1
|
Balez R, Stevens CH, Lenk K, Maksour S, Sidhu K, Sutherland G, Ooi L. Increased Neuronal Nitric Oxide Synthase in Alzheimer's Disease Mediates Spontaneous Calcium Signaling and Divergent Glutamatergic Calcium Responses. Antioxid Redox Signal 2024; 41:255-277. [PMID: 38299492 DOI: 10.1089/ars.2023.0395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Affiliation(s)
- Rachelle Balez
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, Australia
| | - Claire H Stevens
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, Australia
| | - Kerstin Lenk
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Institute of Neural Engineering, Graz University of Technology, Graz, Austria
- BioTechMed, Graz, Austria
| | - Simon Maksour
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, Australia
| | - Kuldip Sidhu
- Centre for Healthy Brain Ageing (CheBA), University of New South Wales, Sydney, Australia
| | - Greg Sutherland
- Charles Perkins Centre, University of Sydney, Glebe, Australia
| | - Lezanne Ooi
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, Australia
| |
Collapse
|
2
|
Vassileff N, Spiers JG, Bamford SE, Lowe RGT, Datta KK, Pigram PJ, Hill AF. Microglial activation induces nitric oxide signalling and alters protein S-nitrosylation patterns in extracellular vesicles. J Extracell Vesicles 2024; 13:e12455. [PMID: 38887871 PMCID: PMC11183937 DOI: 10.1002/jev2.12455] [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: 10/04/2023] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 06/20/2024] Open
Abstract
Neuroinflammation is an underlying feature of neurodegenerative conditions, often appearing early in the aetiology of a disease. Microglial activation, a prominent initiator of neuroinflammation, can be induced through lipopolysaccharide (LPS) treatment resulting in expression of the inducible form of nitric oxide synthase (iNOS), which produces nitric oxide (NO). NO post-translationally modifies cysteine thiols through S-nitrosylation, which can alter function of the target protein. Furthermore, packaging of these NO-modified proteins into extracellular vesicles (EVs) allows for the exertion of NO signalling in distant locations, resulting in further propagation of the neuroinflammatory phenotype. Despite this, the NO-modified proteome of activated microglial EVs has not been investigated. This study aimed to identify the protein post-translational modifications NO signalling induces in neuroinflammation. EVs isolated from LPS-treated microglia underwent mass spectral surface imaging using time of flight-secondary ion mass spectrometry (ToF-SIMS), in addition to iodolabelling and comparative proteomic analysis to identify post-translation S-nitrosylation modifications. ToF-SIMS imaging successfully identified cysteine thiol side chains modified through NO signalling in the LPS treated microglial-derived EV proteins. In addition, the iodolabelling proteomic analysis revealed that the EVs from LPS-treated microglia carried S-nitrosylated proteins indicative of neuroinflammation. These included known NO-modified proteins and those associated with LPS-induced microglial activation that may play an essential role in neuroinflammatory communication. Together, these results show activated microglia can exert broad NO signalling changes through the selective packaging of EVs during neuroinflammation.
Collapse
Affiliation(s)
- Natasha Vassileff
- The Department of Biochemistry and Chemistry, La Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVictoriaAustralia
| | - Jereme G. Spiers
- The Department of Biochemistry and Chemistry, La Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVictoriaAustralia
- Clear Vision Research, Eccles Institute of Neuroscience, John Curtin School of Medical Research, College of Health and MedicineThe Australian National UniversityActonAustralia
- School of Medicine and Psychology, College of Health and MedicineThe Australian National UniversityActonAustralia
| | - Sarah E. Bamford
- Centre for Materials and Surface Science and Department of Mathematical and Physical SciencesLa Trobe UniversityBundooraVictoriaAustralia
| | - Rohan G. T. Lowe
- La Trobe University Proteomics and Metabolomics PlatformLa Trobe UniversityBundooraVictoriaAustralia
| | - Keshava K. Datta
- La Trobe University Proteomics and Metabolomics PlatformLa Trobe UniversityBundooraVictoriaAustralia
| | - Paul J. Pigram
- Centre for Materials and Surface Science and Department of Mathematical and Physical SciencesLa Trobe UniversityBundooraVictoriaAustralia
| | - Andrew F. Hill
- The Department of Biochemistry and Chemistry, La Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVictoriaAustralia
- Institute for Health and SportVictoria UniversityMelbourneAustralia
| |
Collapse
|
3
|
Li Y, Pan K, Gao Y, Li J, Zang Y, Li X. Deconvoluting nitric oxide-protein interactions with spatially resolved multiplex imaging. Chem Sci 2024; 15:6562-6571. [PMID: 38699271 PMCID: PMC11062118 DOI: 10.1039/d4sc00767k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/28/2024] [Indexed: 05/05/2024] Open
Abstract
Simultaneous imaging of nitric oxide (NO) and its proximal proteins should facilitate the deconvolution of NO-protein interactions. While immunostaining is a primary assay to localize proteins in non-genetically manipulated samples, NO imaging probes with immunostaining-compatible signals remain unexplored. Herein, probe NOP-1 was developed with an NO-triggered proximal protein labeling capacity and fluorogenic signals. The trick is to fuse the native chemical ligation of acyl benzotriazole with the protein-conjugation-induced fluorogenic response of Si-rhodamine fluorophore. NOP-1 predominantly existed in the non-fluorescent spirocyclic form. Yet, its acyl o-phenylenediamine moiety was readily activated by NO into acyl benzotriazole to conjugate proximal proteins, providing a fluorogenic response and translating the transient cellular NO signal into a permanent stain compatible with immunostaining. NOP-1 was utilized to investigate NO signaling in hypoglycemia-induced neurological injury, providing direct evidence of NO-induced apoptosis during hypoglycemia. Mechanistically, multiplex imaging revealed the overlap of cellular NOP-1 fluorescence with immunofluorescence for α-tubulin and NO2-Tyr. Importantly, α-tubulin was resolved from NOP-1 labeled proteins. These results suggest that NO played a role in hypoglycemia-induced apoptosis, at least in part, through nitrating α-tubulin. This study fills a crucial gap in current imaging probes, providing a valuable tool for unraveling the complexities of NO signaling in biological processes.
Collapse
Affiliation(s)
- Yi Li
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University 866 Yuhangtang Street Hangzhou 310058 China
| | - Kaijun Pan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University Nanjing 210009 China
| | - Yanan Gao
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University Nanjing 210009 China
| | - Jia Li
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University Nanjing 210009 China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medical, Chinese Academy of Sciences Shanghai 201203 China
| | - Yi Zang
- Lingang Laboratory Shanghai 201203 China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medical, Chinese Academy of Sciences Shanghai 201203 China
| | - Xin Li
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University 866 Yuhangtang Street Hangzhou 310058 China
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University Jiashan 314100 China
| |
Collapse
|
4
|
Bonomi CG, Martorana A, Fiorelli D, Nuccetelli M, Placidi F, Mercuri NB, Motta C. Constitutive NOS Production Is Modulated by Alzheimer's Disease Pathology Depending on APOE Genotype. Int J Mol Sci 2024; 25:3725. [PMID: 38612537 PMCID: PMC11011586 DOI: 10.3390/ijms25073725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Both the endothelial (eNOS) and the neuronal (nNOS) isoforms of constitutive Nitric Oxide Synthase have been implicated in vascular dysfunctions in Alzheimer's disease (AD). We aimed to explore the relationship between amyloid pathology and NO dynamics by comparing the cerebrospinal fluid (CSF) levels of nNOS and eNOS of 8 healthy controls (HC) and 27 patients with a clinical diagnosis of Alzheimer's disease and isolated CSF amyloid changes, stratified according to APOE ε genotype (APOE ε3 = 13, APOE ε4 = 14). Moreover, we explored the associations between NOS isoforms, CSF AD biomarkers, age, sex, cognitive decline, and blood-brain barrier permeability. In our cohort, both eNOS and nNOS levels were increased in APOE ε3 with respect to HC and APOE ε4. CSF eNOS inversely correlated with CSF Amyloid-β42 selectively in carriers of APOE ε3; CSF nNOS was negatively associated with age and CSF p-tau only in the APOE ε4 subgroup. Increased eNOS could represent compensative vasodilation to face progressive Aβ-induced vasoconstriction in APOE ε3, while nNOS could represent the activation of NO-mediated plasticity strategies in the same group. Our results confirm previous findings that the APOE genotype is linked with different vascular responses to AD pathology.
Collapse
Affiliation(s)
- Chiara Giuseppina Bonomi
- UOSD Memory Clinic, Policlinico Tor Vergata, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.G.B.); (C.M.)
| | - Alessandro Martorana
- UOSD Memory Clinic, Policlinico Tor Vergata, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.G.B.); (C.M.)
| | - Denise Fiorelli
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (D.F.); (M.N.)
| | - Marzia Nuccetelli
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (D.F.); (M.N.)
| | - Fabio Placidi
- Neurology Unit, Policlinico Tor Vergata, University of Rome “Tor Vergata”, 00133 Rome, Italy; (F.P.); (N.B.M.)
| | - Nicola Biagio Mercuri
- Neurology Unit, Policlinico Tor Vergata, University of Rome “Tor Vergata”, 00133 Rome, Italy; (F.P.); (N.B.M.)
| | - Caterina Motta
- UOSD Memory Clinic, Policlinico Tor Vergata, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.G.B.); (C.M.)
| |
Collapse
|
5
|
Ma C, Zhang W, Zhang J, Du T. Modification-Specific Proteomic Analysis Reveals Cysteine S-Nitrosylation Mediated the Effect of Preslaughter Transport Stress on Pork Quality Development. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:20260-20273. [PMID: 38085829 DOI: 10.1021/acs.jafc.3c05254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
This study aimed to explore the effects of preslaughter transport stress on protein S-nitrosylation levels and S-nitrosylated proteome in post-mortem pork longissimus thoracis (LT) muscle. Pigs (N= 16) were randomly divided into 3 h transport (high-stress group, HS) and 3 h transport followed by 3 h resting treatments (low-stress control group, LS). Results demonstrated that high transport stress levels induced nitric oxide (NO) overproduction by promoting NO synthase (NOS) activity and neuronal NOS (nNOS) expression, which thereby notably increased protein S-nitrosylation levels in post-mortem muscle (p < 0.05). Proteomic analysis indicated that 133 S-nitrosylation-modified cysteines belonging to 85 proteins were significantly differential, of which 101 cysteines of 63 proteins were higher in the HS group (p < 0.05). Differential proteins including cytoskeletal and calcium-handling proteins, glycolytic enzymes, and oxidoreductase were mainly involved in the regulation of muscle contraction and energy metabolism that might together mediate meat quality development. Overall, this study provided direct evidence for changes in S-nitrosylation levels and proteome in post-mortem muscle in response to preslaughter transport stress and revealed the potential impact of S-nitrosylated proteins on meat quality.
Collapse
Affiliation(s)
- Chao Ma
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wangang Zhang
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jian Zhang
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Tongyao Du
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
6
|
Clemente-Suárez VJ, Redondo-Flórez L, Beltrán-Velasco AI, Ramos-Campo DJ, Belinchón-deMiguel P, Martinez-Guardado I, Dalamitros AA, Yáñez-Sepúlveda R, Martín-Rodríguez A, Tornero-Aguilera JF. Mitochondria and Brain Disease: A Comprehensive Review of Pathological Mechanisms and Therapeutic Opportunities. Biomedicines 2023; 11:2488. [PMID: 37760929 PMCID: PMC10526226 DOI: 10.3390/biomedicines11092488] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Mitochondria play a vital role in maintaining cellular energy homeostasis, regulating apoptosis, and controlling redox signaling. Dysfunction of mitochondria has been implicated in the pathogenesis of various brain diseases, including neurodegenerative disorders, stroke, and psychiatric illnesses. This review paper provides a comprehensive overview of the intricate relationship between mitochondria and brain disease, focusing on the underlying pathological mechanisms and exploring potential therapeutic opportunities. The review covers key topics such as mitochondrial DNA mutations, impaired oxidative phosphorylation, mitochondrial dynamics, calcium dysregulation, and reactive oxygen species generation in the context of brain disease. Additionally, it discusses emerging strategies targeting mitochondrial dysfunction, including mitochondrial protective agents, metabolic modulators, and gene therapy approaches. By critically analysing the existing literature and recent advancements, this review aims to enhance our understanding of the multifaceted role of mitochondria in brain disease and shed light on novel therapeutic interventions.
Collapse
Affiliation(s)
- Vicente Javier Clemente-Suárez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (V.J.C.-S.); (J.F.T.-A.)
- Group de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| | - Laura Redondo-Flórez
- Department of Health Sciences, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, C/Tajo s/n, Villaviciosa de Odón, 28670 Madrid, Spain
| | - Ana Isabel Beltrán-Velasco
- Psychology Department, Facultad de Ciencias de la Vida y la Naturaleza, Universidad Antonio de Nebrija, 28240 Madrid, Spain
| | - Domingo Jesús Ramos-Campo
- LFE Research Group, Department of Health and Human Performance, Faculty of Physical Activity and Sport Science-INEF, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Pedro Belinchón-deMiguel
- Department of Nursing and Nutrition, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain;
| | | | - Athanasios A. Dalamitros
- Laboratory of Evaluation of Human Biological Performance, School of Physical Education and Sport Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Rodrigo Yáñez-Sepúlveda
- Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 2520000, Chile;
| | - Alexandra Martín-Rodríguez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (V.J.C.-S.); (J.F.T.-A.)
| | | |
Collapse
|
7
|
Singh N, Sherin GR, Mugesh G. Antioxidant and Prooxidant Nanozymes: From Cellular Redox Regulation to Next-Generation Therapeutics. Angew Chem Int Ed Engl 2023; 62:e202301232. [PMID: 37083312 DOI: 10.1002/anie.202301232] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 04/22/2023]
Abstract
Nanozymes, nanomaterials with enzyme-mimicking activity, have attracted tremendous interest in recent years owing to their ability to replace natural enzymes in various biomedical applications, such as biosensing, therapeutics, drug delivery, and bioimaging. In particular, the nanozymes capable of regulating the cellular redox status by mimicking the antioxidant enzymes in mammalian cells are of great therapeutic significance in oxidative-stress-mediated disorders. As the distinction of physiological oxidative stress (oxidative eustress) and pathological oxidative stress (oxidative distress) occurs at a fine borderline, it is a great challenge to design nanozymes that can differentially sense the two extremes in cells, tissues and organs and mediate appropriate redox chemical reactions. In this Review, we summarize the advances in the development of redox-active nanozymes and their biomedical applications. We primarily highlight the therapeutic significance of the antioxidant and prooxidant nanozymes in various disease model systems, such as cancer, neurodegeneration, and cardiovascular diseases. The future perspectives of this emerging area of research and the challenges associated with the biomedical applications of nanozymes are described.
Collapse
Affiliation(s)
- Namrata Singh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
- Current address: Department of Medical Biochemistry and Biophysics, Karolinska Institute, Biomedicum, Solnavägen 9, 171 65, Solna, Sweden
| | - G R Sherin
- 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
|
8
|
Iova OM, Marin GE, Lazar I, Stanescu I, Dogaru G, Nicula CA, Bulboacă AE. Nitric Oxide/Nitric Oxide Synthase System in the Pathogenesis of Neurodegenerative Disorders-An Overview. Antioxidants (Basel) 2023; 12:antiox12030753. [PMID: 36979000 PMCID: PMC10045816 DOI: 10.3390/antiox12030753] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/24/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Nitric oxide, a ubiquitous molecule found throughout the natural world, is a key molecule implicated in many central and benefic molecular pathways and has a well-established role in the function of the central nervous system, as numerous studies have previously shown. Dysregulation of its metabolism, mainly the upregulation of nitric oxide production, has been proposed as a trigger and/or aggravator for many neurological affections. Increasing evidence supports the implication of this molecule in prevalent neurodegenerative diseases, such as Parkinson's disease, Alzheimer's disease, or amyotrophic lateral sclerosis. The mechanisms proposed for its neurotoxicity mainly center around the increased quantities of nitric oxide that are produced in the brain, their cause, and, most importantly, the pathological metabolic cascades created. These cascades lead to the formation of neuronal toxic substances that impair the neurons' function and structure on multiple levels. The purpose of this review is to present the main causes of increased pathological production, as well as the most important pathophysiological mechanisms triggered by nitric oxide, mechanisms that could help explain a part of the complex picture of neurodegenerative diseases and help develop targeted therapies.
Collapse
Affiliation(s)
- Olga-Maria Iova
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Gheorghe-Eduard Marin
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Izabella Lazar
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ioana Stanescu
- Department of Neurology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Gabriela Dogaru
- Department of Physical Medicine and Rehabilitation, Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Viilor Street, No. 46-50, 400347 Cluj-Napoca, Romania
| | - Cristina Ariadna Nicula
- Department of Ophthalmology, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Adriana Elena Bulboacă
- Department of Pathophysiology, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
9
|
Zhao W, Spiers JG, Vassileff N, Khadka A, Jaehne EJ, van den Buuse M, Hill AF. microRNA-146a modulates behavioural activity, neuroinflammation, and oxidative stress in adult mice. Mol Cell Neurosci 2023; 124:103820. [PMID: 36736750 DOI: 10.1016/j.mcn.2023.103820] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Small non-coding miRNA act as key regulators of several physiological processes due to their ability to interact with numerous target mRNA within a network. Whilst several miRNA can act in concert to regulate target mRNA expression, miR-146a has emerged as a critical modulator of inflammation by targeting key upstream signalling proteins of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway and reductions in this miRNA have been observed in several neurological and neurodegenerative disorders. However, a targeted assessment of behaviour and neural tissues following the loss of miR-146a has not been documented. In this study, we examined the behavioural and neuroinflammatory phenotype of mice lacking miR-146a to determine the role of this miRNA in neurological function. Adult miR-146a-/- mice displayed no overt developmental phenotype with the exception of enlarged spleens. Behavioural testing revealed a mild but significant reduction in exploratory locomotor activity and increase in anxiety-like behaviour, with no changes in short-term spatial memory, fear conditioning, or sensorimotor gating. In the brain, the lack of miR-146a resulted in a significant compensatory miR-155 expression with no significant changes in expression of the target Interleukin 1 Receptor Associated Kinase (Irak) gene family. Despite these effects on upstream NF-κB mediators, downstream expression of cytokine and chemokine messengers was significantly elevated in miR-146a-/- mice compared to wild-type controls. Moreover, this increase in inflammatory cytokines was observed alongside an induction of oxidative stress, driven in part by nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase, and included reduced thiol antioxidant concentrations and increased oxidised protein carbonyl concentrations. In female miR-146a mice, this increase in oxidative stress resulted in an increased expression of superoxide dismutase 1 (SOD1). Together, this suggests miR-146a plays a key role in regulating inflammation even in the absence of inflammatory stimuli and reduced levels of this miRNA have the capacity to induce limited behavioural effects whilst exacerbating both inflammation and oxidative stress in the brain.
Collapse
Affiliation(s)
- Wenting Zhao
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3083, Australia
| | - Jereme G Spiers
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3083, Australia
| | - Natasha Vassileff
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3083, Australia
| | - Arun Khadka
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3083, Australia
| | - Emily J Jaehne
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3083, Australia
| | - Maarten van den Buuse
- Department of Psychology, Counselling and Therapy, School of Psychology and Public Health, La Trobe University, Melbourne, Australia; Department of Pharmacology, University of Melbourne, Melbourne, Australia
| | - Andrew F Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3083, Australia; Institute for Health and Sport, Victoria University, Footscray, Melbourne, Australia.
| |
Collapse
|
10
|
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
|
11
|
Vrettou S, Wirth B. S-Glutathionylation and S-Nitrosylation in Mitochondria: Focus on Homeostasis and Neurodegenerative Diseases. Int J Mol Sci 2022; 23:15849. [PMID: 36555492 PMCID: PMC9779533 DOI: 10.3390/ijms232415849] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/24/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Redox post-translational modifications are derived from fluctuations in the redox potential and modulate protein function, localization, activity and structure. Amongst the oxidative reversible modifications, the S-glutathionylation of proteins was the first to be characterized as a post-translational modification, which primarily protects proteins from irreversible oxidation. However, a growing body of evidence suggests that S-glutathionylation plays a key role in core cell processes, particularly in mitochondria, which are the main source of reactive oxygen species. S-nitrosylation, another post-translational modification, was identified >150 years ago, but it was re-introduced as a prototype cell-signaling mechanism only recently, one that tightly regulates core processes within the cell’s sub-compartments, especially in mitochondria. S-glutathionylation and S-nitrosylation are modulated by fluctuations in reactive oxygen and nitrogen species and, in turn, orchestrate mitochondrial bioenergetics machinery, morphology, nutrients metabolism and apoptosis. In many neurodegenerative disorders, mitochondria dysfunction and oxidative/nitrosative stresses trigger or exacerbate their pathologies. Despite the substantial amount of research for most of these disorders, there are no successful treatments, while antioxidant supplementation failed in the majority of clinical trials. Herein, we discuss how S-glutathionylation and S-nitrosylation interfere in mitochondrial homeostasis and how the deregulation of these modifications is associated with Alzheimer’s, Parkinson’s, amyotrophic lateral sclerosis and Friedreich’s ataxia.
Collapse
Affiliation(s)
- Sofia Vrettou
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Rare Diseases, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
12
|
Spiers JG, Cortina Chen HJ, Barry TL, Bourgognon JM, Steinert JR. Redox stress and metal dys-homeostasis appear as hallmarks of early prion disease pathogenesis in mice. Free Radic Biol Med 2022; 192:182-190. [PMID: 36170956 DOI: 10.1016/j.freeradbiomed.2022.09.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022]
Abstract
Neurodegenerative diseases are associated with a multitude of dysfunctional cellular pathways. One major contributory factor is a redox stress challenge during the development of several protein misfolding conditions including Alzheimer's (AD), Parkinson's disease (PD), and less common conditions such as Creutzfeldt Jakob disease (CJD). CJD is caused by misfolding of the neuronal prion protein and is characterised by a neurotoxic unfolded protein response involving chronic endoplasmic reticulum stress, reduced protein translation and spongiosis leading subsequently to synaptic and neuronal loss. Here we have characterised prion disease in mice to assess redox stress components including nitrergic and oxidative markers associated with neuroinflammatory activation. Aberrant regulation of the homeostasis of several redox metals contributes to the overall cellular redox stress and we have identified altered levels of iron, copper, zinc, and manganese in the hippocampus of prion diseased mice. Our data show that early in disease, there is evidence for oxidative stress in conjunction with reduced antioxidant superoxide dismutase mRNA and protein expression. Moreover, expression of divalent metal transporter proteins was reduced for Atp7b, Atox1, Slc11a2, Slc39a14 at 6-7 weeks but increased for Slc39a14 and Mt1 at 10 weeks of disease. Our data present evidence for a strong pro-oxidant environment and altered redox metal homeostasis in early disease pathology which both may be contributory factors to aggravating this protein misfolding disease.
Collapse
Affiliation(s)
- Jereme G Spiers
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, 3083, Australia.
| | - Hsiao-Jou Cortina Chen
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Tiffany L Barry
- School of Geography, Geology and the Environment, University of Leicester, University Road, Leicester, LE1 7RH, UK
| | - Julie-Myrtille Bourgognon
- School of Infection and Immunity, Sir Graeme Davies Building, 120 University Place, Glasgow, G12 8TA, UK
| | - Joern R Steinert
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham, NG7 2NR, UK.
| |
Collapse
|
13
|
Rahdar M, Hajisoltani R, Davoudi S, Karimi SA, Borjkhani M, Khatibi VA, Hosseinmardi N, Behzadi G, Janahmadi M. Alterations in the intrinsic discharge activity of CA1 pyramidal neurons associated with possible changes in the NADPH diaphorase activity in a rat model of autism induced by prenatal exposure to valproic acid. Brain Res 2022; 1792:148013. [PMID: 35841982 DOI: 10.1016/j.brainres.2022.148013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/24/2022] [Accepted: 07/10/2022] [Indexed: 11/02/2022]
Abstract
Autism spectrum disorder is a neurodevelopmental disorder characterized by sensory abnormalities, social skills impairment and cognitive deficits. Although recent evidence indicated that induction of autism-like behavior in animal models causes abnormal neuronal excitability, the impact of autism on neuronal properties is still an important issue. Thus, new findings at the cellular level may shed light on the pathophysiology of autism and may help to find effective treatment strategies. Here, we investigated the behavioral, electrophysiological and histochemical impacts of prenatal exposure to valproic acid (VPA) in rats. Findings revealed that VPA exposure caused a significant increase in the hot plate response latency. The novel object recognition ability was also impaired in VPA-exposed rats. Along with these behavioral alterations, neurons from VPA-exposed animals exhibited altered excitability features in response to depolarizing current injections relative to control neurons. In the VPA-exposed group, these changes consisted of a significant increase in the amplitude, evoked firing frequency and the steady-state standard deviation of spike timing of action potentials (APs). Moreover, the half-width, the AHP amplitude and the decay time constant of APs were significantly decreased in this group. These changes in the evoked electrophysiological properties were accompanied by intrinsic hyperexcitability and lower spike-frequency adaptation and also a significant increase in the number of NADPH-diaphorase stained neurons in the hippocampal CA1 area of the VPA-exposed rats. Taken together, findings demonstrate that abnormal nociception and recognition memory is associated with alterations in the neuronal responsiveness and nitrergic system in a rat model of autism-like.
Collapse
Affiliation(s)
- Mona Rahdar
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Razieh Hajisoltani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shima Davoudi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Asaad Karimi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Tehran, Iran
| | - Mehdi Borjkhani
- Department of Electrical Engineering, Urmia University of Technology, Urmia, Iran
| | - Vahid Ahli Khatibi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Narges Hosseinmardi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gila Behzadi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Zhang P, Fang H, Lou C, Ye S, Shen G, Chen S, Amin N, Botchway BOA, Fang M. Enhanced Glial Reaction and Altered Neuronal Nitric Oxide Synthase are Implicated in Attention Deficit Hyperactivity Disorder. Front Cell Dev Biol 2022; 10:901093. [PMID: 35800894 PMCID: PMC9255429 DOI: 10.3389/fcell.2022.901093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/25/2022] [Indexed: 11/22/2022] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) has a complex etiology, and its specific causal factors remain to be elucidated. Aberration of nitric oxide synthase (nNOS) and inflammation, together with astrocytic and microglial cells have been continually associated with several neurological disorders, including ADHD. Using spontaneously hypertensive rat (SHR), we investigated the changes in nNOS, inflammatory, microglial and astrocytic markers in the frontal cortex and hippocampus at three different ages: onset of hypertension stage (i.e., 6 weeks after birth of SHR), established hypertension stage (i.e., 12 weeks after birth of SHR) and senescent stage (i.e., 12 months after birth of SHR), and compared with its age-matched normotensive control, Wistar-Kyoto (WKY) rats. A significant upregulation of Iba-1 expression in the senescent stage of SHR was observed. Further, we observed an upregulated nNOS expression in both onset and established stages of SHR, and a downregulated nNOS in the senescent stage. Our study showed an age-related increment of astrogliosis in the cortex and hippocampi of aged SHR. On the basis of our results, alterations in the nNOS and Iba-1 expressions, as well as age-related astrogliosis, may contribute to ADHD pathogenesis.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Psychiatry, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Huyue Fang
- Department of Psychiatry, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Chengjian Lou
- Department of Neurosurgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Shan Ye
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Guanghong Shen
- The Affiliated People’s Hospital of Hangzhou Medical College, Hangzhou Medical College, Hangzhou, China
| | - Shijia Chen
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Nashwa Amin
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
- Department of Zoology, Faculty of Science, Aswan University, Aswan, Egypt
| | | | - Marong Fang
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Marong Fang,
| |
Collapse
|
15
|
Scuto M, Modafferi S, Rampulla F, Zimbone V, Tomasello M, Spano’ S, Ontario M, Palmeri A, Trovato Salinaro A, Siracusa R, Di Paola R, Cuzzocrea S, Calabrese E, Wenzel U, Calabrese V. Redox modulation of stress resilience by Crocus Sativus L. for potential neuroprotective and anti-neuroinflammatory applications in brain disorders: From molecular basis to therapy. Mech Ageing Dev 2022; 205:111686. [DOI: 10.1016/j.mad.2022.111686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 12/13/2022]
|
16
|
Simultaneous administration of bromodomain and histone deacetylase I inhibitors alleviates cognition deficit in Alzheimer's model of rats. Brain Res Bull 2021; 179:49-56. [PMID: 34915044 DOI: 10.1016/j.brainresbull.2021.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Histone deacetylases (HDACs) target various genes responsible for cognitive functions. However, chromatin readers, particularly bromodomain-containing protein 4 (BRD4), are capable to change the final products of genes. The objective of this study was to evaluate the simultaneous effects of inhibition of HDACs and BRD4 on spatial and aversive memories impaired by amyloid β (Aβ) in a rat model of Alzheimer's disease (AD) considering CREB and TNF-α signaling. METHODS Forty male Wistar rats aged 3 months were randomly divided into five groups: saline +DMSO, Aβ+saline+DMSO, Aβ+JQ1, Aβ+MS-275, Aβ+JQ1+MS-275, and received the related treatments. MS-275, is the second generation of HDACs inhibitor, and JQ1 is a potent inhibitor of the BET family of bromodomain proteins in mammals. After the treatments, cognitive function was assessed by Morris water maze (MWM) and passive avoidance learning (PAL). The hippocampal level of mRNA for CREB and TNF-α, and also phosphorylated CREB were measured using real-time PCR and western blotting respectively. RESULTS Administration of JQ1 and MS-275, either separately or simultaneously, improved acquisition and retrieval of spatial and aversive memories as it was evident by decreased escape latency and increased time spent in the target quadrant (TTS) in Morris water maze (MWM), together with increase in step-through latency, but reduced time spent in the dark zone time in passive avoidance learning (PAL) compared with Aβ+saline+DMSO. Furthermore, there was a significant rise in the hippocampal level of CREB mRNA and phosphorylated CREB, but a reduction in TNF-α expression in comparison with Aβ + Saline. CONCLUSION Simultaneous administration of JQ1 and MS-275 improves acquisition and retrieval of both spatial and aversive memories partly via CREB and TNF-α signaling with no superiority to monotherapy.
Collapse
|
17
|
Dietary uptake of Salvia macilenta extract improves Nrf2 antioxidant signaling pathway and diminishes inflammation and apoptosis in amyloid beta-induced rats. Mol Biol Rep 2021; 48:7667-7676. [PMID: 34724130 DOI: 10.1007/s11033-021-06772-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/22/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Studies showed the protective role of Salvia in traditional medicine against neurodegenerative diseases. Salvia macilenta is one of the potent antioxidant herbs among Salvia species against oxidative stress. In the current study, the effect of oral administration of S. macilenta in the antioxidant, anti-inflammatory activities of Aβ-injected male albino Wistar rats was determined. METHODS Rats were received S. macilenta (50 mg/kg/day) orally, for ten successive days and then some of them received Aβ (10 ng/µl) in their hippocampus (CA1 region). Proteins involved in antioxidant defense system and inflammatory signaling pathways in the hippocampus and prefrontal cortex were evaluated using Western blotting technique. To study apoptosis, Western blotting technique and histological staining were used. Catalase activity, glutathione peroxidase (GSH) and nitric oxide levels were measured. RESULTS Results demonstrated that S. macilenta increased Nrf2 protein level and decreased TNFα and IL-6 protein level in Aβ-injected rats compared to the Aβ-injected group in the hippocampus and prefrontal cortex. Histological analysis showed pretreatment with S. macilenta decreased apoptosis levels in the hippocampus and prefrontal cortex, about 41 and 42%, compared to Aβ-injected rats, respectively. This study showed that catalase activity was changed in the S. macilenta + Aβ group compared to the Aβ-injected rats. Also, GSH level was increased in the S. macilenta + Aβ group compared to the Aβ-injected rat. CONCLUSION Orally treatment of S. macilenta extract in Aβ-injected rats could ameliorate protective pathways and, so, it can be one of the proposed dietary supplements for the prevention of Alzheimer's disease and dementia.
Collapse
|
18
|
Inhibition of neuroinflammatory nitric oxide signaling suppresses glycation and prevents neuronal dysfunction in mouse prion disease. Proc Natl Acad Sci U S A 2021; 118:2009579118. [PMID: 33653950 PMCID: PMC7958397 DOI: 10.1073/pnas.2009579118] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Several neurodegenerative diseases associated with protein misfolding (Alzheimer's and Parkinson's disease) exhibit oxidative and nitrergic stress following initiation of neuroinflammatory pathways. Associated nitric oxide (NO)-mediated posttranslational modifications impact upon protein functions that can exacerbate pathology. Nonenzymatic and irreversible glycation signaling has been implicated as an underlying pathway that promotes protein misfolding, but the direct interactions between both pathways are poorly understood. Here we investigated the therapeutic potential of pharmacologically suppressing neuroinflammatory NO signaling during early disease progression of prion-infected mice. Mice were injected daily with an NO synthase (NOS) inhibitor at early disease stages, hippocampal gene and protein expression levels of oxidative and nitrergic stress markers were analyzed, and electrophysiological characterization of pyramidal CA1 neurons was performed. Increased neuroinflammatory signaling was observed in mice between 6 and 10 wk postinoculation (w.p.i.) with scrapie prion protein. Their hippocampi were characterized by enhanced nitrergic stress associated with a decline in neuronal function by 9 w.p.i. Daily in vivo administration of the NOS inhibitor L-NAME between 6 and 9 w.p.i. at 20 mg/kg prevented the functional degeneration of hippocampal neurons in prion-diseased mice. We further found that this intervention in diseased mice reduced 3-nitrotyrosination of triose-phosphate isomerase, an enzyme involved in the formation of disease-associated glycation. Furthermore, L-NAME application led to a reduced expression of the receptor for advanced glycation end-products and the diminished accumulation of hippocampal prion misfolding. Our data suggest that suppressing neuroinflammatory NO signaling slows functional neurodegeneration and reduces nitrergic and glycation-associated cellular stress.
Collapse
|
19
|
Gheibihayat SM, Cabezas R, Nikiforov NG, Jamialahmadi T, Johnston TP, Sahebkar A. CD47 in the Brain and Neurodegeneration: An Update on the Role in Neuroinflammatory Pathways. Molecules 2021; 26:molecules26133943. [PMID: 34203368 PMCID: PMC8271744 DOI: 10.3390/molecules26133943] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/16/2021] [Accepted: 06/22/2021] [Indexed: 01/02/2023] Open
Abstract
CD47 is a receptor belonging to the immunoglobulin (Ig) superfamily and broadly expressed on cell membranes. Through interactions with ligands such as SIRPα, TSP-1, integrins, and SH2-domain bearing protein tyrosine phosphatase substrate-1 (SHPS-1), CD47 regulates numerous functions like cell adhesion, proliferation, apoptosis, migration, homeostasis, and the immune system. In this aspect, previous research has shown that CD47 modulates phagocytosis via macrophages, the transmigration of neutrophils, and the activation of T-cells, dendritic cells, and B-cells. Moreover, several studies have reported the increased expression of the CD47 receptor in a variety of diseases, including acute lymphoblastic leukemia (ALL), chronic myeloid leukemia, non-Hodgkin’s lymphoma (NHL), multiple myeloma (MM), bladder cancer, acute myeloid leukemia (AML), Gaucher disease, Multiple Sclerosis and stroke among others. The ubiquitous expression of the CD47 cell receptor on most resident cells of the CNS has previously been established through different methodologies. However, there is little information concerning its precise functions in the development of different neurodegenerative pathologies in the CNS. Consequently, further research pertaining to the specific functions and roles of CD47 and SIRP is required prior to its exploitation as a druggable approach for the targeting of various neurodegenerative diseases that affect the human population. The present review attempts to summarize the role of both CD47 and SIRP and their therapeutic potential in neurodegenerative disorders.
Collapse
Affiliation(s)
- Seyed Mohammad Gheibihayat
- Department of Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd 8916188635, Iran;
| | - Ricardo Cabezas
- Department of Physiology, School of Medicine, Universidad Antonio Nariño, Bogotá 111511, Colombia;
| | - Nikita G. Nikiforov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, Tsyurupa Street, 117418 Moscow, Russia;
- Laboratory of Medical Genetics, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 121552 Moscow, Russia
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan 19395/1495, Iran;
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran
| | - Thomas P. Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO 64131, USA;
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran
- School of Medicine, The University of Western Australia, Perth 6907, Australia
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran
- Correspondence: or
| |
Collapse
|
20
|
Gas phase microdialysis and chemiluminescence detection: A small, fast, selective, and sensitive method to monitor aqueous nitric oxide. Talanta 2021; 233:122599. [PMID: 34215087 DOI: 10.1016/j.talanta.2021.122599] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 01/06/2023]
Abstract
A method using a gas-phase microdialysis probe interfaced with a modified commercially available nitric oxide (NO) detector is shown to selectively measure aqueous NO at low μM levels with high selectivity. The detector measures chemiluminescence resulting from the gas-phase reaction of NO with ozone. The microdialysis probe is small enough (3 mm × 200 μm) to be used in vivo. Because the processes of extraction across the microdialysis membrane and transport from the probe to the detector are both very fast, the response time is shorter than 5 s. The method was verified using two different quantifiable sources of NO: nitrite and methylamine hexamethylene methylamine (MAHMA) NONOates. To demonstrate ruggedness and to show the impact of matrix on NO generation, the method was used to measure NO in a cell culture matrix. The continuous extraction, fast response time, and rugged nature make the method useful for monitoring NO in biological applications. Our results also show that predicting NO concentration for in vitro experiments based on NONOate concentration may be a poor assumption due to the pH dependence of NO formation and the rapid decline in NO concentration.
Collapse
|
21
|
Zhu HY, Hong FF, Yang SL. The Roles of Nitric Oxide Synthase/Nitric Oxide Pathway in the Pathology of Vascular Dementia and Related Therapeutic Approaches. Int J Mol Sci 2021; 22:ijms22094540. [PMID: 33926146 PMCID: PMC8123648 DOI: 10.3390/ijms22094540] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022] Open
Abstract
Vascular dementia (VaD) is the second most common form of dementia worldwide. It is caused by cerebrovascular disease, and patients often show severe impairments of advanced cognitive abilities. Nitric oxide synthase (NOS) and nitric oxide (NO) play vital roles in the pathogenesis of VaD. The functions of NO are determined by its concentration and bioavailability, which are regulated by NOS activity. The activities of different NOS subtypes in the brain are partitioned. Pathologically, endothelial NOS is inactivated, which causes insufficient NO production and aggravates oxidative stress before inducing cerebrovascular endothelial dysfunction, while neuronal NOS is overactive and can produce excessive NO to cause neurotoxicity. Meanwhile, inflammation stimulates the massive expression of inducible NOS, which also produces excessive NO and then induces neuroinflammation. The vicious circle of these kinds of damage having impacts on each other finally leads to VaD. This review summarizes the roles of the NOS/NO pathway in the pathology of VaD and also proposes some potential therapeutic methods that target this pathway in the hope of inspiring novel ideas for VaD therapeutic approaches.
Collapse
Affiliation(s)
- Han-Yan Zhu
- Department of Physiology, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China;
- Queen Marry College, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China
| | - Fen-Fang Hong
- Teaching Center, Department of Experimental, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China
- Correspondence: (F.-F.H.); (S.-L.Y.)
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China;
- Correspondence: (F.-F.H.); (S.-L.Y.)
| |
Collapse
|
22
|
Martorell M, Lucas X, Alarcón-Zapata P, Capó X, Quetglas-Llabrés MM, Tejada S, Sureda A. Targeting Xanthine Oxidase by Natural Products as a Therapeutic Approach for Mental Disorders. Curr Pharm Des 2021; 27:367-382. [PMID: 32564744 DOI: 10.2174/1381612826666200621165839] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/08/2020] [Indexed: 11/22/2022]
Abstract
Mental disorders comprise diverse human pathologies, including depression, bipolar affective disorder, schizophrenia, and dementia that affect millions of people around the world. The causes of mental disorders are unclear, but growing evidence suggests that oxidative stress and the purine/adenosine system play a key role in their development and progression. Xanthine oxidase (XO) is a flavoprotein enzyme essential for the catalysis of the oxidative hydroxylation of purines -hypoxanthine and xanthine- to generate uric acid. As a consequence of the oxidative reaction of XO, reactive oxygen species (ROS) such as superoxide and hydrogen peroxide are produced and, further, contribute to the pathogenesis of mental disorders. Altered XO activity has been associated with free radical-mediated neurotoxicity inducing cell damage and inflammation. Diverse studies reported a direct association between an increased activity of XO and diverse mental diseases including depression or schizophrenia. Small-molecule inhibitors, such as the well-known allopurinol, and dietary flavonoids, can modulate the XO activity and subsequent ROS production. In the present work, we review the available literature on XO inhibition by small molecules and their potential therapeutic application in mental disorders. In addition, we discuss the chemistry and molecular mechanism of XO inhibitors, as well as the use of structure-based and computational methods to design specific inhibitors with the capability of modulating XO activity.
Collapse
Affiliation(s)
- Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, and Centre for Healthy Living, University of Concepcion, 4070386 Concepcion, Chile
| | - Xavier Lucas
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel CH-4070, Switzerland
| | - Pedro Alarcón-Zapata
- Clinical Biochemistry and Immunology Department, Faculty of Pharmacy, University of Concepcion, 4070386 Concepcion, Chile
| | - Xavier Capó
- Research Group in Community Nutrition and Oxidative Stress, University of Balearic Islands & Health Research Institute of the Balearic Islands (IdISBa), E-07122, Palma, Balearic Islands, Spain
| | - Maria Magdalena Quetglas-Llabrés
- Laboratory of Neurophysiology, Department of Biology, University of Balearic Islands & Health Research Institute of the Balearic Islands (IdISBa), E-07122, Palma, Balearic Islands, Spain
| | - Silvia Tejada
- Laboratory of Neurophysiology, Department of Biology, University of Balearic Islands & Health Research Institute of the Balearic Islands (IdISBa), E-07122, Palma, Balearic Islands, Spain
| | - Antoni Sureda
- Research Group in Community Nutrition and Oxidative Stress, University of Balearic Islands & Health Research Institute of the Balearic Islands (IdISBa), E-07122, Palma, Balearic Islands, Spain
| |
Collapse
|
23
|
Rivastigmine attenuates the Alzheimer's disease related protein degradation and apoptotic neuronal death signalling. Biochem J 2021; 478:1435-1451. [PMID: 33660768 DOI: 10.1042/bcj20200754] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022]
Abstract
Rivastigmine is a clinical drug for patients of Alzheimer's disease (AD) exerting its inhibitory effect on acetylcholinesterase activity however, its effect on other disease-related pathological mechanisms are not yet known. This study was conducted to evaluate the effect of rivastigmine on protein aggregation and degradation related mechanisms employing streptozotocin (STZ) induced experimental rat model. The known inhibitory effect of rivastigmine on cognition and acetylcholinesterase activity was observed in both cortex and hippocampus and further its effect on tau level, amyloid aggregation, biochemical alterations, endoplasmic reticulum (ER) stress, calcium homeostasis, proteasome activity and apoptosis was estimated. STZ administration in rat brain caused significant cognitive impairment, augmented acetylcholinesterase activity, tau phosphorylation and amyloid aggregation which were significantly inhibited with rivastigmine treatment. STZ also caused significant biochemical alterations which were attenuated with rivastigmine treatment. Since AD pathology is related to protein aggregation and we have found disease-related amyloid aggregation, further the investigation was done to decipher the ER functionality and apoptotic signalling. STZ caused significantly altered level of ER stress related markers (GRP78, GADD153 and caspase-12) which were significantly inhibited with rivastigmine treatment. Furthermore, the effect of rivastigmine was estimated on proteasome activity in both regions. Rivastigmine treatment significantly enhances the proteasome activity and may contributes in removal of amyloid aggregation. In conclusion, findings suggested that along with inhibitory effect of rivastigmine on acetylcholinesterase activity and up to some extent on cognition, it has significant effect on disease-related biochemical alterations, ER functionality, protein degradation machinery and neuronal apoptosis.
Collapse
|
24
|
Patthy Á, Murai J, Hanics J, Pintér A, Zahola P, Hökfelt TGM, Harkany T, Alpár A. Neuropathology of the Brainstem to Mechanistically Understand and to Treat Alzheimer's Disease. J Clin Med 2021; 10:jcm10081555. [PMID: 33917176 PMCID: PMC8067882 DOI: 10.3390/jcm10081555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder as yet without effective therapy. Symptoms of this disorder typically reflect cortical malfunction with local neurohistopathology, which biased investigators to search for focal triggers and molecular mechanisms. Cortex, however, receives massive afferents from caudal brain structures, which do not only convey specific information but powerfully tune ensemble activity. Moreover, there is evidence that the start of AD is subcortical. The brainstem harbors monoamine systems, which establish a dense innervation in both allo- and neocortex. Monoaminergic synapses can co-release neuropeptides either by precisely terminating on cortical neurons or, when being “en passant”, can instigate local volume transmission. Especially due to its early damage, malfunction of the ascending monoaminergic system emerges as an early sign and possible trigger of AD. This review summarizes the involvement and cascaded impairment of brainstem monoaminergic neurons in AD and discusses cellular mechanisms that lead to their dysfunction. We highlight the significance and therapeutic challenges of transmitter co-release in ascending activating system, describe the role and changes of local connections and distant afferents of brainstem nuclei in AD, and summon the rapidly increasing diagnostic window during the last few years.
Collapse
Affiliation(s)
- Ágoston Patthy
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - János Murai
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - János Hanics
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, H-1094 Budapest, Hungary
| | - Anna Pintér
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - Péter Zahola
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - Tomas G. M. Hökfelt
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, 17165 Stockholm, Sweden; (T.G.M.H.); (T.H.)
| | - Tibor Harkany
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, 17165 Stockholm, Sweden; (T.G.M.H.); (T.H.)
- Center for Brain Research, Department of Molecular Neurosciences, Medical University of Vienna, 1090 Vienna, Austria
| | - Alán Alpár
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, H-1094 Budapest, Hungary
- Correspondence:
| |
Collapse
|
25
|
A review on α-mangostin as a potential multi-target-directed ligand for Alzheimer's disease. Eur J Pharmacol 2021; 897:173950. [PMID: 33607107 DOI: 10.1016/j.ejphar.2021.173950] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 02/03/2021] [Accepted: 02/12/2021] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by progressive memory loss, declining language skills and other cognitive disorders. AD has brought great mental and economic burden to patients, families and society. However due to the complexity of AD's pathology, drugs developed for the treatment of AD often fail in clinical or experimental trials. The main problems of current anti-AD drugs are low efficacy due to mono-target method or side effects, especially high hepatotoxicity. To tackle these two main problems, multi-target-directed ligand (MTDL) based on "one molecule, multiple targets" has been studied. MTDLs can regulate multiple biological targets at the same time, so it has shown higher efficacy, better safety. As a natural active small molecule, α-mangostin (α-M) has shown potential multi-factor anti-AD activities in a series of studies, furthermore it also has a certain hepatoprotective effect. The good availability of α-M also provides support for its application in clinical research. In this work, multiple activities of α-M related to AD therapy were reviewed, which included anti-cholinesterase, anti-amyloid-cascade, anti-inflammation, anti-oxidative stress, low toxicity, hepatoprotective effects and drug formulation. It shows that α-M is a promising candidate for the treatment of AD.
Collapse
|
26
|
Aversano A, Rossi FW, Cammarota F, De Paulis A, Izzo P, De Rosa M. Nitrodi thermal water downregulates protein S‑nitrosylation in RKO cells. Int J Mol Med 2020; 46:1359-1366. [PMID: 32945437 PMCID: PMC7447308 DOI: 10.3892/ijmm.2020.4676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 06/23/2020] [Indexed: 11/07/2022] Open
Abstract
Balneotherapy and spa therapy have been used in the treatment of ailments since time immemorial. Moreover, there is evidence to suggest that the beneficial effects of thermal water continue for months following the completion of treatment. The mechanisms through which thermal water exerts its healing effects remain unknown. Both balneological and hydroponic therapy at 'the oldest spa in the world', namely, the Nitrodi spring on the Island of Ischia (Southern Italy) are effective in a number of diseases and conditions. The aim of the present study was to investigate the molecular basis underlying the therapeutic effects of Nitrodi spring water in low-grade inflammation and stress-related conditions. For this purpose, an in vitro model was devised in which RKO colorectal adenocarcinoma cells were treated with phosphate-buffered saline or phosphate-buffered saline prepared with Nitrodi water for 4 h daily, 5 days a week for 6 weeks. The RKO cells were then subjected to the following assays: 3-(4,5- Dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide assay, Transwell migration assay, western blot analysis, the fluorimetric detection of protein S-nitrosothiols and S-nitrosylation western blot analysis. The results revealed that Nitrodi spring water promoted cell migration and cell viability, and downregulated protein S-nitrosylation, probably also the nitrosylated active form of the cyclooxygenase (COX)-2 protein. These results concur with all the previously reported therapeutic properties of Nitrodi spring water, and thus rein-force the concept that this natural resource is an important complementary therapy to traditional medicine.
Collapse
Affiliation(s)
- Antonietta Aversano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, I-80131 Naples, Italy
| | - Francesca Wanda Rossi
- Department of Translational Medical Sciences, University of Naples Federico II, I-80131 Naples, Italy
| | - Francesca Cammarota
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, I-80131 Naples, Italy
| | - Amato De Paulis
- Department of Translational Medical Sciences, University of Naples Federico II, I-80131 Naples, Italy
| | - Paola Izzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, I-80131 Naples, Italy
| | - Marina De Rosa
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, I-80131 Naples, Italy
| |
Collapse
|
27
|
Blood Oxidative Stress Modulates Alveolar Bone Loss in Chronically Stressed Rats. Int J Mol Sci 2020; 21:ijms21103728. [PMID: 32466304 PMCID: PMC7279478 DOI: 10.3390/ijms21103728] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 01/29/2023] Open
Abstract
We aimed to investigate the effects of chronic stress (CS) on experimental periodontitis (EP) in rats. For this, 28 Wistar rats were divided into four groups: control, ligature-induced experimental periodontitis (EP), chronic stress (CS; by physical restraint model) and CS+EP (association of chronic stress and ligature-induced periodontitis). The experimental period lasted 30 days, including exposure to CS every day and ligature was performed on the 15th experimental day. After 30 days, the animals were submitted to the behavioral test of the elevated plus maze (EPM). Next, rats were euthanized for blood and mandible collection in order to evaluate the oxidative biochemistry (by nitric oxide (NO), reduced-glutathione activity (GSH), and thiobarbituric acid reactive substance levels (TBARS)) and alveolar bone characterization (by morphometric, micro-CT, and immunohistochemistry), respectively. The behavioral parameters evaluated in EPM indicated higher anxiogenic activity in the CS and CS+EP, groups, which is a behavioral reflex of CS. The results showed that CS was able to change the blood oxidative biochemistry in CS and CS+EP groups, decrease GSH activity in the blood, and increase the NO and TBARS concentrations. Thus, CS induces oxidative blood imbalance, which can potentialize or generate morphological, structural, and metabolic damages to the alveolar bone.
Collapse
|
28
|
Laranjinha J, Nunes C, Ledo A, Lourenço C, Rocha B, Barbosa RM. The Peculiar Facets of Nitric Oxide as a Cellular Messenger: From Disease-Associated Signaling to the Regulation of Brain Bioenergetics and Neurovascular Coupling. Neurochem Res 2020; 46:64-76. [PMID: 32193753 DOI: 10.1007/s11064-020-03015-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/07/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022]
Abstract
In this review, we address the regulatory and toxic role of ·NO along several pathways, from the gut to the brain. Initially, we address the role on ·NO in the regulation of mitochondrial respiration with emphasis on the possible contribution to Parkinson's disease via mechanisms that involve its interaction with a major dopamine metabolite, DOPAC. In parallel with initial discoveries of the inhibition of mitochondrial respiration by ·NO, it became clear the potential for toxic ·NO-mediated mechanisms involving the production of more reactive species and the post-translational modification of mitochondrial proteins. Accordingly, we have proposed a novel mechanism potentially leading to dopaminergic cell death, providing evidence that NO synergistically interact with DOPAC in promoting cell death via mechanisms that involve GSH depletion. The modulatory role of NO will be then briefly discussed as a master regulator on brain energy metabolism. The energy metabolism in the brain is central to the understanding of brain function and disease. The core role of ·NO in the regulation of brain metabolism and vascular responses is further substantiated by discussing its role as a mediator of neurovascular coupling, the increase in local microvessels blood flow in response to spatially restricted increase of neuronal activity. The many facets of NO as intracellular and intercellular messenger, conveying information associated with its spatial and temporal concentration dynamics, involve not only the discussion of its reactions and potential targets on a defined biological environment but also the regulation of its synthesis by the family of nitric oxide synthases. More recently, a novel pathway, out of control of NOS, has been the subject of a great deal of controversy, the nitrate:nitrite:NO pathway, adding new perspectives to ·NO biology. Thus, finally, this novel pathway will be addressed in connection with nitrate consumption in the diet and the beneficial effects of protein nitration by reactive nitrogen species.
Collapse
Affiliation(s)
- João Laranjinha
- Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548, Coimbra, Portugal. .,Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal.
| | - Carla Nunes
- Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| | - Ana Ledo
- Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| | - Cátia Lourenço
- Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| | - Bárbara Rocha
- Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| | - Rui M Barbosa
- Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| |
Collapse
|