are modulated by its fast reaction with superoxide radical ([Formula: see text]), which yields an unusual and reactive peroxide, peroxynitrite, representing the merging of the oxygen radicals and •NO pathways. In this Inaugural Article, I summarize early and remarkable developments in free radical biochemistry and the later evolution of the field toward molecular medicine; this transition includes our contributions disclosing the relationship of •NO with redox intermediates and metabolism. The biochemical characterization, identification, and quantitation of peroxynitrite and its role in disease processes have concentrated much of our attention. Being a mediator of protein oxidation and nitration, lipid peroxidation, mitochondrial dysfunction, and cell death, peroxynitrite represents both a pathophysiologically relevant endogenous cytotoxin and a cytotoxic effector against invading pathogens.
Collapse
|
42
|
Cao J, An W, Reeves AG, Lippert AR. A chemiluminescent probe for cellular peroxynitrite using a self-immolative oxidative decarbonylation reaction. Chem Sci 2018; 9:2552-2558. [PMID: 29732134 PMCID: PMC5914148 DOI: 10.1039/c7sc05087a] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/31/2018] [Indexed: 01/04/2023] Open
Abstract
Peroxynitrite is a damaging agent of oxidative stress that has been difficult to monitor in living cells. Here, an isatin-based chemiluminescent probe for peroxynitrite is reported.
Peroxynitrite (ONOO–) is a highly reactive oxygen species which has been recognized as an endogenous mediator of physiological activities like the immune response as well as a damaging agent of oxidative stress under pathological conditions. While its biological importance is becoming clearer, many of the details of its production and mechanism of action remain elusive due to the lack of available selective and sensitive detection methods. Herein, we report the development, characterization, and biological applications of a reaction-based chemiluminescent probe for ONOO– detection, termed as PNCL. PNCL reacts with ONOO–via an isatin moiety through an oxidative decarbonylation reaction to initiate light emission that can be observed instantly with high selectivity against other reactive sulphur, oxygen, and nitrogen species. Detailed studies were performed to study the reaction between isatin and ONOO–, which confirm selectivity for ONOO– over NO2˙. PNCL has been applied for ONOO– detection in aqueous solution and live cells. Moreover, PNCL can be employed to detect cellular ONOO– generated in macrophages stimulated to mount an immune response with lipopolysaccharide (LPS). The sensitivity granted by chemiluminescent detection together with the specificity of the oxidative decarbonylation reaction provides a useful tool to explore ONOO– chemistry and biology.
Collapse
Affiliation(s)
- Jian Cao
- Department of Chemistry , Southern Methodist University , Dallas , TX 75275-0314 , USA . .,Center for Drug Discovery , Design, and Delivery (CD4) , Southern Methodist University , Dallas , TX 75275-0314 , USA
| | - Weiwei An
- Department of Chemistry , Southern Methodist University , Dallas , TX 75275-0314 , USA . .,Center for Drug Discovery , Design, and Delivery (CD4) , Southern Methodist University , Dallas , TX 75275-0314 , USA
| | - Audrey G Reeves
- Department of Chemistry , Southern Methodist University , Dallas , TX 75275-0314 , USA .
| | - Alexander R Lippert
- Department of Chemistry , Southern Methodist University , Dallas , TX 75275-0314 , USA . .,Center for Drug Discovery , Design, and Delivery (CD4) , Southern Methodist University , Dallas , TX 75275-0314 , USA.,Center for Global Health Impact (CGHI) , Southern Methodist University , Dallas , TX 75275-0314 , USA
| |
Collapse
|
43
|
Zarkovic K, Jakovcevic A, Zarkovic N. Contribution of the HNE-immunohistochemistry to modern pathological concepts of major human diseases. Free Radic Biol Med 2017; 111:110-126. [PMID: 27993730 DOI: 10.1016/j.freeradbiomed.2016.12.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/05/2016] [Accepted: 12/07/2016] [Indexed: 02/06/2023]
Abstract
Excessive production of reactive oxygen species can induce peroxidation of the polyunsaturated fatty acids thus generating reactive aldehydes like 4-hydroxy-2-nonenal (HNE), denoted as "the second messenger of free radicals". Because HNE has high binding affinity for cysteine, histidine and lysine it forms relatively stable and hardly metabolized protein adducts. By changing structure and function of diverse structural and regulatory proteins, HNE achieves not only cytotoxic, but also regulatory functions in various pathophysiological processes. Numerous animal model studies and clinical trials confirmed HNE as one of the crucial factors in development and progression of many disorders, in particular of cancer, (neuro)degenerative, metabolic and inflammatory diseases. Since HNE has multiple biological effects and is in the living system usually bound to proteins and peptides, many research groups work on development of specific immunochemical methods targeting the HNE-histidine adducts as major bioactive marker of lipid peroxidation, following the research pathway initiated by Hermann Esterbauer, who discovered HNE in 60's. Such immunohistochemical studies did not only prove the high biomedical importance of HNE, but have also given new insights into major diseases of the modern man. Immunohistochemical studies have shown reversibility of formation of the HNE-protein adducts, as well as differential onset of the HNE-mediated lipid peroxidation between age- associated atherosclerosis and photoaging, revealing eventually selective anti-cancer effects of HNE produced by non-malignant cells in vicinity of cancer. This review summarizes some of the HNE-histidine immunohistochemistry findings we believe are of broad biomedical interest and could inspire new studies in the field.
Collapse
Affiliation(s)
- Kamelija Zarkovic
- University of Zagreb, School of Medicine, Clinical Hospital Centre Zagreb, Croatia.
| | - Antonia Jakovcevic
- University of Zagreb, School of Medicine, Clinical Hospital Centre Zagreb, Croatia
| | - Neven Zarkovic
- Rudjer Boskovic Institute, Laboratory for Oxidative Stress, Zagreb, Croatia
| |
Collapse
|
44
|
Exploratory double-blind, parallel-group, placebo-controlled extension study of edaravone (MCI-186) in amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2017; 18:20-31. [DOI: 10.1080/21678421.2017.1362000] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
45
|
Stamenković S, Pavićević A, Mojović M, Popović-Bijelić A, Selaković V, Andjus P, Bačić G. In vivo EPR pharmacokinetic evaluation of the redox status and the blood brain barrier permeability in the SOD1 G93A ALS rat model. Free Radic Biol Med 2017; 108:258-269. [PMID: 28366802 DOI: 10.1016/j.freeradbiomed.2017.03.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/25/2017] [Accepted: 03/27/2017] [Indexed: 12/14/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder affecting the motor pathways of the central nervous system. Although a number of pathophysiological mechanisms have been described in the disease, post mortem and animal model studies indicate blood-brain barrier (BBB) disruption and elevated production of reactive oxygen species as major contributors to disease pathology. In this study, the BBB permeability and the brain tissue redox status of the SOD1G93A ALS rat model in the presymptomatic (preALS) and symptomatic (ALS) stages of the disease were investigated by in vivo EPR spectroscopy using three aminoxyl radicals with different cell membrane and BBB permeabilities, Tempol, 3-carbamoyl proxyl (3CP), and 3-carboxy proxyl (3CxP). Additionally, the redox status of the two brain regions previously implicated in disease pathology, brainstem and hippocampus, was investigated by spectrophotometric biochemical assays. The EPR results indicated that among the three spin probes, 3CP is the most suitable for reporting the intracellular redox status changes, as Tempol was reduced in vivo within minutes (t1/2 =2.0±0.5min), thus preventing reliable kinetic modeling, whereas 3CxP reduction kinetics gave divergent conclusions, most probably due to its membrane impermeability. It was observed that the reduction kinetics of 3CP in vivo, in the head of preALS and ALS SOD1G93A rats was altered compared to the controls. Pharmacokinetic modeling of 3CP reduction in vivo, revealed elevated tissue distribution and tissue reduction rate constants indicating an altered brain tissue redox status, and possibly BBB disruption in these animals. The preALS and ALS brain tissue homogenates also showed increased nitrilation, superoxide production, lipid peroxidation and manganese superoxide dismutase activity, and a decreased copper-zinc superoxide dismutase activity. The present study highlights in vivo EPR spectroscopy as a reliable tool for the investigation of changes in BBB permeability and for the unprecedented in vivo monitoring of the brain tissue redox status, as early markers of ALS.
Collapse
Affiliation(s)
- Stefan Stamenković
- University of Belgrade - Faculty of Biology, Center for Laser Microscopy, Studentski trg 3, 11158 Belgrade, Serbia
| | - Aleksandra Pavićević
- University of Belgrade - Faculty of Physical Chemistry, EPR Laboratory, Studentski trg 12-16, 11158 Belgrade, Serbia
| | - Miloš Mojović
- University of Belgrade - Faculty of Physical Chemistry, EPR Laboratory, Studentski trg 12-16, 11158 Belgrade, Serbia
| | - Ana Popović-Bijelić
- University of Belgrade - Faculty of Physical Chemistry, EPR Laboratory, Studentski trg 12-16, 11158 Belgrade, Serbia
| | - Vesna Selaković
- Institute for Medical Research, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia
| | - Pavle Andjus
- University of Belgrade - Faculty of Biology, Center for Laser Microscopy, Studentski trg 3, 11158 Belgrade, Serbia.
| | - Goran Bačić
- University of Belgrade - Faculty of Physical Chemistry, EPR Laboratory, Studentski trg 12-16, 11158 Belgrade, Serbia
| |
Collapse
|
46
|
Li ZH, Liu R, Tan ZL, He L, Lu ZL, Gong B. Aromatization of 9,10-Dihydroacridine Derivatives: Discovering a Highly Selective and Rapid-Responding Fluorescent Probe for Peroxynitrite. ACS Sens 2017; 2:501-505. [PMID: 28723194 DOI: 10.1021/acssensors.7b00139] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
As part of an effort to develop generally applicable strategies for creating probes suitable for detecting important molecular and ionic species, the oxidative aromatization of nonfluorescent 9,10-dihydroacridine derivatives triggered by peroxynitrite (ONOO-) led to the identification of compound 2H, 9-phenyl-9,10-dihydroacridine-4-carboxylic acid, as a rapid-responding fluorescent probe capable of detecting ONOO- with an extraordinary selectivity. Adding a little more than 1 equiv of ONOO- to a solution of 2H resulted in over 100-fold fluorescence enhancement. In sharp contrast, treating 2H with excessive amounts of other oxidants that often interfere with the detection of ONOO- failed to lead to noticeable fluorescence increase. The reaction of ONOO- with 2H shows a similar efficiency in the pH range of 2-8. Low cytotoxicity was observed for 2H and its aromatized product. Bioimaging experiments revealed the promising potential of 2H as a new fluorescent probe for the selective detection of intracellular ONOO-.
Collapse
Affiliation(s)
- Zhi-heng Li
- College
of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Rui Liu
- College
of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Zheng-li Tan
- College
of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Lan He
- National Institute for Food and Drug Control, Beijing 100050, China
| | - Zhong-lin Lu
- College
of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Bing Gong
- College
of Chemistry, Beijing Normal University, Beijing 100875, China
- Department
of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| |
Collapse
|
47
|
Peroxynitrite Activates the NLRP3 Inflammasome Cascade in SOD1(G93A) Mouse Model of Amyotrophic Lateral Sclerosis. Mol Neurobiol 2017; 55:2350-2361. [PMID: 28357805 DOI: 10.1007/s12035-017-0502-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 03/20/2017] [Indexed: 01/02/2023]
Abstract
Neuroinflammation, characterized by the appearance of reactive microglial and astroglial cells, is one of the several pathogenic mechanisms of amyotrophic lateral sclerosis (ALS), a fast-progressing and fatal neurodegenerative disease. Cerebrospinal fluid and spinal cord of ALS patients and SOD1 mutant mice show high concentrations of IL-1β. This interleukin, expressed as an inactive precursor, undergoes a proteolytic maturation by caspase1, whose activation, in turn, depends on inflammasomes. Whether and how inflammasome is activated in ALS models is still to be clarified. The mechanism of inflammasome activation was studied in murine microglial cells overexpressing hSOD1(G93A) and verified in the spinal cord of hSOD1(G93A) mice. Murine microglial hSOD1(G93A) cells express all the inflammasome components and LPS activates caspase1 leading to an increase in the secretion of IL-1β. By activating NF-κB, LPS increases ROS and NO levels that spontaneously react to form peroxynitrite, thus leading to protein nitration. Reduction in peroxynitrite levels results in a decrease in caspase1 activity. Protein nitration and caspase1 activity are concomitantly increased in the spinal cord of pre-symptomatic SOD1(G93A) mice. Oxidative/nitrosative stress induces peroxynitrite formation that may be a key trigger of caspase1/inflammasome activation. Peroxynitrite formation may play a critical role in inflammasome activation and might be exploited as potential therapeutic target for ALS.
Collapse
|
48
|
Abstract
ALS is a relentless neurodegenerative disease in which motor neurons are the susceptible neuronal population. Their death results in progressive paresis of voluntary and respiratory muscles. The unprecedented rate of discoveries over the last two decades have broadened our knowledge of genetic causes and helped delineate molecular pathways. Here we critically review ALS epidemiology, genetics, pathogenic mechanisms, available animal models, and iPS cell technologies with a focus on their translational therapeutic potential. Despite limited clinical success in treatments to date, the new discoveries detailed here offer new models for uncovering disease mechanisms as well as novel strategies for intervention.
Collapse
|
49
|
Kalita B, Ranjan R, Singh A, Yashavarddhan MH, Bajaj S, Gupta ML. A Combination of Podophyllotoxin and Rutin Attenuates Radiation Induced Gastrointestinal Injury by Negatively Regulating NF-κB/p53 Signaling in Lethally Irradiated Mice. PLoS One 2016; 11:e0168525. [PMID: 28036347 PMCID: PMC5201299 DOI: 10.1371/journal.pone.0168525] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 12/03/2016] [Indexed: 12/15/2022] Open
Abstract
Development of an effective radio protector to minimise radiation-inflicted damages have largely failed owing to inherent toxicity of most of the agents examined so far. This study is centred towards delivering protection to lethally irradiated mice by pre-administration of a safe formulation G-003M (combination of podophyllotoxin and rutin) majorly through regulation of inflammatory and cell death pathways in mice. Single intramuscular dose of G-003M injected 60 min prior to 9 Gy exposure rescued 89% of whole body lethally irradiated C57BL/6J mice. Studies have revealed reduction in radiation induced reactive oxygen species (ROS), nitric oxide (NO) generation, prostaglandin E2 (PGE2) levels and intestinal apoptosis in G-003M pre-treated mice intestine. Restricted nuclear translocation of redox-sensitive Nuclear factor-κB (NF-κB) and subsequent downregulation of cyclo-oxygenase 2 (COX-2), inducible nitric oxide synthase (iNOS; EC 1.14.13.39) and tumor necrosis factor (TNF-α) levels demonstrated the anti-inflammatory effect that G-003M exerts. Support to early hematopoietic recovery was exhibited through G-003M mediated induction of granulocyte colony stimulating factor (G-CSF) and interleukin (IL-6) levels in lethally irradiated mice. Considerable attenuation in radiation induced morphological damage to the intestinal villi, crypts and mucosal layers was observed in G-003M pre-treated mice. Additionally, our formulation did not reduce the sensitivity of tumor tissue to radiation. Altogether, these results suggest that G-003M ameliorates the deleterious effects of radiation exposure by minimising ROS and NO generation and effectively regulating inflammatory and cell death pathways. Mechanism of protection elucidated in the current study demonstrates that G-003M can be used as a safe and effective radio protective agent in radiotherapy for human application.
Collapse
Affiliation(s)
- Bhargab Kalita
- Division of Radioprotective Drug Development and Research, Institute of Nuclear Medicine and Allied Sciences, Brig.S.K Mazumdar Marg, Delhi, INDIA
| | - Rajiv Ranjan
- Division of Radioprotective Drug Development and Research, Institute of Nuclear Medicine and Allied Sciences, Brig.S.K Mazumdar Marg, Delhi, INDIA
| | - Abhinav Singh
- Division of Radioprotective Drug Development and Research, Institute of Nuclear Medicine and Allied Sciences, Brig.S.K Mazumdar Marg, Delhi, INDIA
| | - M. H. Yashavarddhan
- Division of Radioprotective Drug Development and Research, Institute of Nuclear Medicine and Allied Sciences, Brig.S.K Mazumdar Marg, Delhi, INDIA
| | - Sania Bajaj
- Division of Radioprotective Drug Development and Research, Institute of Nuclear Medicine and Allied Sciences, Brig.S.K Mazumdar Marg, Delhi, INDIA
| | - Manju Lata Gupta
- Division of Radioprotective Drug Development and Research, Institute of Nuclear Medicine and Allied Sciences, Brig.S.K Mazumdar Marg, Delhi, INDIA
- * E-mail:
| |
Collapse
|
50
|
Johnston-Carey HK, Pomatto LCD, Davies KJA. The Immunoproteasome in oxidative stress, aging, and disease. Crit Rev Biochem Mol Biol 2016; 51:268-81. [PMID: 27098648 DOI: 10.3109/10409238.2016.1172554] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The Immunoproteasome has traditionally been viewed primarily for its role in peptide production for antigen presentation by the major histocompatibility complex, which is critical for immunity. However, recent research has shown that the Immunoproteasome is also very important for the clearance of oxidatively damaged proteins in homeostasis, and especially during stress and disease. The importance of the Immunoproteasome in protein degradation has become more evident as diseases characterized by protein aggregates have also been linked to deficiencies of the Immunoproteasome. Additionally, there are now diseases defined by mutations or polymorphisms within Immunoproteasome-specific subunit genes, further suggesting its crucial role in cytokine signaling and protein homeostasis (or "proteostasis"). The purpose of this review is to highlight our growing understanding of the importance of the Immunoproteasome in the management of protein quality control, and the detrimental impact of its dysregulation during disease and aging.
Collapse
Affiliation(s)
- Helen K Johnston-Carey
- a Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center , The University of Southern California , Los Angeles , CA , USA
| | - Laura C D Pomatto
- a Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center , The University of Southern California , Los Angeles , CA , USA
| | - Kelvin J A Davies
- a Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center , The University of Southern California , Los Angeles , CA , USA ;,b Division of Molecular & Computational Biology, Department of Biological Sciences, Dornsife College of Letters, Arts, & Sciences , Los Angeles , CA , USA
| |
Collapse
|