1
|
Lokhandwala J, Matlack JK, Smalley TB, Miner RE, Tran TH, Binning JM. Structural basis for FN3K-mediated protein deglycation. Structure 2024; 32:1711-1724.e5. [PMID: 39173621 PMCID: PMC11455621 DOI: 10.1016/j.str.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/05/2024] [Accepted: 07/28/2024] [Indexed: 08/24/2024]
Abstract
Protein glycation is a universal, non-enzymatic modification that occurs when a sugar covalently attaches to a primary amine. These spontaneous modifications may have deleterious or regulatory effects on protein function, and their removal is mediated by the conserved metabolic kinase fructosamine-3-kinase (FN3K). Despite its crucial role in protein repair, we currently have a poor understanding of how FN3K engages or phosphorylates its substrates. By integrating structural biology and biochemistry, we elucidated the catalytic mechanism for FN3K-mediated protein deglycation. Our work identifies key amino acids required for binding and phosphorylating glycated substrates and reveals the molecular basis of an evolutionarily conserved protein repair pathway. Additional structural-functional studies revealed unique structural features of human FN3K as well as differences in the dimerization behavior and regulation of FN3K family members. Our findings improve our understanding of the structure of FN3K and its catalytic mechanism, which opens new avenues for therapeutically targeting FN3K.
Collapse
Affiliation(s)
- Jameela Lokhandwala
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Jenet K Matlack
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Tracess B Smalley
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Robert E Miner
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; Cancer Chemical Biology PhD Program, University of South Florida, Tampa, FL 33612, USA
| | - Timothy H Tran
- Chemical Biology Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Jennifer M Binning
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
2
|
Bejaoui S, Chetoui I, Ghribi F, Belhassen D, Abdallah BB, Fayala CB, Boubaker S, Mili S, Soudani N. Exposure to different cobalt chloride levels produces oxidative stress and lipidomic changes and affects the liver structure of Cyprinus carpio juveniles. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:51658-51672. [PMID: 39117974 DOI: 10.1007/s11356-024-34578-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 07/26/2024] [Indexed: 08/10/2024]
Abstract
The present investigation was undertaken to evaluate the toxic effects of CoCl2-induced hepatotoxicity and fatty acid changes in juvenile Cyprinus carpio. Fish were divided into six experimental groups in duplicate. The first group served as controls. The second group received the lowest exposure dose at 2.5 µg/L. In the third group, fish were exposed to 25 µg/L of CoCl2. The fourth group was exposed to 50 µg/L of CoCl2. The last two groups were exposed to the highest doses, 100 and 500 µg/L of CoCl2. Total antioxidant activities were estimated using a colorimetric method. Liver fatty acid compositions were analyzed by high-performance gas chromatography (GC). Hepatopathy was identified through microscopic analysis. Exposure of C. carpio to CoCl2 resulted in hepatotoxicity, indicated by increased levels of malondialdehyde (MDA), hydrogen peroxide (H2O2), protein carbonyls (PCO), and alterations in the ferric reducing antioxidant power system (FRAP). Superoxide dismutase (SOD), glutathione-S-transferase (GST), glutathione peroxidase (GPx), reduced glutathione (GSH), metallothioneins (MTs), and low thiol levels (L-SH) significantly increased, particularly under exposure to the highest CoCl2 doses (100 and 500 µg/L). Acetylcholinesterase activity decreased significantly in C. carpio exposed to graded CoCl2 doses. Additionally, there was a decrease in polyunsaturated fatty acids (PUFA), primarily n-3 PUFA, docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA), while an increase in monounsaturated (MUFA) and saturated fatty acids (SFA), including palmitic (C16:0), stearic (C18:0), palmitoleic (C16:1), and oleic (C18:1) acids, was observed. Histopathological examination of the liver confirmed hepatopathy revealing characteristic tissue changes such as leucocyte infiltration, hepatic cell membrane degradation, vacuolization, and lipid inclusions. The study provided ethnophysiology insights into the responses of C. carpio to CoCl2-induced oxidative stress and lipidomic alteration, underscoring its potential as a bioindicator for assessing environmental impacts and metal contamination.
Collapse
Affiliation(s)
- Safa Bejaoui
- Laboratory of Ecology, Biology and Physiology of Aquatic Organisms, Department of Biology, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia.
| | - Imene Chetoui
- Laboratory of Ecology, Biology and Physiology of Aquatic Organisms, Department of Biology, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
| | - Feriel Ghribi
- Laboratory of Ecology, Biology and Physiology of Aquatic Organisms, Department of Biology, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
- High Institute of Aquaculture and Fishing of Bizerte, BP15, 7080, Menzel Jemil, Tunisia
| | - Dalya Belhassen
- Laboratory of Ecology, Biology and Physiology of Aquatic Organisms, Department of Biology, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
| | - Boutheina Ben Abdallah
- Laboratory of Ecology, Biology and Physiology of Aquatic Organisms, Department of Biology, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
| | - Chaima Ben Fayala
- Unit of Pathological and Experimental Human Anatomy, Institute of Pasteur of Tunis, 1002, Tunis-Belvedere, Tunisia
| | - Samir Boubaker
- Unit of Pathological and Experimental Human Anatomy, Institute of Pasteur of Tunis, 1002, Tunis-Belvedere, Tunisia
| | - Sami Mili
- High Institute of Aquaculture and Fishing of Bizerte, BP15, 7080, Menzel Jemil, Tunisia
| | - Nejla Soudani
- Laboratory of Ecology, Biology and Physiology of Aquatic Organisms, Department of Biology, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
| |
Collapse
|
3
|
Tarbali S, Dadkhah M, Saadati H. Lipophilic fluorescent products as a potential biomarker of oxidative stress: A link between central (brain) and peripheral (blood). JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2024; 29:38. [PMID: 39239084 PMCID: PMC11376719 DOI: 10.4103/jrms.jrms_671_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/06/2024] [Accepted: 04/22/2024] [Indexed: 09/07/2024]
Abstract
Oxidative stress plays a key role in brain damage because of the sensitivity of brain tissue to oxidative damage. Biomarkers with easy measurement can be a candidate for reflecting the oxidative stress issue in humans. For this reason, we need to focus on specific metabolic products of the brain. End products of free radical reactions such as malondialdehydes form fluorescent products known as lipophilic fluorescent products (LFPs). The distinctive feature of LFPs is their autofluorescent properties. LFPs are detectable in the brain and cerebrospinal fluid. Furthermore, because of the diffusion into the bloodstream, these lipophilic molecules can be detected in the blood. Accumulations of these compounds produce more reactive oxygen species and increase the sensitivity of cells to oxidative damage. Hence, LFPs can be considered a danger signal for neurons and can be introduced as a strong index of oxidative damage both in the central and in the peripheral.
Collapse
Affiliation(s)
- Sepideh Tarbali
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hakimeh Saadati
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Department of Physiology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
4
|
Sultana R, Butterfield DA. Protein Oxidation in Aging and Alzheimer's Disease Brain. Antioxidants (Basel) 2024; 13:574. [PMID: 38790679 PMCID: PMC11117785 DOI: 10.3390/antiox13050574] [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: 04/09/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Proteins are essential molecules that play crucial roles in maintaining cellular homeostasis and carrying out biological functions such as catalyzing biochemical reactions, structural proteins, immune response, etc. However, proteins also are highly susceptible to damage by reactive oxygen species (ROS) and reactive nitrogen species (RNS). In this review, we summarize the role of protein oxidation in normal aging and Alzheimer's disease (AD). The major emphasis of this review article is on the carbonylation and nitration of proteins in AD and mild cognitive impairment (MCI). The oxidatively modified proteins showed a strong correlation with the reported changes in brain structure, carbohydrate metabolism, synaptic transmission, cellular energetics, etc., of both MCI and AD brains compared to the controls. Some proteins were found to be common targets of oxidation and were observed during the early stages of AD, suggesting that those changes might be critical in the onset of symptoms and/or formation of the pathological hallmarks of AD. Further studies are required to fully elucidate the role of protein oxidation and nitration in the progression and pathogenesis of AD.
Collapse
Affiliation(s)
- Rukhsana Sultana
- Department of Neuroscience, School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Rd., Richardson, TX 75080, USA;
| | - D. Allan Butterfield
- Department of Chemistry, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
5
|
Lin LW, Durbin-Johnson BP, Rocke DM, Salemi M, Phinney BS, Rice RH. Environmental pro-oxidants induce altered envelope protein profiles in human keratinocytes. Toxicol Sci 2023; 197:16-26. [PMID: 37788135 PMCID: PMC10734632 DOI: 10.1093/toxsci/kfad103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023] Open
Abstract
Cornified envelopes (CEs) of human epidermis ordinarily consist of transglutaminase-mediated cross-linked proteins and are essential for skin barrier function. However, in addition to enzyme-mediated isopeptide bonding, protein cross-linking could also arise from oxidative damage. Our group recently demonstrated abnormal incorporation of cellular proteins into CEs by pro-oxidants in woodsmoke. In this study, we focused on 2,3-dimethoxy-1,4-naphthoquinone (DMNQ), mesquite liquid smoke (MLS), and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), to further understand the mechanisms through which environmental pro-oxidants induce CE formation and alter the CE proteome. CEs induced by the ionophore X537A were used for comparison. Similar to X537A, DMNQ- and MLS-induced CE formation was associated with membrane permeabilization. However, since DMNQ is non-adduct forming, its CEs were similar in protein profile to those from X537A. By contrast, MLS, rich in reactive carbonyls that can form protein adducts, caused a dramatic change in the CE proteome. TCDD-CEs were found to contain many CE precursors, such as small proline-rich proteins and late cornified envelope proteins, encoded by the epidermal differentiation complex. Since expression of these proteins is mediated by the aryl hydrocarbon receptor (AhR), and its well-known downstream protein, CYP1A1, was exclusively present in the TCDD group, we suggest that TCDD alters the CE proteome through persistent AhR activation. This study demonstrates the potential of environmental pro-oxidants to alter the epidermal CE proteome and indicates that the cellular redox state has an important role in CE formation.
Collapse
Affiliation(s)
- Lo-Wei Lin
- Department of Environmental Toxicology, University of California, Davis, California 95616, USA
| | - Blythe P Durbin-Johnson
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, California 95616, USA
| | - David M Rocke
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, California 95616, USA
| | - Michelle Salemi
- Proteomics Core Facility, University of California, Davis, California 95616, USA
| | - Brett S Phinney
- Proteomics Core Facility, University of California, Davis, California 95616, USA
| | - Robert H Rice
- Department of Environmental Toxicology, University of California, Davis, California 95616, USA
| |
Collapse
|
6
|
Han J. Copper trafficking systems in cells: insights into coordination chemistry and toxicity. Dalton Trans 2023; 52:15277-15296. [PMID: 37702384 DOI: 10.1039/d3dt02166a] [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: 09/14/2023]
Abstract
Transition metal ions, such as copper, are indispensable components in the biological system. Copper ions which primarily exist in two major oxidation states Cu(I) and Cu(II) play crucial roles in various cellular processes including antioxidant defense, biosynthesis of neurotransmitters, and energy metabolism, owing to their inherent redox activity. The disturbance in copper homeostasis can contribute to the development of copper metabolism disorders, cancer, and neurodegenerative diseases, highlighting the significance of understanding the copper trafficking system in cellular environments. This review aims to offer a comprehensive overview of copper homeostatic machinery, with an emphasis on the coordination chemistry of copper transporters and trafficking proteins. While copper chaperones and the corresponding metalloenzymes are thoroughly discussed, we also explore the potential existence of low-molecular-mass metal complexes within cellular systems. Furthermore, we summarize the toxicity mechanisms originating from copper deficiency or accumulation, which include the dysregulation of oxidative stress, signaling pathways, signal transduction, and amyloidosis. This perspective review delves into the current knowledge regarding the intricate aspects of the copper trafficking system, providing valuable insights into potential treatment strategies from the standpoint of bioinorganic chemistry.
Collapse
Affiliation(s)
- Jiyeon Han
- Department of Applied Chemistry, University of Seoul, Seoul 02504, Republic of Korea.
| |
Collapse
|
7
|
Cazzaro S, Woo JAA, Wang X, Liu T, Rego S, Kee TR, Koh Y, Vázquez-Rosa E, Pieper AA, Kang DE. Slingshot homolog-1-mediated Nrf2 sequestration tips the balance from neuroprotection to neurodegeneration in Alzheimer's disease. Proc Natl Acad Sci U S A 2023; 120:e2217128120. [PMID: 37463212 PMCID: PMC10374160 DOI: 10.1073/pnas.2217128120] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 06/16/2023] [Indexed: 07/20/2023] Open
Abstract
Oxidative damage in the brain is one of the earliest drivers of pathology in Alzheimer's disease (AD) and related dementias, both preceding and exacerbating clinical symptoms. In response to oxidative stress, nuclear factor erythroid 2-related factor 2 (Nrf2) is normally activated to protect the brain from oxidative damage. However, Nrf2-mediated defense against oxidative stress declines in AD, rendering the brain increasingly vulnerable to oxidative damage. Although this phenomenon has long been recognized, its mechanistic basis has been a mystery. Here, we demonstrate through in vitro and in vivo models, as well as human AD brain tissue, that Slingshot homolog-1 (SSH1) drives this effect by acting as a counterweight to neuroprotective Nrf2 in response to oxidative stress and disease. Specifically, oxidative stress-activated SSH1 suppresses nuclear Nrf2 signaling by sequestering Nrf2 complexes on actin filaments and augmenting Kelch-like ECH-associated protein 1 (Keap1)-Nrf2 interaction, independently of SSH1 phosphatase activity. We also show that Ssh1 elimination in AD models increases Nrf2 activation, which mitigates tau and amyloid-β accumulation and protects against oxidative injury, neuroinflammation, and neurodegeneration. Furthermore, loss of Ssh1 preserves normal synaptic function and transcriptomic patterns in tauP301S mice. Importantly, we also show that human AD brains exhibit highly elevated interactions of Nrf2 with both SSH1 and Keap1. Thus, we demonstrate here a unique mode of Nrf2 blockade that occurs through SSH1, which drives oxidative damage and ensuing pathogenesis in AD. Strategies to inhibit SSH1-mediated Nrf2 suppression while preserving normal SSH1 catalytic function may provide new neuroprotective therapies for AD and related dementias.
Collapse
Affiliation(s)
- Sara Cazzaro
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Molecular Medicine, University of South Florida Health College of Medicine, Tampa, FL33620
| | - Jung-A A. Woo
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Xinming Wang
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Tian Liu
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Shanon Rego
- Department of Molecular Medicine, University of South Florida Health College of Medicine, Tampa, FL33620
| | - Teresa R. Kee
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Molecular Medicine, University of South Florida Health College of Medicine, Tampa, FL33620
| | - Yeojung Koh
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Institute for Transformative Molecular Medicine, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Edwin Vázquez-Rosa
- Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Institute for Transformative Molecular Medicine, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Andrew A. Pieper
- Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Institute for Transformative Molecular Medicine, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Neuroscience, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Cleveland Veteran Affairs Medical Center, Cleveland, OH44106
- Brain Health Medicines, Center Harrington Discovery Institute, Cleveland, OH44106
| | - David E. Kang
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Louis Stokes Cleveland Veteran Affairs Medical Center, Cleveland, OH44106
| |
Collapse
|
8
|
Dakterzada F, Jové M, Cantero JL, Pamplona R, Piñoll-Ripoll G. Plasma and cerebrospinal fluid nonenzymatic protein damage is sustained in Alzheimer's disease. Redox Biol 2023; 64:102772. [PMID: 37339560 DOI: 10.1016/j.redox.2023.102772] [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: 05/03/2023] [Revised: 06/01/2023] [Accepted: 06/03/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND Oxidative stress is considered to play an important role in the pathogenesis of Alzheimer's disease (AD). It has been observed that oxidative damage to specific protein targets affecting particular functional networks is one of the mechanisms by which oxidative stress contributes to neuronal failure and consequently loss of cognition and AD progression. Studies are lacking in which oxidative damage is measured at both systemic and central fluid levels and in the same cohort of patients. We aimed to determine the levels of both plasma and cerebrospinal fluid (CSF) nonenzymatic protein damage in patients in the continuum of AD and to evaluate the relation of this damage with clinical progression from mild cognitive impairment (MCI) to AD. METHODS Different markers of nonenzymatic post-translational protein modification, mostly from oxidative processes, were detected and quantified in plasma and CSF by isotope dilution gas chromatography‒mass spectrometry using selected ion monitoring (SIM-GC/MS) for 289 subjects: 103 AD, 92 MCI, and 94 control subjects. Characteristics of the study population such as age, sex, Mini-mental state examination, CSF AD biomarkers, and APOE ϵ4, were also considered. RESULTS Forty-seven (52.8%) MCI patients progressed to AD during follow-up (58 ± 12.5 months). After controlling for age, sex, and APOE ϵ4 allele, plasma and CSF concentrations of protein damage markers were not associated with either diagnosis of AD or MCI. The CSF levels of nonenzymatic protein damage markers were associated with none of the CSF AD biomarkers. In addition, neither in CSF nor in plasma were the levels of protein damage associated with the MCI to AD progression. CONCLUSION The lack of association between both CSF and plasma concentrations of nonenzymatic protein damage markers and AD diagnosis and progression suggests that oxidative damage in AD is a pathogenic mechanism specifically expressed at the cell-tissue level, not in extracellular fluids.
Collapse
Affiliation(s)
- Farida Dakterzada
- Cognitive Disorders Unit, Clinical Neuroscience Research, Hospital Universitari Santa Maria, IRBLleida, Lleida, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida, IRBLleida, Lleida, Spain
| | - José Luís Cantero
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Seville, Spain; CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida, IRBLleida, Lleida, Spain
| | - Gerard Piñoll-Ripoll
- Cognitive Disorders Unit, Clinical Neuroscience Research, Hospital Universitari Santa Maria, IRBLleida, Lleida, Spain.
| |
Collapse
|
9
|
Mowaad NA, El-Shamarka MEA, Khadrawy YA. The Behavioral and Neurochemical Changes Induced by Boldenone and/or Tramadol in Adult Male Rats. Neurochem Res 2022; 48:1320-1333. [PMID: 36449200 PMCID: PMC10066173 DOI: 10.1007/s11064-022-03827-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/17/2022] [Accepted: 11/14/2022] [Indexed: 12/02/2022]
Abstract
AbstractBoldenone and tramadol are abused among large sectors of adolescents. Therefore, the behavioral changes concerned with memory and cognitive functions and neurochemical variations were investigated in the cortex of rats treated with boldenone and/or tramadol. Rats were divided into control and rats treated with boldenone, tramadol, or both drugs. At the end of the treatment period, the memory and cognitive functions were evaluated by the Y-maze test (YMT) and elevated plus maze test (EPMT) and the motor activity was determined by the open field test (OFT). The cortex was dissected to carry out the neurochemical analyses. Rats treated with boldenone and/or tramadol showed impaired memory and cognitive functions and reduced motor activity. A significant increase in lipid peroxidation (MDA), nitric oxide (NO), and a significant decrease in reduced glutathione (GSH) were observed in the cortex of rats treated with boldenone and/or tramadol. The levels of acetylcholinesterase (AChE) and monoamine oxidase (MAO) decreased significantly. Western blot data showed a significant decrease in Bcl2 and a significant increase in caspase-3 and inducible nitric oxide synthase (iNOS) in rats treated with boldenone and/or tramadol. These changes were associated with neuronal death as indicated from the histopathological examination.The present findings indicate that boldenone and/or tramadol induced impairment in memory and cognitive functions. These changes could be mediated by the increase in oxidative stress, neuroinflammation, reduced AChE level, and reduced number of survived neurons in the cortex as indicated from the decreased Bcl2 level and the histological examination.
Collapse
Affiliation(s)
- Noha A Mowaad
- Department of Narcotics, Ergogenic Aids and Poisons,Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Marwa E A El-Shamarka
- Department of Narcotics, Ergogenic Aids and Poisons,Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Yasser A Khadrawy
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Centre, El-Behouth St, Giza, Egypt.
| |
Collapse
|
10
|
Viedma-Poyatos Á, González-Jiménez P, Pajares MA, Pérez-Sala D. Alexander disease GFAP R239C mutant shows increased susceptibility to lipoxidation and elicits mitochondrial dysfunction and oxidative stress. Redox Biol 2022; 55:102415. [PMID: 35933901 PMCID: PMC9364016 DOI: 10.1016/j.redox.2022.102415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 01/04/2023] Open
Abstract
Alexander disease is a fatal neurological disorder caused by mutations in the intermediate filament protein Glial Fibrillary Acidic Protein (GFAP), which is key for astrocyte homeostasis. These mutations cause GFAP aggregation, astrocyte dysfunction and neurodegeneration. Remarkably, most of the known GFAP mutations imply a change by more nucleophilic amino acids, mainly cysteine or histidine, which are more susceptible to oxidation and lipoxidation. Therefore, we hypothesized that a higher susceptibility of Alexander disease GFAP mutants to oxidative or electrophilic damage, which frequently occurs during neurodegeneration, could contribute to disease pathogenesis. To address this point, we have expressed GFP-GFAP wild type or the harmful Alexander disease GFP-GFAP R239C mutant in astrocytic cells. Interestingly, GFAP R239C appears more oxidized than the wild type under control conditions, as indicated both by its lower cysteine residue accessibility and increased presence of disulfide-bonded oligomers. Moreover, GFP-GFAP R239C undergoes lipoxidation to a higher extent than GFAP wild type upon treatment with the electrophilic mediator 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2). Importantly, GFAP R239C filament organization is altered in untreated cells and is earlier and more severely disrupted than GFAP wild type upon exposure to oxidants (diamide, H2O2) or electrophiles (4-hydroxynonenal, 15d-PGJ2), which exacerbate GFAP R239C aggregation. Furthermore, H2O2 causes reversible alterations in GFAP wild type, but irreversible damage in GFAP R239C expressing cells. Finally, we show that GFAP R239C expression induces a more oxidized cellular status, with decreased free thiol content and increased mitochondrial superoxide generation. In addition, mitochondria show decreased mass, increased colocalization with GFAP and altered morphology. Notably, a GFP-GFAP R239H mutant recapitulates R239C-elicited alterations whereas an R239G mutant induces a milder phenotype. Together, our results outline a deleterious cycle involving altered GFAP R239C organization, mitochondrial dysfunction, oxidative stress, and further GFAP R239C protein damage and network disruption, which could contribute to astrocyte derangement in Alexander disease.
Collapse
Affiliation(s)
- Álvaro Viedma-Poyatos
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, C.S.I.C., 28040, Madrid, Spain
| | - Patricia González-Jiménez
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, C.S.I.C., 28040, Madrid, Spain
| | - María A Pajares
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, C.S.I.C., 28040, Madrid, Spain
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, C.S.I.C., 28040, Madrid, Spain.
| |
Collapse
|
11
|
Ding J, Chen Y, Zhao YJ, Chen F, Dong L, Zhang HL, Hu WR, Li SF, Zhou RP, Hu W. Acid-sensitive ion channel 1a mediates osteoarthritis chondrocyte senescence by promoting Lamin B1 degradation. Biochem Pharmacol 2022; 202:115107. [PMID: 35643339 DOI: 10.1016/j.bcp.2022.115107] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is a common and debilitating chronic joint disease, which is characterized by degeneration of articular cartilage and the aging of chondrocytes. Acid-sensitive ion channel 1a (ASIC1a) is a proton-activated cationic channel abundant in chondrocytes, which senses and regulates joint cavity pH. Our previous study demonstrated that ASIC1a was involved in acid-induced rat articular chondrocyte senescence, but the mechanistic basis remained unclear. In this study, we explored the mechanism of ASIC1a in chondrocyte senescence and OA. The results showed that senescence-related-β-galactosidase, senescence-related markers (p53 and p21) and the autophagy-related protein Beclin-1 were found to be increased, but Lamin B1 was found to be reduced with acid (pH 6.0) treatment. These effects were inhibited by ASIC1a-specific blocker psalmotoxin-1 or ASIC1a-short hairpin RNA respectively in chondrocytes. Moreover, Silencing of Lamin B1 enhanced ASIC1a-mediated chondrocyte senescence, this effect was reversed by overexpression of Lamin B1, indicating that Lamin B1 was involved in ASIC1a-mediated chondrocyte senescence. Further, blockade of ASIC1a inhibits acid-induced autophagosomes and Beclin-1 protein expression, suggesting that ASIC1a is involved in acid-induced chondrocyte autophagy. Blocking autophagy with chloroquine inhibited Beclin-1 and increased Lamin B1 in acid-induced chondrocyte senescence. We further demonstrated that ASIC1a-mediated reduction of Lamin B1 expression was caused by autophagy pathway-dependent protein degradation. Finally, blocking ASIC1a protected cartilage tissue, restored Lamin B1 levels and inhibited chondrocyte senescence in a rat OA model. In summary, these findings suggest that ASIC1a may promote Lamin B1 degradation to mediate osteoarthritis chondrocyte senescence through the autophagy pathway.
Collapse
Affiliation(s)
- Jie Ding
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Yong Chen
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Ying-Jie Zhao
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Fan Chen
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Lei Dong
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Hai-Lin Zhang
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Wei-Rong Hu
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Shu-Fang Li
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Ren-Peng Zhou
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China.
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China.
| |
Collapse
|
12
|
Lee J, Kim HJ. Normal Aging Induces Changes in the Brain and Neurodegeneration Progress: Review of the Structural, Biochemical, Metabolic, Cellular, and Molecular Changes. Front Aging Neurosci 2022; 14:931536. [PMID: 35847660 PMCID: PMC9281621 DOI: 10.3389/fnagi.2022.931536] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/13/2022] [Indexed: 11/30/2022] Open
Abstract
Aging is accompanied by many changes in brain and contributes to progressive cognitive decline. In contrast to pathological changes in brain, normal aging brain changes have relatively mild but important changes in structural, biochemical and molecular level. Representatively, aging associated brain changes include atrophy of tissues, alteration in neurotransmitters and damage accumulation in cellular environment. These effects have causative link with age associated changes which ultimately results in cognitive decline. Although several evidences were found in normal aging changes of brain, it is not clearly integrated. Figuring out aging related changes in brain is important as aging is the process that everyone goes through, and comprehensive understanding may help to progress further studies. This review clarifies normal aging brain changes in an asymptotic and comprehensive manner, from a gross level to a microscopic and molecular level, and discusses potential approaches to seek the changes with cognitive decline.
Collapse
Affiliation(s)
- Jiseon Lee
- Department of Neurology, Hanyang University Hospital, Seoul, South Korea
| | - Hee-Jin Kim
- Department of Neurology, Hanyang University Hospital, Seoul, South Korea
| |
Collapse
|
13
|
Tiwari AK, Adhikari A, Mishra LC, Srivastava A. Current Status of Our Understanding for Brain Integrated Functions and its Energetics. Neurochem Res 2022; 47:2499-2512. [PMID: 35689788 DOI: 10.1007/s11064-022-03633-w] [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: 11/30/2021] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 10/18/2022]
Abstract
Human/animal brain is a unique organ with substantially high metabolism but it contains no energy reserve that is the reason it requires continuous supply of O2 and energy fluxes through CBF. The main source of energy remains glucose as the other biomolecules do not able to cross the blood-brain barrier. The speed of glucose metabolism is heterogeneous throughout the brain. One of the major flux consumption is Neuron-astrocyte cycling of glutamate and glutamine in glutamatergic neurons (approximately 80% of glucose metabolism in brain). The quantification of cellular glucose and other related substrate in resting, activated state can be analyzed through [18 F]FDG -positron-emission tomography (studying CMRglc) and [13 C/31P -MRS: for neuroenergetics & neurotransmitter cycling &31P-MRS: for energy induction & redox state). Merging basic in vitro studies with these techniques will help to develop new treatment paradigms for human brain diseased conditions.
Collapse
Affiliation(s)
- Anjani Kumar Tiwari
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University (A Central University), 226025, Lucknow, Uttar Pradesh, India.
| | - Anupriya Adhikari
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University (A Central University), 226025, Lucknow, Uttar Pradesh, India
| | - Lokesh Chandra Mishra
- Department of Zoology, Hansraj College, University of Delhi, North Campus, 110007, Delhi, India
| | | |
Collapse
|
14
|
González-Ruiz V, Cores Á, Caja MM, Sridharan V, Villacampa M, Martín MA, Olives AI, Menéndez JC. Fluorescence Sensors Based on Hydroxycarbazole for the Determination of Neurodegeneration-Related Halide Anions. BIOSENSORS 2022; 12:175. [PMID: 35323445 PMCID: PMC8946780 DOI: 10.3390/bios12030175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 11/21/2022]
Abstract
The environmental presence of anions of natural origin or anthropogenic origin is gradually increasing. As a tool to tackle this problem, carbazole derivatives are an attractive gateway to the development of luminescent chemosensors. Considering the different mechanisms proposed for anion recognition, the fluorescence properties and anion-binding response of several newly synthesised carbazole derivatives were studied. Potential anion sensors were designed so that they combined the native fluorescence of carbazole with the presence of hydrogen bonding donor groups in critical positions for anion recognition. These compounds were synthesised by a feasible and non-expensive procedure using palladium-promoted cyclodehydrogenation of suitable diarylamine under microwave irradiation. In comparison to the other carbazole derivatives studied, 1-hydroxycarbazole proved to be useful as a fluorescent sensor for anions, as it was able to sensitively recognise fluoride and chloride anions by establishing hydrogen bond interactions through the hydrogen atoms on the pyrrolic nitrogen and the hydroxy group. Solvent effects and excited-state proton transfer (ESPT) of the carbazole derivatives are described to discard the role of the anions as Brönsted bases on the observed fluorescence behaviour of the sensors. The anion-sensor interaction was confirmed by 1H-NMR. Molecular modelling was employed to propose a mode of recognition of the sensor in terms of complex stability and interatomic distances. 1-hydroxycarbazole was employed for the quantitation of fluoride and chloride anions in commercially available medicinal spring water and mouthwash samples.
Collapse
Affiliation(s)
- Víctor González-Ruiz
- Unidad de Química Analítica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain; (V.G.-R.); (M.M.C.); (M.A.M.)
- School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
- Swiss Centre for Applied Human Toxicology (SCATH), 4055 Basel, Switzerland
| | - Ángel Cores
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Á.C.); (V.S.); (M.V.)
| | - M. Mar Caja
- Unidad de Química Analítica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain; (V.G.-R.); (M.M.C.); (M.A.M.)
| | - Vellaisamy Sridharan
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Á.C.); (V.S.); (M.V.)
- Department of Chemistry and Chemical Sciences, Central University of Jammu, Rahya-Suchani (Bagla), District-Samba, Jammu 181143, J&K, India
| | - Mercedes Villacampa
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Á.C.); (V.S.); (M.V.)
| | - M. Antonia Martín
- Unidad de Química Analítica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain; (V.G.-R.); (M.M.C.); (M.A.M.)
| | - Ana I. Olives
- Unidad de Química Analítica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain; (V.G.-R.); (M.M.C.); (M.A.M.)
| | - J. Carlos Menéndez
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Á.C.); (V.S.); (M.V.)
| |
Collapse
|
15
|
Wang H, Wang X, Li P, Dong M, Yao SQ, Tang B. Fluorescent probes for visualizing ROS-associated proteins in disease. Chem Sci 2021; 12:11620-11646. [PMID: 34659698 PMCID: PMC8442704 DOI: 10.1039/d1sc02165f] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/06/2021] [Indexed: 12/14/2022] Open
Abstract
Abnormal expression of proteins, including catalytic and expression dysfunction, is directly related to the development of various diseases in living organisms. Reactive oxygen species (ROS) could regulate protein expression by redox modification or cellular signal pathway and thus influence the development of disease. Determining the expression level and activity of these ROS-associated proteins is of considerable importance in early-stage disease diagnosis and the identification of new drug targets. Fluorescence imaging technology has emerged as a powerful tool for specific in situ imaging of target proteins by virtue of its non-invasiveness, high sensitivity and good spatiotemporal resolution. In this review, we summarize advances made in the past decade for the design of fluorescent probes that have contributed to tracking ROS-associated proteins in disease. We envision that this review will attract significant attention from a wide range of researchers in their utilization of fluorescent probes for in situ investigation of pathological processes synergistically regulated by both ROS and proteins.
Collapse
Affiliation(s)
- Hui Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University Jinan 250014 P. R. China
| | - Xin Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University Jinan 250014 P. R. China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University Jinan 250014 P. R. China
| | - Mingyan Dong
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University Jinan 250014 P. R. China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore Singapore 117543 Singapore
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University Jinan 250014 P. R. China
| |
Collapse
|
16
|
Mohseni M, Sahebkar A, Askari G, Johnston TP, Alikiaii B, Bagherniya M. The clinical use of curcumin on neurological disorders: An updated systematic review of clinical trials. Phytother Res 2021; 35:6862-6882. [PMID: 34528307 DOI: 10.1002/ptr.7273] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 08/05/2021] [Accepted: 08/25/2021] [Indexed: 12/19/2022]
Abstract
Neuroprotective effects of curcumin have been shown in previous studies. This updated systematic review of clinical trials aimed to investigate the effect of curcumin on neurological disorders. Databases including PubMed, Scopus, Web of Science, and Google Scholar were systematically searched to identify clinical trials investigating the effects of curcumin/turmeric supplements alone, or in combination with other ingredients, on neurological diseases. Nineteen studies comprising 1,130 patients met the inclusion criteria. Generally, intervention and study outcomes were heterogeneous. In most of the studies, curcumin had a favorable effect on oxidative stress and inflammation. However, with the exception of AD, curcumin supplementation either alone, or in combination with other ingredients, had beneficial effects on clinical outcomes for the other aforementioned neurodegenerative diseases. For example, the frequency, severity, and duration of migraine attacks, scores on the revised ALS functional rating scale, and the occurrence of motor complications in PD were all significantly improved with curcumin supplementation either alone or in combination with other ingredients. However, in three studies, several adverse side effects (mostly gastrointestinal in nature) were reported. Curcumin supplementation may have favorable effects on inflammatory status and clinical outcomes of patients with neurological disease, although the results were not consistent.
Collapse
Affiliation(s)
- Maryam Mohseni
- Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.,Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Australia.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Askari
- Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.,Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.,Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Babak Alikiaii
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Bagherniya
- Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.,Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.,Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
17
|
Dietary nutrients and their control of the redox bioenergetic networks as therapeutics in redox dysfunctions sustained pathologies. Pharmacol Res 2021; 170:105709. [PMID: 34089868 DOI: 10.1016/j.phrs.2021.105709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/12/2021] [Accepted: 05/31/2021] [Indexed: 02/06/2023]
Abstract
Electrons exchange amongst the chemical species in an organism is a pivotal concomitant activity carried out by individual cells for basic cellular processes and continuously contribute towards the maintenance of bioenergetic networks plus physiological attributes like cell growth, phenotypic differences and nutritional adaptations. Humans exchange matter and energy via complex connections of metabolic pathways (redox reactions) amongst cells being a thermodynamically open system. Usually, these reactions are the real lifeline and driving forces of health and disease in the living entity. Many shreds of evidence support the secondary role of reactive species in the cellular process of control apoptosis and proliferation. Disrupted redox mechanisms are seen in malaises, like degenerative and metabolic disorders, cancerous cells. This review targets the importance of redox reactions in the body's normal functioning and the effects of its alterations in cells to obtain a better understanding. Understanding the redox dynamics in a pathological state can provide an opportunity for cure or diagnosis at the earlier stage and serve as an essential biomarker to predict in advance to give personalized therapy. Understanding redox metabolism can also highlight the use of naturally available antioxidant in the form of diet.
Collapse
|
18
|
Duarte-Silva E, Meiry da Rocha Araújo S, Oliveira WH, Lós DB, Bonfanti AP, Peron G, de Lima Thomaz L, Verinaud L, Peixoto CA. Sildenafil Alleviates Murine Experimental Autoimmune Encephalomyelitis by Triggering Autophagy in the Spinal Cord. Front Immunol 2021; 12:671511. [PMID: 34054847 PMCID: PMC8156813 DOI: 10.3389/fimmu.2021.671511] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
Multiple Sclerosis (MS) is a neuroinflammatory and chronic Central Nervous System (CNS) disease that affects millions of people worldwide. The search for more promising drugs for the treatment of MS has led to studies on Sildenafil, a phosphodiesterase type 5 Inhibitor (PDE5I) that has been shown to possess neuroprotective effects in the Experimental Autoimmune Encephalomyelitis (EAE), an animal model of MS. We have previously shown that Sildenafil improves the clinical score of EAE mice via modulation of apoptotic pathways, but other signaling pathways were not previously covered. Therefore, the aim of the present study was to further investigate the effects of Sildenafil treatment on autophagy and nitrosative stress signaling pathways in EAE. 24 female C57BL/6 mice were divided into the following groups: (A) Control - received only water; (B) EAE - EAE untreated mice; (C) SILD - EAE mice treated with 25mg/kg of Sildenafil s.c. The results showed that EAE mice presented a pro-nitrosative profile characterized by high tissue nitrite levels, lowered levels of p-eNOS and high levels of iNOS. Furthermore, decreased levels of LC3, beclin-1 and ATG5, suggests impaired autophagy, and decreased levels of AMPK in the spinal cord were also detected in EAE mice. Surprisingly, treatment with Sildenafil inhibited nitrosative stress and augmented the levels of LC3, beclin-1, ATG5, p-CREB and BDNF and decreased mTOR levels, as well as augmented p-AMPK. In conclusion, we propose that Sildenafil alleviates EAE by activating autophagy via the eNOS-NO-AMPK-mTOR-LC3-beclin1-ATG5 and eNOS-NO-AMPK-mTOR-CREB-BDNF pathways in the spinal cord.
Collapse
Affiliation(s)
- Eduardo Duarte-Silva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Recife, Brazil
- Postgraduate Program in Biosciences and Biotechnology for Health (PPGBBS), Oswaldo Cruz Foundation (FIOCRUZ-PE)/Aggeu Magalhães Institute (IAM), Recife, Brazil
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Recife, Brazil
| | - Shyrlene Meiry da Rocha Araújo
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Recife, Brazil
- Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, Brazil
| | - Wilma Helena Oliveira
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Recife, Brazil
- Postgraduate Program in Biological Sciences/Center of Biosciences, Federal University of Pernambuco (UFPE), Recife, Brazil
| | - Deniele Bezerra Lós
- Postgraduate Program in Biotechnology/Northeast Network in Biotechnology (RENORBIO), Federal University of Pernambuco (UFPE), Recife, Brazil
| | - Amanda Pires Bonfanti
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Gabriela Peron
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Livia de Lima Thomaz
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Liana Verinaud
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Christina Alves Peixoto
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Recife, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
19
|
Jové M, Mota-Martorell N, Torres P, Ayala V, Portero-Otin M, Ferrer I, Pamplona R. The Causal Role of Lipoxidative Damage in Mitochondrial Bioenergetic Dysfunction Linked to Alzheimer's Disease Pathology. Life (Basel) 2021; 11:life11050388. [PMID: 33923074 PMCID: PMC8147054 DOI: 10.3390/life11050388] [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: 03/19/2021] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 01/18/2023] Open
Abstract
Current shreds of evidence point to the entorhinal cortex (EC) as the origin of the Alzheimer’s disease (AD) pathology in the cerebrum. Compared with other cortical areas, the neurons from this brain region possess an inherent selective vulnerability derived from particular oxidative stress conditions that favor increased mitochondrial molecular damage with early bioenergetic involvement. This alteration of energy metabolism is the starting point for subsequent changes in a multitude of cell mechanisms, leading to neuronal dysfunction and, ultimately, cell death. These events are induced by changes that come with age, creating the substrate for the alteration of several neuronal pathways that will evolve toward neurodegeneration and, consequently, the development of AD pathology. In this context, the present review will focus on description of the biological mechanisms that confer vulnerability specifically to neurons of the entorhinal cortex, the changes induced by the aging process in this brain region, and the alterations at the mitochondrial level as the earliest mechanism for the development of AD pathology. Current findings allow us to propose the existence of an altered allostatic mechanism at the entorhinal cortex whose core is made up of mitochondrial oxidative stress, lipid metabolism, and energy production, and which, in a positive loop, evolves to neurodegeneration, laying the basis for the onset and progression of AD pathology.
Collapse
Affiliation(s)
- Mariona Jové
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), 25198 Lleida, Spain; (M.J.); (N.M.-M.); (P.T.); (V.A.); (M.P.-O.)
| | - Natàlia Mota-Martorell
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), 25198 Lleida, Spain; (M.J.); (N.M.-M.); (P.T.); (V.A.); (M.P.-O.)
| | - Pascual Torres
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), 25198 Lleida, Spain; (M.J.); (N.M.-M.); (P.T.); (V.A.); (M.P.-O.)
| | - Victoria Ayala
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), 25198 Lleida, Spain; (M.J.); (N.M.-M.); (P.T.); (V.A.); (M.P.-O.)
| | - Manuel Portero-Otin
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), 25198 Lleida, Spain; (M.J.); (N.M.-M.); (P.T.); (V.A.); (M.P.-O.)
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Bellvitge University Hospital/Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08907 Barcelona, Spain
- Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), ISCIII, 28220 Madrid, Spain
- Correspondence: (I.F.); (R.P.)
| | - Reinald Pamplona
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), Lleida University (UdL), 25198 Lleida, Spain; (M.J.); (N.M.-M.); (P.T.); (V.A.); (M.P.-O.)
- Correspondence: (I.F.); (R.P.)
| |
Collapse
|
20
|
Salivary Biomarkers of Oxidative Stress and Inflammation in Stroke Patients: From Basic Research to Clinical Practice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5545330. [PMID: 33897941 PMCID: PMC8052150 DOI: 10.1155/2021/5545330] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 12/11/2022]
Abstract
Cerebral stroke is a serious worldwide health problem, as can be seen by the global epidemic of the disease. In this disorder, when the blood flow is compromised by ruptures or blocked arteries, sudden death of neurons is observed as a result of a lack of oxygen and nutrients. Numerous severe problems and frequent complications also exist in stroke patients; therefore, there is an urgent need to develop new therapeutic, diagnostic, and prognostic methods for the disease. At present, the diagnosis of stroke is based on a neurological examination, medical history, and neuroimaging, due to the fact that rapid and noninvasive diagnostic tests are unavailable. Nevertheless, oxidative stress and inflammation are considered key factors in stroke pathogenesis. Oxygen free radicals are responsible for oxidation of lipids, proteins, and DNA/RNA, which in turn contributes to oxidative damage of the brain. Toxic products of the oxidation reactions act cytostatically on the cell by damaging cell membranes and leading to neuronal death by apoptosis or necrosis. Thus, it seems that redox/inflammatory biomarkers might be used in the diagnosis of the disease. Nowadays, saliva is of increasing interest in clinical laboratory medicine. Redox biomarkers could be obtained easily, noninvasively, cheaply, and stress-free from saliva. This minireview is aimed at presenting the current knowledge concerning the use of salivary biomarkers of oxidative stress and inflammation in the diagnosis and prognosis of stroke.
Collapse
|
21
|
Avcil S, Uysal P, Yenisey Ç, Abas BI. Elevated Melatonin Levels in Children With Attention Deficit Hyperactivity Disorder: Relationship to Oxidative and Nitrosative Stress. J Atten Disord 2021; 25:693-703. [PMID: 30819002 DOI: 10.1177/1087054719829816] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Objective: The aim of this article is to measure serum antioxidant melatonin, the oxidants of nitric oxide, and malondialdehyde levels to calculate the serum oxidant-antioxidant balance based on the nitric oxide/melatonin and malondialdehyde/melatonin ratios in children with ADHD. Method: The serum melatonin, nitric oxide, malondialdehyde, and the nitric oxide/melatonin and malondialdehyde/melatonin ratios were calculated and compared between the children with ADHD (n = 103) and healthy control participants (n = 73). Results: Serum melatonin and nitric oxide levels were higher, and the nitric oxide/melatonin and malondialdehyde/melatonin ratios were lower in ADHD children than the control group. Melatonin was found to be significantly high, and the malondialdehyde/melatonin ratio was found to be significantly low in children with a positive ADHD family history. Conclusion: The serum oxidant-antioxidant balance was impaired in children with ADHD. Within the ADHD group, higher melatonin levels were determined in the children with a positive family history.
Collapse
Affiliation(s)
| | - Pınar Uysal
- Aydın Adnan Menderes University, Aydın, Turkey
| | | | | |
Collapse
|
22
|
Catalase Inhibitors with Dual Pro‐Oxidant Effect as New Therapeutic Agents in Castration‐Resistant Prostate Cancer. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
23
|
Dos Santos Cardoso F, Dos Santos JCC, Gonzalez-Lima F, Araújo BHS, Lopes-Martins RÁB, Gomes da Silva S. Effects of Chronic Photobiomodulation with Transcranial Near-Infrared Laser on Brain Metabolomics of Young and Aged Rats. Mol Neurobiol 2021; 58:2256-2268. [PMID: 33417219 DOI: 10.1007/s12035-020-02247-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/03/2020] [Indexed: 12/25/2022]
Abstract
Since laser photobiomodulation has been found to enhance brain energy metabolism and cognition, we conducted the first metabolomics study to systematically analyze the metabolites modified by brain photobiomodulation. Aging is often accompanied by cognitive decline and susceptibility to neurodegeneration, including deficits in brain energy metabolism and increased susceptibility of nerve cells to oxidative stress. Changes in oxidative stress and energetic homeostasis increase neuronal vulnerability, as observed in diseases related to brain aging. We evaluated and compared the cortical and hippocampal metabolic pathways of young (4 months old) and aged (20 months old) control rats with those of rats exposed to transcranial near-infrared laser over 58 consecutive days. Statistical analyses of the brain metabolomics data indicated that chronic transcranial photobiomodulation (1) significantly enhances the metabolic pathways of young rats, particularly for excitatory neurotransmission and oxidative metabolism, and (2) restores the altered metabolic pathways of aged rats towards levels found in younger rats, mainly in the cerebral cortex. These novel metabolomics findings may help complement other laser-induced neurocognitive, neuroprotective, anti-inflammatory, and antioxidant effects described in the literature.
Collapse
Affiliation(s)
- Fabrízio Dos Santos Cardoso
- Núcleo de Pesquisas Tecnológicas, Universidade de Mogi das Cruzes, Av. Cândido Xavier de Almeida e Souza, 200, Mogi das Cruzes, SP, CEP 08780-911, Brazil.,Department of Psychology and Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - Júlio César Claudino Dos Santos
- Laboratory of Neuroscience, Department of Neurology and Neurosurgery, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Francisco Gonzalez-Lima
- Department of Psychology and Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - Bruno Henrique Silva Araújo
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| | - Rodrigo Álvaro Brandão Lopes-Martins
- Laboratory of Biophotonics and Experimental Therapeutics, Institute of Research and Development, University of Vale do Paraíba (UNIVAP), São José dos Campos, SP, Brazil
| | - Sérgio Gomes da Silva
- Núcleo de Pesquisas Tecnológicas, Universidade de Mogi das Cruzes, Av. Cândido Xavier de Almeida e Souza, 200, Mogi das Cruzes, SP, CEP 08780-911, Brazil. .,Centro Universitário UNIFAMINAS (UNIFAMINAS), Muriaé, MG, Brazil. .,Hospital do Câncer de Muriaé, Fundação Cristiano Varella (FCV), Muriaé, MG, Brazil.
| |
Collapse
|
24
|
Yang Z, Lin P, Chen B, Zhang X, Xiao W, Wu S, Huang C, Feng D, Zhang W, Zhang J. Autophagy alleviates hypoxia-induced blood-brain barrier injury via regulation of CLDN5 (claudin 5). Autophagy 2020; 17:3048-3067. [PMID: 33280500 DOI: 10.1080/15548627.2020.1851897] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Blood-brain barrier (BBB) disruption is a key event in triggering secondary damage to the central nervous system (CNS) under stroke, and is frequently associated with abnormal macroautophagy/autophagy in brain microvascular endothelial cells (BMECs). However, the underlying mechanism of autophagy in maintaining BBB integrity remains unclear. Here we report that in BMECs of patients suffering stroke, CLDN5 (claudin 5) abnormally aggregates in the cytosol accompanied by autophagy activation. In vivo zebrafish and in vitro cell studies reveal that BBB breakdown is partially caused by CAV1 (caveolin 1)-mediated redistribution of membranous CLDN5 into the cytosol under hypoxia. Meanwhile, autophagy is activated and contributes mainly to the degradation of CAV1 and aggregated CLDN5 in the cytosol of BMECs, therefore alleviating BBB breakdown. Blockage of autophagy by genetic methods or chemicals aggravates cytosolic aggregation of CLDN5, resulting in severer BBB impairment. These data demonstrate that autophagy functions in the protection of BBB integrity by regulating CLDN5 redistribution and provide a potential therapeutic strategy for BBB disorder-related cerebrovascular disease.Abbreviations: BBB: blood-brain barrier; BECN1: beclin 1; BMEC: brain microvascular endothelial cell; CAV1: caveolin 1; CCA: common carotid artery; CLDN5: claudin 5; CNS: central nervous system; CQ: chloroquine; HIF1A: hypoxia inducible factor 1 subunit alpha; MCAO: middle cerebral artery occlusion-reperfusion; OCLN: occludin; ROS: reactive oxygen species; STED: stimulated emission depletion; TEER: trans-endothelial electrical resistance; TEM: transmission electron microscopy; TJ: tight junction; TJP1: tight junction protein 1; UPS: ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Zhenguo Yang
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, China
| | - Panpan Lin
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, China
| | - Bing Chen
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, China
| | - Xiaoqi Zhang
- Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Wei Xiao
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, China
| | - Shuilong Wu
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, China
| | - Chunnian Huang
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, China
| | - Du Feng
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Wenqing Zhang
- Laboratory of Developmental Biology and Regenerative Medicine, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jingjing Zhang
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
25
|
Gámez-Valero A, Guisado-Corcoll A, Herrero-Lorenzo M, Solaguren-Beascoa M, Martí E. Non-Coding RNAs as Sensors of Oxidative Stress in Neurodegenerative Diseases. Antioxidants (Basel) 2020; 9:E1095. [PMID: 33171576 PMCID: PMC7695195 DOI: 10.3390/antiox9111095] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/03/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress (OS) results from an imbalance between the production of reactive oxygen species and the cellular antioxidant capacity. OS plays a central role in neurodegenerative diseases, where the progressive accumulation of reactive oxygen species induces mitochondrial dysfunction, protein aggregation and inflammation. Regulatory non-protein-coding RNAs (ncRNAs) are essential transcriptional and post-transcriptional gene expression controllers, showing a highly regulated expression in space (cell types), time (developmental and ageing processes) and response to specific stimuli. These dynamic changes shape signaling pathways that are critical for the developmental processes of the nervous system and brain cell homeostasis. Diverse classes of ncRNAs have been involved in the cell response to OS and have been targeted in therapeutic designs. The perturbed expression of ncRNAs has been shown in human neurodegenerative diseases, with these changes contributing to pathogenic mechanisms, including OS and associated toxicity. In the present review, we summarize existing literature linking OS, neurodegeneration and ncRNA function. We provide evidences for the central role of OS in age-related neurodegenerative conditions, recapitulating the main types of regulatory ncRNAs with roles in the normal function of the nervous system and summarizing up-to-date information on ncRNA deregulation with a direct impact on OS associated with major neurodegenerative conditions.
Collapse
Affiliation(s)
- Ana Gámez-Valero
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/Casanova 143, 08036 Barcelona, Spain; (A.G.-V.); (A.G.-C.); (M.H.-L.); (M.S.-B.)
| | - Anna Guisado-Corcoll
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/Casanova 143, 08036 Barcelona, Spain; (A.G.-V.); (A.G.-C.); (M.H.-L.); (M.S.-B.)
| | - Marina Herrero-Lorenzo
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/Casanova 143, 08036 Barcelona, Spain; (A.G.-V.); (A.G.-C.); (M.H.-L.); (M.S.-B.)
| | - Maria Solaguren-Beascoa
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/Casanova 143, 08036 Barcelona, Spain; (A.G.-V.); (A.G.-C.); (M.H.-L.); (M.S.-B.)
| | - Eulàlia Martí
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/Casanova 143, 08036 Barcelona, Spain; (A.G.-V.); (A.G.-C.); (M.H.-L.); (M.S.-B.)
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Ministerio de Ciencia Innovación y Universidades, 28046 Madrid, Spain
| |
Collapse
|
26
|
Type III intermediate filaments as targets and effectors of electrophiles and oxidants. Redox Biol 2020; 36:101582. [PMID: 32711378 PMCID: PMC7381704 DOI: 10.1016/j.redox.2020.101582] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/05/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
Intermediate filaments (IFs) play key roles in cell mechanics, signaling and homeostasis. Their assembly and dynamics are finely regulated by posttranslational modifications. The type III IFs, vimentin, desmin, peripherin and glial fibrillary acidic protein (GFAP), are targets for diverse modifications by oxidants and electrophiles, for which their conserved cysteine residue emerges as a hot spot. Pathophysiological examples of these modifications include lipoxidation in cell senescence and rheumatoid arthritis, disulfide formation in cataracts and nitrosation in endothelial shear stress, although some oxidative modifications can also be detected under basal conditions. We previously proposed that cysteine residues of vimentin and GFAP act as sensors for oxidative and electrophilic stress, and as hinges influencing filament assembly. Accumulating evidence indicates that the structurally diverse cysteine modifications, either per se or in combination with other posttranslational modifications, elicit specific functional outcomes inducing distinct assemblies or network rearrangements, including filament stabilization, bundling or fragmentation. Cysteine-deficient mutants are protected from these alterations but show compromised cellular performance in network assembly and expansion, organelle positioning and aggresome formation, revealing the importance of this residue. Therefore, the high susceptibility to modification of the conserved cysteine of type III IFs and its cornerstone position in filament architecture sustains their role in redox sensing and integration of cellular responses. This has deep pathophysiological implications and supports the potential of this residue as a drug target. Type III intermediate filaments can be modified by many oxidants and electrophiles. Oxidative modifications of type III IFs occur in normal and pathological conditions. The conserved cysteine residue acts as a hub for redox/electrophilic modifications. Cysteine modifications elicit structure-dependent type III IF rearrangements. Type III intermediate filaments act as sensors for oxidative and electrophilic stress.
Collapse
|
27
|
Abstract
Redox proteomics is a field of proteomics that is concerned with the characterization of the oxidation state of proteins to gain information about their modulated structure, function, activity, and involvement in different physiological pathways. Oxidative modifications of proteins have been shown to be implicated in normal physiological processes of cells as well as in pathomechanisms leading to the development of cancer, diabetes, neurodegenerative diseases, and some rare hereditary metabolic diseases, like classic galactosemia. Reactive oxygen species generate a variety of reversible and irreversible modifications in amino acid residue side chains and within the protein backbone. These oxidative post-translational modifications (Ox-PTMs) can participate in the activation of signal transduction pathways and mediate the toxicity of harmful oxidants. Thus the application of advanced redox proteomics technologies is important for gaining insights into molecular mechanisms of diseases. Mass-spectrometry-based proteomics is one of the most powerful methods that can be used to give detailed qualitative and quantitative information on protein modifications and allows us to characterize redox proteomes associated with diseases. This Review illustrates the role and biological consequences of Ox-PTMs under basal and oxidative stress conditions by focusing on protein carbonylation and S-glutathionylation, two abundant modifications with an impact on cellular pathways that have been intensively studied during the past decade.
Collapse
Affiliation(s)
- Atef Mannaa
- Borg AlArab Higher Institute of Engineering and Technology , New Borg AlArab City , Alexandria , Egypt
| | - Franz-Georg Hanisch
- Institute of Biochemistry II, Medical Faculty , University of Cologne , Joseph-Stelzmann-Str. 52 , 50931 Cologne , Germany
| |
Collapse
|
28
|
Hamon MP, Ahmed EK, Baraibar MA, Friguet B. Proteome Oxidative Modifications and Impairment of Specific Metabolic Pathways During Cellular Senescence and Aging. Proteomics 2019; 20:e1800421. [PMID: 31507063 DOI: 10.1002/pmic.201800421] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/05/2019] [Indexed: 12/13/2022]
Abstract
Accumulation of oxidatively modified proteins is a hallmark of organismal aging in vivo and of cellular replicative senescence in vitro. Failure of protein maintenance is a major contributor to the age-associated accumulation of damaged proteins that is believed to participate to the age-related decline in cellular function. In this context, quantitative proteomics approaches, including 2-D gel electrophoresis (2-DE)-based methods, represent powerful tools for monitoring the extent of protein oxidative modifications at the proteome level and for identifying the targeted proteins, also referred as to the "oxi-proteome." Previous studies have identified proteins targeted by oxidative modifications during replicative senescence of human WI-38 fibroblasts and myoblasts and have been shown to represent a restricted set within the total cellular proteome that fall in key functional categories, such as energy metabolism, protein quality control, and cellular morphology. To provide mechanistic support into the role of oxidized proteins in the development of the senescent phenotype, untargeted metabolomic profiling is also performed for young and senescent myoblasts and fibroblasts. Metabolomic profiling is indicative of energy metabolism impairment in both senescent myoblasts and fibroblasts, suggesting a link between oxidative protein modifications and the altered cellular metabolism associated with the senescent phenotype of human myoblasts and fibroblasts.
Collapse
Affiliation(s)
- Marie-Paule Hamon
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Ageing, B2A-IBPS, F-75005, Paris, France
| | - Emad K Ahmed
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | | | - Bertrand Friguet
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Ageing, B2A-IBPS, F-75005, Paris, France
| |
Collapse
|
29
|
Colombo G, Garavaglia ML, Astori E, Giustarini D, Rossi R, Milzani A, Dalle-Donne I. Protein carbonylation in human bronchial epithelial cells exposed to cigarette smoke extract. Cell Biol Toxicol 2019; 35:345-360. [PMID: 30648195 DOI: 10.1007/s10565-019-09460-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/02/2019] [Indexed: 12/16/2022]
Abstract
Cigarette smoke is a well-established exogenous risk factor containing toxic reactive molecules able to induce oxidative stress, which in turn contributes to smoking-related diseases, including cardiovascular, pulmonary, and oral cavity diseases. We investigated the effects of cigarette smoke extract on human bronchial epithelial cells. Cells were exposed to various concentrations (2.5-5-10-20%) of cigarette smoke extract for 1, 3, and 24 h. Carbonylation was assessed by 2,4-dinitrophenylhydrazine using both immunocytochemical and Western immunoblotting assays. Cigarette smoke induced increasing protein carbonylation in a concentration-dependent manner. The main carbonylated proteins were identified by means of two-dimensional electrophoresis coupled to MALDI-TOF mass spectrometry analysis and database search (redox proteomics). We demonstrated that exposure of bronchial cells to cigarette smoke extract induces carbonylation of a large number of proteins distributed throughout the cell. Proteins undergoing carbonylation are involved in primary metabolic processes, such as protein and lipid metabolism and metabolite and energy production as well as in fundamental cellular processes, such as cell cycle and chromosome segregation, thus confirming that reactive carbonyl species contained in cigarette smoke markedly alter cell homeostasis and functions.
Collapse
Affiliation(s)
- Graziano Colombo
- Department of Biosciences, Università degli Studi di Milano, via Celoria 26, 20133, Milan, Italy.
| | - Maria Lisa Garavaglia
- Department of Biosciences, Università degli Studi di Milano, via Celoria 26, 20133, Milan, Italy
| | - Emanuela Astori
- Department of Biosciences, Università degli Studi di Milano, via Celoria 26, 20133, Milan, Italy
| | - Daniela Giustarini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100, Siena, Italy
| | - Ranieri Rossi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100, Siena, Italy
| | - Aldo Milzani
- Department of Biosciences, Università degli Studi di Milano, via Celoria 26, 20133, Milan, Italy
| | - Isabella Dalle-Donne
- Department of Biosciences, Università degli Studi di Milano, via Celoria 26, 20133, Milan, Italy
| |
Collapse
|
30
|
Zhang Y, Cui G, Wang Y, Gong Y, Wang Y. SIRT1 activation alleviates brain microvascular endothelial dysfunction in peroxisomal disorders. Int J Mol Med 2019; 44:995-1005. [PMID: 31257461 PMCID: PMC6657955 DOI: 10.3892/ijmm.2019.4250] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 06/11/2019] [Indexed: 02/03/2023] Open
Abstract
Peroxisomal disorders are genetically heterogeneous metabolic disorders associated with a deficit of very long chain fatty acid β-oxidation that commonly manifest as early-onset neurodegeneration. Brain microvascular endothelial dysfunction with increased permeability to monocytes has been described in X-linked adrenoleukodystrophy, one of the most common peroxisomal disorders caused by mutations of the ATP binding cassette subfamily D member 1 (ABCD1) gene. The present study demonstrated that dysregulation of sirtuin 1 (SIRT1) in human brain microvascular endothelial cells (HBMECs) mediates changes in adhesion molecules and tight-junction protein expression, as well as increased adhesion to monocytes associated with peroxisomal dysfunction due to ABCD1 or hydroxysteroid 17-β dehydrogenase 4 silencing. Furthermore, enhancement of the function of SIRT1 by resve-ratrol attenuated this molecular and functional dysregulation of HBMECs via modulation of the nuclear factor-κB and Krüppel-like factor 4 signaling pathways.
Collapse
Affiliation(s)
- Yunshan Zhang
- Department of Anatomy and Embryology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Guiyun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Yue Wang
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Yi Gong
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Yulan Wang
- Department of Anatomy and Embryology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| |
Collapse
|
31
|
Yang Z, Huang C, Wu Y, Chen B, Zhang W, Zhang J. Autophagy Protects the Blood-Brain Barrier Through Regulating the Dynamic of Claudin-5 in Short-Term Starvation. Front Physiol 2019; 10:2. [PMID: 30713499 PMCID: PMC6345697 DOI: 10.3389/fphys.2019.00002] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/07/2019] [Indexed: 01/14/2023] Open
Abstract
The blood-brain barrier (BBB) is essential for the exchange of nutrient and ions to maintain the homeostasis of central nervous system (CNS). BBB dysfunction is commonly associated with the disruption of endothelial tight junctions and excess permeability, which results in various CNS diseases. Therefore, maintaining the structural integrity and proper function of the BBB is essential for the homeostasis and physiological function of the CNS. Here, we showed that serum starvation disrupted the function of endothelial barrier as evidenced by decreased trans-endothelial electrical resistance, increased permeability, and redistribution of tight junction proteins such as Claudin-5 (Cldn5). Further analyses revealed that autophagy was activated and protected the integrity of endothelial barrier by scavenging ROS and inhibiting the redistribution of Cldn5 under starvation, as evidenced by accumulation of autophagic vacuoles and increased expression of LC3II/I, ATG5 and LAMP1. In addition, autophagosome was observed to package and eliminate the aggregated Cldn5 in cytosol as detected by immunoelectron microscopy (IEM) and stimulated emission depletion (STED) microscope. Moreover, Akt-mTOR-p70S6K pathway was found to be involved in the protective autophagy induced by starvation. Our data demonstrated that autophagy played an essential role in maintaining the integrity of endothelial barrier by regulating the localization of Cldn5 under starvation.
Collapse
Affiliation(s)
- Zhenguo Yang
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chunnian Huang
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yongfu Wu
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Bing Chen
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Wenqing Zhang
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jingjing Zhang
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
32
|
Abstract
Astrocytes are involved in many diseases of the central nervous system, not only as reactive cells to neuronal damage but also as primary actors in the pathological process. Astrogliopathy is a term used to designate the involvement of astrocytes as key elements in the pathogenesis and pathology of diseases and injuries of the central nervous system. Astrocytopathy is utilized to name non-reactive astrogliosis covering hypertrophy, atrophy and astroglial degeneration with loss of function in astrocytes and pathological remodeling, as well as senescent changes. Astrogliopathy and astrocytopathy are hallmarks of tauopathies—neurodegenerative diseases with abnormal hyper-phosphorylated tau aggregates in neurons and glial cells. The involvement of astrocytes covers different disease-specific types such as tufted astrocytes, astrocytic plaques, thorn-shaped astrocytes, granular/fuzzy astrocytes, ramified astrocytes and astrocytes with globular inclusions, as well as others which are unnamed but not uncommon in familial frontotemporal degeneration linked to mutations in the tau gene. Knowledge of molecular differences among tau-containing astrocytes is only beginning, and their distinct functional implications remain rather poorly understood. However, tau-containing astrocytes in certain conditions have deleterious effects on neuronal function and nervous system integrity. Moreover, recent studies have shown that tau-containing astrocytes obtained from human brain tauopathies have a capacity for abnormal tau seeding and spreading in wild type mice. Inclusive conceptions include a complex scenario involving neurons, glial cells and local environmental factors that potentiate each other and promote disease progression in tauopathies.
Collapse
|
33
|
Park HJ, Lee KW, Oh S, Yan R, Zhang J, Beach TG, Adler CH, Voronkov M, Braithwaite SP, Stock JB, Mouradian MM. Protein Phosphatase 2A and Its Methylation Modulating Enzymes LCMT-1 and PME-1 Are Dysregulated in Tauopathies of Progressive Supranuclear Palsy and Alzheimer Disease. J Neuropathol Exp Neurol 2018; 77:139-148. [PMID: 29281045 PMCID: PMC6251692 DOI: 10.1093/jnen/nlx110] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Hyperphosphorylated tau aggregates are characteristic of tauopathies including progressive supranuclear palsy (PSP) and Alzheimer disease (AD), but factors contributing to pathologic tau phosphorylation are not well understood. Here, we studied the regulation of the major tau phosphatase, the heterotrimeric AB55αC protein phosphatase 2 A (PP2A), in PSP and AD. The assembly and activity of this PP2A isoform are regulated by reversible carboxyl methylation of its catalytic C subunit, while the B subunit confers substrate specificity. We sought to address whether the decreases in PP2A methylation and its methylating enzyme, leucine carboxyl methyltransferase (LCMT-1), which are reported in AD, relate to tau pathology or to concomitant amyloid pathology by comparing them in the relatively pure tauopathy PSP. Immunohistochemical analysis of frontal cortices showed that methyl-PP2A is reduced while demethyl-PP2A is increased, with no changes in total PP2A or B55α subunit, resulting in a reduction in the methyl/demethyl PP2A ratio of 63% in PSP and 75% in AD compared to controls. Similarly, Western blot analyses showed a decrease of methyl-PP2A and an increase of demethyl-PP2A with a concomitant reduction in the methyl/demethyl PP2A ratio in both PSP (74%) and AD (76%) brains. This was associated with a decrease in LCMT-1 and an increase in the demethylating enzyme, protein phosphatase methylesterase (PME-1), in both diseases. These findings suggest that PP2A dysregulation in tauopathies may contribute to the accumulation of hyperphosphorylated tau and to neurodegeneration.
Collapse
Affiliation(s)
- Hye-Jin Park
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers—Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Kang-Woo Lee
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers—Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Stephanie Oh
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers—Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Run Yan
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers—Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Jie Zhang
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers—Robert Wood Johnson Medical School, Piscataway, New Jersey
| | | | | | | | | | - Jeffry B Stock
- Signum Biosciences, Princeton, New Jersey
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | - M Maral Mouradian
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers—Robert Wood Johnson Medical School, Piscataway, New Jersey
| |
Collapse
|
34
|
Ferrer I. Sisyphus in Neverland. J Alzheimers Dis 2018; 62:1023-1047. [PMID: 29154280 PMCID: PMC5870014 DOI: 10.3233/jad-170609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2017] [Indexed: 11/24/2022]
Abstract
The study of life and living organisms and the way in which these interact and organize to form social communities have been central to my career. I have been fascinated by biology, neurology, and neuropathology, but also by history, sociology, and art. Certain current historical, political, and social events, some occurring proximally but others affecting people in apparently distant places, have had an impact on me. Epicurus, Seneca, and Camus shared their philosophical positions which I learned from. Many scientists from various disciplines have been exciting sources of knowledge as well. I have created a world of hypothesis and experiments but I have also got carried away by serendipity following unexpected observations. It has not been an easy path; errors and wanderings are not uncommon, and opponents close to home much more abundant than one might imagine. Ambition, imagination, resilience, and endurance have been useful in moving ahead in response to setbacks. In the end, I have enjoyed my dedication to science and I am grateful to have glimpsed beauty in it. These are brief memories of a Spanish neuropathologist born and raised in Barcelona, EU.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona; Service of Pathological Anatomy, Bellvitge University Hospital; CIBERNED; Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
35
|
Berkiks I, Benmhammed H, Mesfioui A, Ouichou A, El Hasnaoui A, Mouden S, Touil T, Bahbiti Y, Nakache R, El Hessni A. Postnatal melatonin treatment protects against affective disorders induced by early-life immune stimulation by reducing the microglia cell activation and oxidative stress. Int J Neurosci 2017; 128:495-504. [PMID: 29077529 DOI: 10.1080/00207454.2017.1398156] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Systemic inflammation induced by neonatal infection may result as long-term hyper-activation of microglial cells followed by an overproduction of pro-inflammatory cytokines, such as tumor necrosis factor-alpha, nitric oxide and lipid peroxidation. Those inflammation mediators can trigger behavioral disruption and/or cognitive disorders. OBJECTIVE The present work aims to evaluate the effect of melatonin (a cytokine release modulator and antioxidant agent) in the reduction of the prefrontal microglia activation and depressive-like behaviors induced by lipopolysaccharide (LPS) injection in adult rats. RESULTS The effect of melatonin (5 mg/kg) was compared to minocycline (50 mg/kg), a well-known anti-inflammatory drug with potent inhibitory effect on microglial activation. Our results showed that LPS injection induced a significant increase in prefrontal cortex tumor necrosis factor-alpha and nitric oxide levels. Furthermore, lipid peroxidation and microglial activation were highly increased in the prefrontal cortex compared to control. The melatonin treatment induced a significant decrease on nitric oxide and lipid peroxidation levels in the prefrontal cortex and significant decrease on tumor necrosis factor-alpha and microglia activation. Melatonin can also induce a significant reduction in the anxiety and depression-like effect induced by PND9 LPS administration. CONCLUSION Our results demonstrated that melatonin possesses potent protective effect against the depression and anxiety induced by LPS. The underlying effect of melatonin is probably due to the reduction of nitric oxide toxic effect and lipid peroxidation in addition to its anti-inflammatory effect.
Collapse
Affiliation(s)
- I Berkiks
- a Department of Biology, Laboratory of Genetic, Neuroendocrinology, and Biotechnology, Faculty of Sciences , Ibn Tofail University , Kenitra , Morocco
| | - H Benmhammed
- a Department of Biology, Laboratory of Genetic, Neuroendocrinology, and Biotechnology, Faculty of Sciences , Ibn Tofail University , Kenitra , Morocco
| | - A Mesfioui
- a Department of Biology, Laboratory of Genetic, Neuroendocrinology, and Biotechnology, Faculty of Sciences , Ibn Tofail University , Kenitra , Morocco
| | - A Ouichou
- a Department of Biology, Laboratory of Genetic, Neuroendocrinology, and Biotechnology, Faculty of Sciences , Ibn Tofail University , Kenitra , Morocco
| | - A El Hasnaoui
- a Department of Biology, Laboratory of Genetic, Neuroendocrinology, and Biotechnology, Faculty of Sciences , Ibn Tofail University , Kenitra , Morocco
| | - S Mouden
- b Provincial Laboratory of Serology , Diagnostic Centre, Regional Hospital El Idrissi , Kenitra
| | - T Touil
- a Department of Biology, Laboratory of Genetic, Neuroendocrinology, and Biotechnology, Faculty of Sciences , Ibn Tofail University , Kenitra , Morocco
| | - Y Bahbiti
- a Department of Biology, Laboratory of Genetic, Neuroendocrinology, and Biotechnology, Faculty of Sciences , Ibn Tofail University , Kenitra , Morocco
| | - R Nakache
- a Department of Biology, Laboratory of Genetic, Neuroendocrinology, and Biotechnology, Faculty of Sciences , Ibn Tofail University , Kenitra , Morocco
| | - A El Hessni
- a Department of Biology, Laboratory of Genetic, Neuroendocrinology, and Biotechnology, Faculty of Sciences , Ibn Tofail University , Kenitra , Morocco
| |
Collapse
|
36
|
Baraibar MA, Hyzewicz J, Rogowska-Wrzesinska A, Bulteau AL, Prip-Buus C, Butler-Browne G, Friguet B. Impaired energy metabolism of senescent muscle satellite cells is associated with oxidative modifications of glycolytic enzymes. Aging (Albany NY) 2017; 8:3375-3389. [PMID: 27922824 PMCID: PMC5270674 DOI: 10.18632/aging.101126] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/16/2016] [Indexed: 01/05/2023]
Abstract
Accumulation of oxidized proteins is a hallmark of cellular and organismal aging. Adult muscle stem cell (or satellite cell) replication and differentiation is compromised with age contributing to sarcopenia. However, the molecular events related to satellite cell dysfunction during aging are not completely understood. In the present study we have addressed the potential impact of oxidatively modified proteins on the altered metabolism of senescent human satellite cells. By using a modified proteomics analysis we have found that proteins involved in protein quality control and glycolytic enzymes are the main targets of oxidation (carbonylation) and modification with advanced glycation/lipid peroxidation end products during the replicative senescence of satellite cells. Inactivation of the proteasome appeared to be a likely contributor to the accumulation of such damaged proteins. Metabolic and functional analyses revealed an impaired glucose metabolism in senescent cells. A metabolic shift leading to increased mobilization of non-carbohydrate substrates such as branched chain amino acids or long chain fatty acids was observed. Increased levels of acyl-carnitines indicated an increased turnover of storage and membrane lipids for energy production. Taken together, these results support a link between oxidative protein modifications and the altered cellular metabolism associated with the senescent phenotype of human myoblasts.
Collapse
Affiliation(s)
- Martín A Baraibar
- Sorbonne Universités, UPMC Univ Paris 06, UMR 8256, Biological Adaptation and Ageing- IBPS, CNRS UMR 8256, INSERM U1164, Paris, France
| | - Janek Hyzewicz
- Sorbonne Universités, UPMC Univ Paris 06, UMR 8256, Biological Adaptation and Ageing- IBPS, CNRS UMR 8256, INSERM U1164, Paris, France
| | | | - Anne-Laure Bulteau
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, INSERM U1016, CNRS UMR 8104, Institut Cochin, Paris, France
| | - Carina Prip-Buus
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, INSERM U1016, CNRS UMR 8104, Institut Cochin, Paris, France
| | - Gillian Butler-Browne
- Institut de Myologie, UPMC Univ Paris 06, UMRS INSERM U974, CNRS UMR 7215, CHU Pitié-Salpétrière, Sorbonne Universités, Paris, France
| | - Bertrand Friguet
- Sorbonne Universités, UPMC Univ Paris 06, UMR 8256, Biological Adaptation and Ageing- IBPS, CNRS UMR 8256, INSERM U1164, Paris, France
| |
Collapse
|
37
|
Laugeray A, Herzine A, Perche O, Richard O, Montecot-Dubourg C, Menuet A, Mazaud-Guittot S, Lesné L, Jegou B, Mortaud S. In utero and lactational exposure to low-doses of the pyrethroid insecticide cypermethrin leads to neurodevelopmental defects in male mice-An ethological and transcriptomic study. PLoS One 2017; 12:e0184475. [PMID: 29020013 PMCID: PMC5636066 DOI: 10.1371/journal.pone.0184475] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 08/19/2017] [Indexed: 01/03/2023] Open
Abstract
Accumulating evidence suggests that developmental exposure to environmental chemicals may modify the course of brain development, ultimately leading to neuropsychiatric / neurodegenerative disorders later in life. In the present study, we assessed the impact of one of the most frequently used pesticides in both residential and agricultural applications − the synthetic pyrethroid cypermethrin (CYP) − on developmental neurotoxicity (DNT). Female mice were perinatally exposed to low doses of CYP (5 and 20 mg/kg body weight) from gestation to postnatal day 15. Behavioral analyses were performed during the offspring’s early life and during adulthood. Postnatal analyses revealed that perinatal exposure to CYP disturbed motor development without modifying sensory and communicative skills. We found that later in life, CYP-exposed offspring expressed maladaptive behaviors in response to highly challenging tasks and abnormal sociability. Transcriptomic analyses performed in the offspring’s brain at the end of the exposure, highlighted mitochondrial dysfunction as a relevant pathomechanism underlying CYP-induced DNT. Interestingly, several genes involved in proteostasis maintenance were also shown to be dysregulated suggesting that alterations in biogenesis, folding, trafficking and degradation of proteins may significantly contribute to CYP-related DNT. From a regulatory perspective, this study highlights that behavioral and transcriptomic analyses are complementary tools providing useful direction for better DNT characterization, and as such, should be used together more systematically.
Collapse
Affiliation(s)
- Anthony Laugeray
- Immunologie et Neurogénétique Expérimentales et Moléculaires – UMR7355 CNRS – Orléans, France
- * E-mail: (AL); (SM)
| | - Ameziane Herzine
- Immunologie et Neurogénétique Expérimentales et Moléculaires – UMR7355 CNRS – Orléans, France
| | - Olivier Perche
- Immunologie et Neurogénétique Expérimentales et Moléculaires – UMR7355 CNRS – Orléans, France
- Département de génétique, Center Hospitalier Régional, Orléans, France
| | - Olivier Richard
- Immunologie et Neurogénétique Expérimentales et Moléculaires – UMR7355 CNRS – Orléans, France
| | - Céline Montecot-Dubourg
- Immunologie et Neurogénétique Expérimentales et Moléculaires – UMR7355 CNRS – Orléans, France
| | - Arnaud Menuet
- Immunologie et Neurogénétique Expérimentales et Moléculaires – UMR7355 CNRS – Orléans, France
| | | | | | - Bernard Jegou
- IRSET INSERM U 1085, Université de Rennes I, Rennes, France
| | - Stéphane Mortaud
- Immunologie et Neurogénétique Expérimentales et Moléculaires – UMR7355 CNRS – Orléans, France
- * E-mail: (AL); (SM)
| |
Collapse
|
38
|
Vasquez-Vivar J, Shi Z, Luo K, Thirugnanam K, Tan S. Tetrahydrobiopterin in antenatal brain hypoxia-ischemia-induced motor impairments and cerebral palsy. Redox Biol 2017; 13:594-599. [PMID: 28803128 PMCID: PMC5554922 DOI: 10.1016/j.redox.2017.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 07/28/2017] [Accepted: 08/01/2017] [Indexed: 12/24/2022] Open
Abstract
Antenatal brain hypoxia-ischemia, which occurs in cerebral palsy, is considered a significant cause of motor impairments in children. The mechanisms by which antenatal hypoxia-ischemia causes brain injury and motor deficits still need to be elucidated. Tetrahydrobiopterin is an important enzyme cofactor that is necessary to produce neurotransmitters and to maintain the redox status of the brain. A genetic deficiency of this cofactor from mutations of biosynthetic or recycling enzymes is a well-recognized factor in the development of childhood neurological disorders characterized by motor impairments, developmental delay, and encephalopathy. Experimental hypoxia-ischemia causes a decline in the availability of tetrahydrobiopterin in the immature brain. This decline coincides with the loss of brain function, suggesting this occurrence contributes to neuronal dysfunction and motor impairments. One possible mechanism linking tetrahydrobiopterin deficiency, hypoxia-ischemia, and neuronal injury is oxidative injury. Evidence of the central role of the developmental biology of tetrahydrobiopterin in response to hypoxic ischemic brain injury, especially the development of motor deficits, is discussed.
Collapse
Affiliation(s)
- Jeannette Vasquez-Vivar
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| | - Zhongjie Shi
- Wayne State University School of Medicine and Children's Hospital of Michigan, 3901 Beaubien, Room 5177, Carls Bldg., Detroit, MI 48201, USA
| | - Kehuan Luo
- Wayne State University School of Medicine and Children's Hospital of Michigan, 3901 Beaubien, Room 5177, Carls Bldg., Detroit, MI 48201, USA
| | - Karthikeyan Thirugnanam
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Sidhartha Tan
- Wayne State University School of Medicine and Children's Hospital of Michigan, 3901 Beaubien, Room 5177, Carls Bldg., Detroit, MI 48201, USA.
| |
Collapse
|
39
|
Ferrer I. Diversity of astroglial responses across human neurodegenerative disorders and brain aging. Brain Pathol 2017; 27:645-674. [PMID: 28804999 PMCID: PMC8029391 DOI: 10.1111/bpa.12538] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/24/2017] [Indexed: 12/11/2022] Open
Abstract
Astrogliopathy refers to alterations of astrocytes occurring in diseases of the nervous system, and it implies the involvement of astrocytes as key elements in the pathogenesis and pathology of diseases and injuries of the central nervous system. Reactive astrocytosis refers to the response of astrocytes to different insults to the nervous system, whereas astrocytopathy indicates hypertrophy, atrophy/degeneration and loss of function and pathological remodeling occurring as a primary cause of a disease or as a factor contributing to the development and progression of a particular disease. Reactive astrocytosis secondary to neuron loss and astrocytopathy due to intrinsic alterations of astrocytes occur in neurodegenerative diseases, overlap each other, and, together with astrocyte senescence, contribute to disease-specific astrogliopathy in aging and neurodegenerative diseases with abnormal protein aggregates in old age. In addition to the well-known increase in glial fibrillary acidic protein and other proteins in reactive astrocytes, astrocytopathy is evidenced by deposition of abnormal proteins such as β-amyloid, hyper-phosphorylated tau, abnormal α-synuclein, mutated huntingtin, phosphorylated TDP-43 and mutated SOD1, and PrPres , in Alzheimer's disease, tauopathies, Lewy body diseases, Huntington's disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease, respectively. Astrocytopathy in these diseases can also be manifested by impaired glutamate transport; abnormal metabolism and release of neurotransmitters; altered potassium, calcium and water channels resulting in abnormal ion and water homeostasis; abnormal glucose metabolism; abnormal lipid and, particularly, cholesterol metabolism; increased oxidative damage and altered oxidative stress responses; increased production of cytokines and mediators of the inflammatory response; altered expression of connexins with deterioration of cell-to-cell networks and transfer of gliotransmitters; and worsening function of the blood brain barrier, among others. Increased knowledge of these aspects will permit a better understanding of brain aging and neurodegenerative diseases in old age as complex disorders in which neurons are not the only players.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental TherapeuticsUniversity of BarcelonaBarcelonaSpain
- Institute of NeuropathologyPathologic Anatomy Service, Bellvitge University Hospital, IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- Biomedical Network Research Center on Neurodegenerative Diseases (CIBERNED), Institute Carlos IIIMadridSpain
| |
Collapse
|
40
|
|
41
|
Garcia-Esparcia P, Koneti A, Rodríguez-Oroz MC, Gago B, Del Rio JA, Ferrer I. Mitochondrial activity in the frontal cortex area 8 and angular gyrus in Parkinson's disease and Parkinson's disease with dementia. Brain Pathol 2017; 28:43-57. [PMID: 27984680 DOI: 10.1111/bpa.12474] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 12/01/2016] [Indexed: 11/30/2022] Open
Abstract
Altered mitochondrial function is characteristic in the substantia nigra in Parkinson's disease (PD). Information about mitochondria in other brain regions such as the cerebral cortex is conflicting mainly because most studies have not contemplated the possibility of variable involvement depending on the region, stage of disease progression and clinical symptoms such as the presence or absence of dementia. RT-qPCR of 18 nuclear mRNAs encoding subunits of mitochondrial complexes and 12 mRNAs encoding energy metabolism-related enzymes; western blotting of mitochondrial proteins; and analysis of enzymatic activities of complexes I, II, II, IV and V of the respiratory chain were assessed in frontal cortex area 8 and the angular gyrus of middle-aged individuals (MA), and those with incidental PD (iPD), long-lasting PD with parkinsonism without dementia (PD) and long-lasting PD with dementia (PDD). Up-regulation of several genes was found in frontal cortex area 8 in PD when compared with MA and in the angular gyrus in iPD when compared with MA. Marked down-regulation of genes encoding mitochondrial subunits and energy metabolism-related enzymes occurs in frontal cortex but only of genes coding for energy metabolism-related enzymes in the angular gyrus in PDD. Significant decrease in the protein expression levels of several mitochondrial subunits encoded by these genes occurs in frontal cortex area 8 and angular gyrus in PDD. Moreover, expression of MT-ND1 which is encoded by mitochondrial DNA is also reduced in PDD. Reduced enzymatic activity of complex III in frontal cortex area 8 and angular gyrus is observed in PD, but dramatic reduction in the activity of complexes I, II, II and IV in both regions characterizes PDD. Dementia in the context of PD is linked to region-specific deregulation of genomic genes encoding subunits of mitochondrial complexes and to marked reduction in the activity of mitochondrial complexes I, II, III and IV.
Collapse
Affiliation(s)
- Paula Garcia-Esparcia
- Institute of Neuropathology, Bellvitge University Hospital, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de LLobregat, Catalonia, Spain
| | - Anusha Koneti
- Institute of Neuropathology, Bellvitge University Hospital, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de LLobregat, Catalonia, Spain
| | - M Cruz Rodríguez-Oroz
- University Hospital Donostia, San Sebastián, Spain.,Ikerbasque (Basque Foundation for Science), Bilbao, Spain.,Biodonostia Research Institute, San Sebastián, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Belen Gago
- Biodonostia Research Institute, San Sebastián, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - José Antonio Del Rio
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Department of Cell Biology, University of Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Isidro Ferrer
- Institute of Neuropathology, Bellvitge University Hospital, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de LLobregat, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
42
|
Naudí A, Cabré R, Dominguez-Gonzalez M, Ayala V, Jové M, Mota-Martorell N, Piñol-Ripoll G, Gil-Villar MP, Rué M, Portero-Otín M, Ferrer I, Pamplona R. Region-specific vulnerability to lipid peroxidation and evidence of neuronal mechanisms for polyunsaturated fatty acid biosynthesis in the healthy adult human central nervous system. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:485-495. [PMID: 28185952 DOI: 10.1016/j.bbalip.2017.02.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 12/23/2016] [Accepted: 02/05/2017] [Indexed: 12/28/2022]
Abstract
Lipids played a determinant role in the evolution of the brain. It is postulated that the morphological and functional diversity among neural cells of the human central nervous system (CNS) is projected and achieved through the expression of particular lipid profiles. The present study was designed to evaluate the differential vulnerability to oxidative stress mediated by lipids through a cross-regional comparative approach. To this end, we compared 12 different regions of CNS of healthy adult subjects, and the fatty acid profile and vulnerability to lipid peroxidation, were determined by gas chromatography (GC) and gas chromatography/mass spectrometry (GC/MS), respectively. In addition, different components involved in PUFA biosynthesis, as well as adaptive defense mechanisms against lipid peroxidation, were also measured by western blot and immunohistochemistry, respectively. We found that: i) four fatty acids (18.1n-9, 22:6n-3, 20:1n-9, and 18:0) are significant discriminators among CNS regions; ii) these differential fatty acid profiles generate a differential selective neural vulnerability (expressed by the peroxidizability index); iii) the cross-regional differences for the fatty acid profiles follow a caudal-cranial gradient which is directly related to changes in the biosynthesis pathways which can be ascribed to neuronal cells; and iv) the higher the peroxidizability index for a given human brain region, the lower concentration of the protein damage markers, likely supported by the presence of adaptive antioxidant mechanisms. In conclusion, our results suggest that there is a region-specific vulnerability to lipid peroxidation and offer evidence of neuronal mechanisms for polyunsaturated fatty acid biosynthesis in the human central nervous system.
Collapse
Affiliation(s)
- Alba Naudí
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), E-25198 Lleida, Spain
| | - Rosanna Cabré
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), E-25198 Lleida, Spain
| | - Mayelin Dominguez-Gonzalez
- Institute of Neuropathology, Bellvitge University Hospital, Department of Pathology and Experimental Therapeutics, University of Barcelona, E-08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Victoria Ayala
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), E-25198 Lleida, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), E-25198 Lleida, Spain
| | - Natalia Mota-Martorell
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), E-25198 Lleida, Spain
| | | | | | - Montserrat Rué
- Department of Basic Medical Sciences, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), E-25198 Lleida, Spain
| | - Manuel Portero-Otín
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), E-25198 Lleida, Spain
| | - Isidre Ferrer
- Institute of Neuropathology, Bellvitge University Hospital, Department of Pathology and Experimental Therapeutics, University of Barcelona, E-08908, L'Hospitalet de Llobregat, Barcelona, Spain; Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), ISCIII, Spain.
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), E-25198 Lleida, Spain.
| |
Collapse
|
43
|
Fruhmann G, Seynnaeve D, Zheng J, Ven K, Molenberghs S, Wilms T, Liu B, Winderickx J, Franssens V. Yeast buddies helping to unravel the complexity of neurodegenerative disorders. Mech Ageing Dev 2017; 161:288-305. [DOI: 10.1016/j.mad.2016.05.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/22/2016] [Accepted: 05/02/2016] [Indexed: 12/31/2022]
|
44
|
Butterfield DA, Palmieri EM, Castegna A. Clinical implications from proteomic studies in neurodegenerative diseases: lessons from mitochondrial proteins. Expert Rev Proteomics 2016; 13:259-74. [PMID: 26837425 DOI: 10.1586/14789450.2016.1149470] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mitochondria play a key role in eukaryotic cells, being mediators of energy, biosynthetic and regulatory requirements of these cells. Emerging proteomics techniques have allowed scientists to obtain the differentially expressed proteome or the proteomic redox status in mitochondria. This has unmasked the diversity of proteins with respect to subcellular location, expression and interactions. Mitochondria have become a research 'hot spot' in subcellular proteomics, leading to identification of candidate clinical targets in neurodegenerative diseases in which mitochondria are known to play pathological roles. The extensive efforts to rapidly obtain differentially expressed proteomes and unravel the redox proteomic status in mitochondria have yielded clinical insights into the neuropathological mechanisms of disease, identification of disease early stage and evaluation of disease progression. Although current technical limitations hamper full exploitation of the mitochondrial proteome in neurosciences, future advances are predicted to provide identification of specific therapeutic targets for neurodegenerative disorders.
Collapse
Affiliation(s)
- D Allan Butterfield
- a Department of Chemistry, and Sanders-Brown Center on Aging , University of Kentucky , Lexington , KY , USA
| | - Erika M Palmieri
- b Department of Biosciences, Biotechnologies and Biopharmaceutics , University of Bari 'Aldo Moro' , Bari , Italy
| | - Alessandra Castegna
- b Department of Biosciences, Biotechnologies and Biopharmaceutics , University of Bari 'Aldo Moro' , Bari , Italy
| |
Collapse
|
45
|
Roberts JH, Liu F, Karnuta JM, Fitzgerald MC. Discovery of Age-Related Protein Folding Stability Differences in the Mouse Brain Proteome. J Proteome Res 2016; 15:4731-4741. [PMID: 27806573 DOI: 10.1021/acs.jproteome.6b00927] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Described here is the application of thermodynamic stability measurements to study age-related differences in the folding and stability of proteins in a rodent model of aging. Thermodynamic stability profiles were generated for 809 proteins in brain cell lysates from mice, aged 6 (n = 7) and 18 months (n = 9) using the Stability of Proteins from Rates of Oxidation (SPROX) technique. The biological variability of the protein stability measurements was low and within the experimental error of SPROX. A total of 83 protein hits were detected with age-related stability differences in the brain samples. Remarkably, the large majority of the brain protein hits were destabilized in the old mice, and the hits were enriched in proteins that have slow turnover rates (p < 0.07). Furthermore, 70% of the hits have been previously linked to aging or age-related diseases. These results help validate the use of thermodynamic stability measurements to capture relevant age-related proteomic changes and establish a new biophysical link between these proteins and aging.
Collapse
Affiliation(s)
- Julia H Roberts
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| | - Fang Liu
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| | - Jaret M Karnuta
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| | - Michael C Fitzgerald
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| |
Collapse
|
46
|
Yoshioka K, Komatsu T, Hanaoka K, Ueno T, Terai T, Nagano T, Urano Y. Discovery of a pyruvylated peptide-metabolizing enzyme using a fluorescent substrate-based protein discovery technique. Chem Commun (Camb) 2016; 52:4377-80. [PMID: 26925595 DOI: 10.1039/c6cc00829a] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We employed a fluorescent substrate-based target discovery approach to screen the enzymome for metabolic activity towards pyruvyl-amidated peptides, and identified an acylamino acid-releasing enzyme (APEH). Cells overexpressing APEH exhibited higher metabolic activity towards the probe, N-pyruvyl-leucyl-7-amido-4-methylcoumarin (Pyr-Leu-AMC), while the selective APEH inhibitor AA74-1 blocked the reaction. Metabolism of various pyruvylated peptides in liver lysate was almost completely blocked by AA74-1. Pyruvyl peptides are synthesized in response to oxidative stress, but their biological role is poorly understood; identification of a key contributor to their metabolism should stimulate research on pathways leading from oxidative stress to protein modification and biological output.
Collapse
Affiliation(s)
- Kentaro Yoshioka
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Toru Komatsu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan and Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Kenjiro Hanaoka
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Tasuku Ueno
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Takuya Terai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Tetsuo Nagano
- Drug Discovery Initiative, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan.
| | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan and Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan and Core Research for Evolutional Science and Technology (CREST) Investigator, Japan Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
47
|
Abstract
Reactive oxygen species (ROS), which are both a natural byproduct of oxidative metabolism and an undesirable byproduct of many environmental stressors, can damage all classes of cellular macromolecules and promote diseases from cancer to neurodegeneration. The actions of ROS are mitigated by the transcription factor NRF2, which regulates expression of antioxidant genes via its interaction with cis-regulatory antioxidant response elements (AREs). However, despite the seemingly straightforward relationship between the opposing forces of ROS and NRF2, regulatory precision in the NRF2 network is essential. Genetic variants that alter NRF2 stability or alter ARE sequences have been linked to a range of diseases. NRF2 hyperactivating mutations are associated with tumorigenesis. On the subtler end of the spectrum, single nucleotide variants (SNVs) that alter individual ARE sequences have been linked to neurodegenerative disorders including progressive supranuclear palsy and Parkinson’s disease, as well as other diseases. Although the human health implications of NRF2 dysregulation have been recognized for some time, a systems level view of this regulatory network is beginning to highlight key NRF2-targeted AREs consistently associated with disease.
Collapse
|
48
|
Johansson I, Monsen VT, Pettersen K, Mildenberger J, Misund K, Kaarniranta K, Schønberg S, Bjørkøy G. The marine n-3 PUFA DHA evokes cytoprotection against oxidative stress and protein misfolding by inducing autophagy and NFE2L2 in human retinal pigment epithelial cells. Autophagy 2016; 11:1636-51. [PMID: 26237736 PMCID: PMC4590664 DOI: 10.1080/15548627.2015.1061170] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Accumulation and aggregation of misfolded proteins is a hallmark of several diseases collectively known as proteinopathies. Autophagy has a cytoprotective role in diseases associated with protein aggregates. Age-related macular degeneration (AMD) is the most common neurodegenerative eye disease that evokes blindness in elderly. AMD is characterized by degeneration of retinal pigment epithelial (RPE) cells and leads to loss of photoreceptor cells and central vision. The initial phase associates with accumulation of intracellular lipofuscin and extracellular deposits called drusen. Epidemiological studies have suggested an inverse correlation between dietary intake of marine n-3 polyunsaturated fatty acids (PUFAs) and the risk of developing neurodegenerative diseases, including AMD. However, the disease-preventive mechanism(s) mobilized by n-3 PUFAs is not completely understood. In human retinal pigment epithelial cells we find that physiologically relevant doses of the n-3 PUFA docosahexaenoic acid (DHA) induce a transient increase in cellular reactive oxygen species (ROS) levels that activates the oxidative stress response regulator NFE2L2/NRF2 (nuclear factor, erythroid derived 2, like 2). Simultaneously, there is a transient increase in intracellular protein aggregates containing SQSTM1/p62 (sequestosome 1) and an increase in autophagy. Pretreatment with DHA rescues the cells from cell cycle arrest induced by misfolded proteins or oxidative stress. Cells with a downregulated oxidative stress response, or autophagy, respond with reduced cell growth and survival after DHA supplementation. These results suggest that DHA both induces endogenous antioxidants and mobilizes selective autophagy of misfolded proteins. Both mechanisms could be relevant to reduce the risk of developing aggregate-associate diseases such as AMD.
Collapse
Affiliation(s)
- Ida Johansson
- a Department of Laboratory Medicine ; Children's and Women's Health; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway.,b Department of Technology ; University College of Sør-Trøndelag ; Trondheim , Norway.,c Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine ; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Vivi Talstad Monsen
- a Department of Laboratory Medicine ; Children's and Women's Health; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Kristine Pettersen
- a Department of Laboratory Medicine ; Children's and Women's Health; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway.,b Department of Technology ; University College of Sør-Trøndelag ; Trondheim , Norway.,c Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine ; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Jennifer Mildenberger
- b Department of Technology ; University College of Sør-Trøndelag ; Trondheim , Norway.,c Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine ; Norwegian University of Science and Technology ; Trondheim , Norway.,d Department of Cancer Research and Molecular Medicine ; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Kristine Misund
- d Department of Cancer Research and Molecular Medicine ; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway.,e KG Jebsen Center for Myeloma Research; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Kai Kaarniranta
- f Department of Ophthalmology ; Institute of Clinical Medicine; University of Eastern Finland ; Kuopio , Finland.,g Department of Ophthalmology ; Kuopio University Hospital ; Kuopio , Finland
| | - Svanhild Schønberg
- a Department of Laboratory Medicine ; Children's and Women's Health; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Geir Bjørkøy
- b Department of Technology ; University College of Sør-Trøndelag ; Trondheim , Norway.,c Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine ; Norwegian University of Science and Technology ; Trondheim , Norway
| |
Collapse
|
49
|
Xu J, Begley P, Church SJ, Patassini S, McHarg S, Kureishy N, Hollywood KA, Waldvogel HJ, Liu H, Zhang S, Lin W, Herholz K, Turner C, Synek BJ, Curtis MA, Rivers-Auty J, Lawrence CB, Kellett KAB, Hooper NM, Vardy ERLC, Wu D, Unwin RD, Faull RLM, Dowsey AW, Cooper GJS. Elevation of brain glucose and polyol-pathway intermediates with accompanying brain-copper deficiency in patients with Alzheimer's disease: metabolic basis for dementia. Sci Rep 2016; 6:27524. [PMID: 27276998 DOI: 10.1038/srep27524] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 05/19/2016] [Indexed: 12/25/2022] Open
Abstract
Impairment of brain-glucose uptake and brain-copper regulation occurs in Alzheimer's disease (AD). Here we sought to further elucidate the processes that cause neurodegeneration in AD by measuring levels of metabolites and metals in brain regions that undergo different degrees of damage. We employed mass spectrometry (MS) to measure metabolites and metals in seven post-mortem brain regions of nine AD patients and nine controls, and plasma-glucose and plasma-copper levels in an ante-mortem case-control study. Glucose, sorbitol and fructose were markedly elevated in all AD brain regions, whereas copper was correspondingly deficient throughout (all P < 0.0001). In the ante-mortem case-control study, by contrast, plasma-glucose and plasma-copper levels did not differ between patients and controls. There were pervasive defects in regulation of glucose and copper in AD brain but no evidence for corresponding systemic abnormalities in plasma. Elevation of brain glucose and deficient brain copper potentially contribute to the pathogenesis of neurodegeneration in AD.
Collapse
Affiliation(s)
- Jingshu Xu
- School of Biological Sciences, and Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, University of Auckland, New Zealand.,Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.,Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, and Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | - Paul Begley
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, and Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | - Stephanie J Church
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, and Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | - Stefano Patassini
- School of Biological Sciences, and Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, University of Auckland, New Zealand.,Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.,Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, and Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | - Selina McHarg
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, and Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | - Nina Kureishy
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, and Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | - Katherine A Hollywood
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, and Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | - Henry J Waldvogel
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Hong Liu
- School of Biological Sciences, and Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, University of Auckland, New Zealand
| | - Shaoping Zhang
- School of Biological Sciences, and Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, University of Auckland, New Zealand
| | - Wanchang Lin
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, and Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | - Karl Herholz
- Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | - Clinton Turner
- Anatomical Pathology, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - Beth J Synek
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.,Anatomical Pathology, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Jack Rivers-Auty
- Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | - Catherine B Lawrence
- Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | - Katherine A B Kellett
- Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | - Nigel M Hooper
- Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | | | - Donghai Wu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Richard D Unwin
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, and Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | - Richard L M Faull
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Andrew W Dowsey
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, and Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| | - Garth J S Cooper
- School of Biological Sciences, and Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, University of Auckland, New Zealand.,Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.,Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, and Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, United Kingdom
| |
Collapse
|
50
|
Domínguez M, de Oliveira E, Odena MA, Portero M, Pamplona R, Ferrer I. Redox proteomic profiling of neuroketal-adducted proteins in human brain: Regional vulnerability at middle age increases in the elderly. Free Radic Biol Med 2016; 95:1-15. [PMID: 26968793 DOI: 10.1016/j.freeradbiomed.2016.02.034] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/03/2016] [Accepted: 02/27/2016] [Indexed: 02/08/2023]
Abstract
Protein lipoxidation was assessed in the parietal cortex (PC), frontal cortex (FC), and cingulate gyrus (CG) in middle-aged and old-aged individuals with no clinical manifestations of cognitive impairment, in order to increase understanding of regional brain vulnerability to oxidative damage during aging. Twenty-five lipoxidized proteins were identified in all the three regions although with regional specificities, by using redox proteomics to detect target proteins of neuroketals (NKT) adduction. The number of cases with NKT-adducted proteins was higher in old-aged individuals but most oxidized proteins were already present in middle-aged individuals. Differences in vulnerability to oxidation were dependent on the sub-cellular localization, secondary structure, and external exposition of certain amino acids. Lipoxidized proteins included those involved in energy metabolism, cytoskeleton, proteostasis, neurotransmission and O2/CO2, and heme metabolism. Total NKT and soluble oligomer levels were estimated employing slot-blot, and these were compared between age groups. Oligomers increased with age in PC and FC; NKT significantly increased with age in FC, whereas total NKT and oligomer levels were not modified in CG, thus highlighting differences in brain regional vulnerability with age. Oligomers significantly correlated with NKT levels in the three cortical regions, suggesting that protein NKT adduction parallels soluble oligomer formation.
Collapse
Affiliation(s)
- Mayelín Domínguez
- Institute of Neuropathology, University Hospital of Bellvitge, IDIBELL (Biomedical Research Institute of Bellvitge), Carrer Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain.
| | | | | | - Manuel Portero
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, 25198 Lleida, Spain.
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida, 25198 Lleida, Spain.
| | - Isidro Ferrer
- Institute of Neuropathology, University Hospital of Bellvitge, IDIBELL (Biomedical Research Institute of Bellvitge), Carrer Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; Department of Pathology and Experimental Therapeutics, University of Barcelona, Carrer Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; CIBERNED (Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas), Spain.
| |
Collapse
|