101
|
Hanaki M, Murakami K, Katayama S, Akagi KI, Irie K. Mechanistic analyses of the suppression of amyloid β42 aggregation by apomorphine. Bioorg Med Chem 2018; 26:1538-1546. [PMID: 29429575 DOI: 10.1016/j.bmc.2018.01.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/29/2018] [Accepted: 01/31/2018] [Indexed: 01/05/2023]
Abstract
(R)-Apomorphine (1) has the potential to reduce the accumulation of amyloid β-protein (Aβ42), a causative agent of Alzheimer's disease (AD). Although the inhibition of Aβ42 aggregation by 1 is ascribable to the antioxidative effect of its phenol moiety, its inhibitory mechanism at the molecular level remains to be fully elucidated. LC-MS and UV analyses revealed that 1 is autoxidized during incubation to produce an unstable o-quinone form (2), which formed a Michael adduct with Lys 16 and 28 of Aβ42. A further autoxidized form of 1 (3) with o-quinone and phenanthrene moieties suppressed Aβ42 aggregation comparable to 1, whereas treating 1 with a reductant, tris(2-carboxyethyl)phosphine diminished its inhibitory activity. 1H-15N SOFAST-HMQC NMR studies suggested that 1 interacts with Arg5, His13,14, Gln15, and Lys16 of the Aβ42 monomer. These regions form intermolecular β-sheets in Aβ42 aggregates. Since 3 did not perturb the chemical shift of monomeric Aβ42, we performed aggregation experiments using 1,1,1,3,3,3-hexafluoro-2-propanol-treated Aβ42 to investigate whether 3 associates with Aβ42 oligomers. Compounds 1 and 3 delayed the onset of the oligomer-driven nucleation phase. Despite their cytotoxicity, they did not exacerbate Aβ42-mediated neurotoxicity in SH-SY5Y neuroblastoma cells. These results demonstrate that extension of the conjugated system in 1 by autoxidation can promote its planarity, which is required for intercalation into the β-sheet of Aβ42 nuclei, thereby suppressing further aggregation.
Collapse
Affiliation(s)
- Mizuho Hanaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Kazuma Murakami
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Sumie Katayama
- National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Ken-Ichi Akagi
- National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Kazuhiro Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| |
Collapse
|
102
|
Neuroprotective effect of berberine against environmental heavy metals-induced neurotoxicity and Alzheimer's-like disease in rats. Food Chem Toxicol 2017; 111:432-444. [PMID: 29170048 DOI: 10.1016/j.fct.2017.11.025] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 11/15/2017] [Accepted: 11/18/2017] [Indexed: 01/20/2023]
Abstract
Heavy metals are reported as neurodegenerative disorders progenitor. They play a role in the precipitation of abnormal β-amyloid protein and hyper-phosphorylated tau, the main hallmarks of Alzheimer's disease (AD). The present study aimed to validate the heavy metals-induced Alzheimer's-like disease in rats as an experimental model of AD and explore the therapeutic effect of berberine via tracking its effect on the oxidative stress-inflammatory pathway. Alzheimer's-like disease was induced in rats orally by a mixture of aluminium, cadmium and fluoride for three months, followed by berberine treatment for another one month. Berberine significantly improved the cognitive behaviors in Morris water maze test and offered a protective effect against heavy metals-induced memory impairment. Docking results showed that berberine inhibited AChE, COX-2 and TACE. Matching with in silico study, berberine downregulated the AChE expression and inhibited its activity in the brain tissues. Also, it normalized the production of TNF- α, IL-12, IL-6 and IL-1β. Moreover, it evoked the production of antioxidant Aβ40 and inhibited the formation of Aβ42, responsible for the aggregations of amyloid-β plaques. Histopathological examination confirmed the neuroprotective effect of berberine. The present data advocate the possible beneficial effect of berberine as therapeutic modality for Alzheimer's disease via its antiinflammatory/antioxidant mechanism.
Collapse
|
103
|
Wijesekara N, Gonçalves RA, De Felice FG, Fraser PE. Impaired peripheral glucose homeostasis and Alzheimer's disease. Neuropharmacology 2017; 136:172-181. [PMID: 29169962 DOI: 10.1016/j.neuropharm.2017.11.027] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/12/2017] [Accepted: 11/16/2017] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia. Recent studies suggest that metabolic disturbances, particularly type 2 diabetes (T2D) increase the risk of cognitive decline and AD. AD is also a risk factor for T2D, and a growing body of evidence indicates that these diseases are connected both at clinical and molecular levels. In T2D, peripheral insulin resistance, hyperglycemia and eventually insulin deficiency develops, leading to an overall decline in tissue health. More recently, brain insulin resistance has been shown to be a key feature of AD that is linked to neuronal dysfunction and cognitive impairment. Furthermore, both AD and T2D are amyloidogenic diseases, with abnormal aggregation of amyloid-β peptide (Aβ) and islet amyloid polypeptide (IAPP) respectively contributing to cellular death and disease pathogenesis. Emerging data suggests that Aβ may have peripheral effects including its co-deposition in the pancreas. In this review, we discuss how peripheral effects of Aβ and metabolic disturbances may impact AD pathogenesis. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Nadeeja Wijesekara
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, Ontario, M5T 2S8, Canada.
| | - Rafaella Araujo Gonçalves
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, Ontario, M5T 2S8, Canada; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, Ontario, M5T 2S8, Canada; Department of Medical Biophysics, University of Toronto, Canada.
| |
Collapse
|
104
|
Mazensky D, Flesarova S, Sulla I. Arterial Blood Supply to the Spinal Cord in Animal Models of Spinal Cord Injury. A Review. Anat Rec (Hoboken) 2017; 300:2091-2106. [PMID: 28972696 DOI: 10.1002/ar.23694] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/06/2017] [Accepted: 07/18/2017] [Indexed: 02/02/2023]
Abstract
Animal models are used to examine the results of experimental spinal cord injury. Alterations in spinal cord blood supply caused by complex spinal cord injuries contribute significantly to the diversity and severity of the spinal cord damage, particularly ischemic changes. However, the literature has not completely clarified our knowledge of anatomy of the complex three-dimensional arterial system of the spinal cord in experimental animals, which can impede the translation of experimental results to human clinical applications. As the literary sources dealing with the spinal cord arterial blood supply in experimental animals are limited and scattered, the authors performed a review of the anatomy of the arterial blood supply to the spinal cord in several experimental animals, including pigs, dogs, cats, rabbits, guinea pigs, rats, and mice and created a coherent format discussing the interspecies differences. This provides researchers with a valuable tool for the selection of the most suitable animal model for their experiments in the study of spinal cord ischemia and provides clinicians with a basis for the appropriate translation of research work to their clinical applications. Anat Rec, 300:2091-2106, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David Mazensky
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Slavka Flesarova
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Igor Sulla
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| |
Collapse
|
105
|
Cavallaro RA, Nicolia V, Fiorenza MT, Scarpa S, Fuso A. S-Adenosylmethionine and Superoxide Dismutase 1 Synergistically Counteract Alzheimer's Disease Features Progression in TgCRND8 Mice. Antioxidants (Basel) 2017; 6:antiox6040076. [PMID: 28973985 PMCID: PMC5745486 DOI: 10.3390/antiox6040076] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/26/2017] [Accepted: 09/27/2017] [Indexed: 01/30/2023] Open
Abstract
Recent evidence emphasizes the role of dysregulated one-carbon metabolism in Alzheimer’s Disease (AD). Exploiting a nutritional B-vitamin deficiency paradigm, we have previously shown that PSEN1 and BACE1 activity is modulated by one-carbon metabolism, leading to increased amyloid production. We have also demonstrated that S-adenosylmethionine (SAM) supplementation contrasted the AD-like features, induced by B-vitamin deficiency. In the present study, we expanded these observations by investigating the effects of SAM and SOD (Superoxide dismutase) association. TgCRND8 AD mice were fed either with a control or B-vitamin deficient diet, with or without oral supplementation of SAM + SOD. We measured oxidative stress by lipid peroxidation assay, PSEN1 and BACE1 expression by Real-Time Polymerase Chain Reaction (PCR), amyloid deposition by ELISA assays and immunohistochemistry. We found that SAM + SOD supplementation prevents the exacerbation of AD-like features induced by B vitamin deficiency, showing synergistic effects compared to either SAM or SOD alone. SAM + SOD supplementation also contrasts the amyloid deposition typically observed in TgCRND8 mice. Although the mechanisms underlying the beneficial effect of exogenous SOD remain to be elucidated, our findings identify that the combination of SAM + SOD could be carefully considered as co-adjuvant of current AD therapies.
Collapse
Affiliation(s)
- Rosaria A Cavallaro
- Department of Surgery "P. Valdoni", Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
| | - Vincenzina Nicolia
- Department of Surgery "P. Valdoni", Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
| | - Maria Teresa Fiorenza
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00183 Rome, Italy.
- IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64-65, 00143 Rome, Italy.
| | - Sigfrido Scarpa
- Department of Surgery "P. Valdoni", Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
| | - Andrea Fuso
- Department of Surgery "P. Valdoni", Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00183 Rome, Italy.
| |
Collapse
|
106
|
Jang JY, Cho H, Park HY, Rhim H, Kang S. ALS-linked mutant SOD1 proteins promote Aβ aggregates in ALS through direct interaction with Aβ. Biochem Biophys Res Commun 2017; 493:697-707. [PMID: 28864422 DOI: 10.1016/j.bbrc.2017.08.127] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 08/28/2017] [Indexed: 12/16/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive degeneration of motor neurons. Aggregation of ALS-linked mutant Cu/Zn superoxide dismutase (SOD1) is a hallmark of a subset of familial ALS (fALS). Recently, intracellular amyloid-β (Aβ) is detected in motor neurons of both sporadic and familial ALS. We have previously shown that intracellular Aβ specifically interacts with G93A, an ALS-linked SOD1 mutant. However, little is known about the pathological and biological effect of this interaction in neurons. In this study, we have demonstrated that the Aβ-binding region is exposed on the SOD1 surface through the conformational changes due to misfolding of SOD1. Interestingly, we found that the intracellular aggregation of Aβ is enhanced through the direct interaction of Aβ with the Aβ-binding region exposed to misfolded SOD1. Ultimately, increased Aβ aggregation by this interaction promotes neuronal cell death. Consistent with this result, Aβ aggregates was three-fold higher in the brains of G93A transgenic mice than those of non Tg. Our study provides the first direct evidence that Aβ, an AD-linked factor, is associated to the pathogenesis of ALS and provides molecular clues to understand common aggregation mechanisms in the pathogenesis of neurodegenerative diseases. Furthermore, it will provide new insights into the development of therapeutic approaches for ALS.
Collapse
Affiliation(s)
- Ja-Young Jang
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Hyungmin Cho
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Hye-Yoon Park
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Hyangshuk Rhim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Seongman Kang
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
107
|
Dietary Modulation of Oxidative Stress in Alzheimer's Disease. Int J Mol Sci 2017; 18:ijms18071583. [PMID: 28753984 PMCID: PMC5536070 DOI: 10.3390/ijms18071583] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/03/2017] [Accepted: 07/12/2017] [Indexed: 02/07/2023] Open
Abstract
Cells generate unpaired electrons, typically via oxygen- or nitrogen-based by-products during normal cellular respiration and under stressed situations. These pro-oxidant molecules are highly unstable and may oxidize surrounding cellular macromolecules. Under normal conditions, the reactive oxygen or nitrogen species can be beneficial to cell survival and function by destroying and degrading pathogens or antigens. However, excessive generation and accumulation of the reactive pro-oxidant species over time can damage proteins, lipids, carbohydrates, and nucleic acids. Over time, this oxidative stress can contribute to a range of aging-related degenerative diseases such as cancer, diabetes, macular degeneration, and Alzheimer’s, and Parkinson’s diseases. It is well accepted that natural compounds, including vitamins A, C, and E, β-carotene, and minerals found in fruits and vegetables are powerful anti-oxidants that offer health benefits against several different oxidative stress induced degenerative diseases, including Alzheimer’s disease (AD). There is increasing interest in developing anti-oxidative therapeutics to prevent AD. There are contradictory and inconsistent reports on the possible benefits of anti-oxidative supplements; however, fruits and vegetables enriched with multiple anti-oxidants (e.g., flavonoids and polyphenols) and minerals may be highly effective in attenuating the harmful effects of oxidative stress. As the physiological activation of either protective or destructive pro-oxidant behavior remains relatively unclear, it is not straightforward to relate the efficacy of dietary anti-oxidants in disease prevention. Here, we review oxidative stress mediated toxicity associated with AD and highlight the modulatory roles of natural dietary anti-oxidants in preventing AD.
Collapse
|
108
|
Ponomarenko P, Chadaeva I, Rasskazov DA, Sharypova E, Kashina EV, Drachkova I, Zhechev D, Ponomarenko MP, Savinkova LK, Kolchanov N. Candidate SNP Markers of Familial and Sporadic Alzheimer's Diseases Are Predicted by a Significant Change in the Affinity of TATA-Binding Protein for Human Gene Promoters. Front Aging Neurosci 2017; 9:231. [PMID: 28775688 PMCID: PMC5517495 DOI: 10.3389/fnagi.2017.00231] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 07/04/2017] [Indexed: 12/14/2022] Open
Abstract
While year after year, conditions, quality, and duration of human lives have been improving due to the progress in science, technology, education, and medicine, only eight diseases have been increasing in prevalence and shortening human lives because of premature deaths according to the retrospective official review on the state of US health, 1990-2010. These diseases are kidney cancer, chronic kidney diseases, liver cancer, diabetes, drug addiction, poisoning cases, consequences of falls, and Alzheimer's disease (AD) as one of the leading pathologies. There are familial AD of hereditary nature (~4% of cases) and sporadic AD of unclear etiology (remaining ~96% of cases; i.e., non-familial AD). Therefore, sporadic AD is no longer a purely medical problem, but rather a social challenge when someone asks oneself: “What can I do in my own adulthood to reduce the risk of sporadic AD at my old age to save the years of my lifespan from the destruction caused by it?” Here, we combine two computational approaches for regulatory SNPs: Web service SNP_TATA_Comparator for sequence analysis and a PubMed-based keyword search for articles on the biochemical markers of diseases. Our purpose was to try to find answers to the question: “What can be done in adulthood to reduce the risk of sporadic AD in old age to prevent the lifespan reduction caused by it?” As a result, we found 89 candidate SNP markers of familial and sporadic AD (e.g., rs562962093 is associated with sporadic AD in the elderly as a complication of stroke in adulthood, where natural marine diets can reduce risks of both diseases in case of the minor allele of this SNP). In addition, rs768454929, and rs761695685 correlate with sporadic AD as a comorbidity of short stature, where maximizing stature in childhood and adolescence as an integral indicator of health can minimize (or even eliminate) the risk of sporadic AD in the elderly. After validation by clinical protocols, these candidate SNP markers may become interesting to the general population [may help to choose a lifestyle (in childhood, adolescence, and adulthood) that can reduce the risks of sporadic AD, its comorbidities, and complications in the elderly].
Collapse
Affiliation(s)
- Petr Ponomarenko
- Children's Hospital Los Angeles, University of Southern CaliforniaLos Angeles, CA, United States
| | - Irina Chadaeva
- Division for System Biology, Institute of Cytology and Genetics of Siberian Branch of Russian Academy of SciencesNovosibirsk, Russia.,Faculty of Natural Sciences, Novosibirsk State UniversityNovosibirsk, Russia
| | - Dmitry A Rasskazov
- Division for System Biology, Institute of Cytology and Genetics of Siberian Branch of Russian Academy of SciencesNovosibirsk, Russia
| | - Ekaterina Sharypova
- Division for System Biology, Institute of Cytology and Genetics of Siberian Branch of Russian Academy of SciencesNovosibirsk, Russia
| | - Elena V Kashina
- Division for System Biology, Institute of Cytology and Genetics of Siberian Branch of Russian Academy of SciencesNovosibirsk, Russia
| | - Irina Drachkova
- Division for System Biology, Institute of Cytology and Genetics of Siberian Branch of Russian Academy of SciencesNovosibirsk, Russia
| | - Dmitry Zhechev
- Division for System Biology, Institute of Cytology and Genetics of Siberian Branch of Russian Academy of SciencesNovosibirsk, Russia
| | - Mikhail P Ponomarenko
- Division for System Biology, Institute of Cytology and Genetics of Siberian Branch of Russian Academy of SciencesNovosibirsk, Russia.,Faculty of Natural Sciences, Novosibirsk State UniversityNovosibirsk, Russia
| | - Ludmila K Savinkova
- Division for System Biology, Institute of Cytology and Genetics of Siberian Branch of Russian Academy of SciencesNovosibirsk, Russia
| | - Nikolay Kolchanov
- Division for System Biology, Institute of Cytology and Genetics of Siberian Branch of Russian Academy of SciencesNovosibirsk, Russia.,Faculty of Natural Sciences, Novosibirsk State UniversityNovosibirsk, Russia
| |
Collapse
|
109
|
Arbo BD, Hoppe JB, Rodrigues K, Garcia-Segura LM, Salbego CG, Ribeiro MF. 4'-Chlorodiazepam is neuroprotective against amyloid-beta in organotypic hippocampal cultures. J Steroid Biochem Mol Biol 2017; 171:281-287. [PMID: 28442392 DOI: 10.1016/j.jsbmb.2017.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 02/23/2017] [Accepted: 04/19/2017] [Indexed: 01/15/2023]
Abstract
The translocator protein (TSPO) is an outer mitochondrial membrane protein involved in the transport of cholesterol into the mitochondria, which is the first step for the synthesis of steroid hormones, as well as in the regulation of mitochondrial permeability transition pore opening and apoptosis. Studies have shown that the activation of TSPO may promote neuroprotective actions in experimental models of neurodegeneration and brain injury. In a previous study, our group showed that 4'-chlorodiazepam (4'-CD), a TSPO ligand, was neuroprotective against amyloid-beta (Aβ) in SHSY-5Y neuroblastoma cells. The aim of this study was to evaluate if 4'-CD was also neuroprotective against Aβ in organotypic hippocampal cultures and to identify its mechanisms of action. Aβ decreased the cell viability of organotypic hippocampal cultures, while 4'-CD had a neuroprotective effect when administered at 100nM and 1000nM. The neuroprotective effects of 4'-CD against Aβ were associated with an increased expression of superoxide dismutase (SOD). No differences were found in the expression of catalase, glial fibrillary acidic protein, Akt and procaspase-3. In summary, our results show that 4'-CD is neuroprotective against Aβ by a mechanism involving the modulation of SOD protein expression.
Collapse
Affiliation(s)
- B D Arbo
- Laboratório de Interação Neuro-Humoral - Department of Physiology - ICBS - Universidade Federal do Rio Grande do Sul (UFRGS), Rua Sarmento Leite, 500, 90050-170, Porto Alegre, RS, Brazil.
| | - J B Hoppe
- Laboratório de Neuroproteção e Sinalização Celular - Department of Biochemistry - Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil
| | - K Rodrigues
- Laboratório de Neuroproteção e Sinalização Celular - Department of Biochemistry - Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil
| | - L M Garcia-Segura
- Instituto Cajal - CSIC, Avenida Doctor Arce, 37, 28002, Madrid, Spain
| | - C G Salbego
- Laboratório de Neuroproteção e Sinalização Celular - Department of Biochemistry - Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, 90035-003, Porto Alegre, RS, Brazil
| | - M F Ribeiro
- Laboratório de Interação Neuro-Humoral - Department of Physiology - ICBS - Universidade Federal do Rio Grande do Sul (UFRGS), Rua Sarmento Leite, 500, 90050-170, Porto Alegre, RS, Brazil
| |
Collapse
|
110
|
Shibuya S, Sakaguchi I, Ito S, Kato E, Watanabe K, Izuo N, Shimizu T. Topical Application of Trisodium Ascorbyl 6-Palmitate 2-Phosphate Actively Supplies Ascorbate to Skin Cells in an Ascorbate Transporter-Independent Manner. Nutrients 2017. [PMID: 28640219 PMCID: PMC5537765 DOI: 10.3390/nu9070645] [Citation(s) in RCA: 8] [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] [Indexed: 12/14/2022] Open
Abstract
Ascorbic acid (AA) possesses multiple beneficial functions, such as regulating collagen biosynthesis and redox balance in the skin. AA derivatives have been developed to overcome this compound’s high fragility and to assist with AA supplementation to the skin. However, how AA derivatives are transferred into cells and converted to AA in the skin remains unclear. In the present study, we showed that AA treatment failed to increase the cellular AA level in the presence of AA transporter inhibitors, indicating an AA transporter-dependent action. In contrast, torisodium ascorbyl 6-palmitate 2-phosphate (APPS) treatment significantly enhanced the cellular AA level in skin cells despite the presence of inhibitors. In ex vivo experiments, APPS treatment also increased the AA content in a human epidermis model. Interestingly, APPS was readily metabolized and converted to AA in keratinocyte lysates via an intrinsic mechanism. Furthermore, APPS markedly repressed the intracellular superoxide generation and promoted viability associated with an enhanced AA level in Sod1-deficient skin cells. These findings indicate that APPS effectively restores the AA level and normalizes the redox balance in skin cells in an AA transporter-independent manner. Topical treatment of APPS is a beneficial strategy for supplying AA and improving the physiology of damaged skin.
Collapse
Affiliation(s)
- Shuichi Shibuya
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba 260-8670, Japan.
| | - Ikuyo Sakaguchi
- Reserch & Development Division, Club Cosmetics Co., Ltd., Ikoma, Nara 630-0222, Japan.
| | - Shintaro Ito
- Reserch & Development Division, Club Cosmetics Co., Ltd., Ikoma, Nara 630-0222, Japan.
| | - Eiko Kato
- Functional Chemicals Division, Showa Denko K.K. Minato-ku, Tokyo 105-8518, Japan.
| | - Kenji Watanabe
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba 260-8670, Japan.
| | - Naotaka Izuo
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba 260-8670, Japan.
| | - Takahiko Shimizu
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba 260-8670, Japan.
| |
Collapse
|
111
|
Oku Y, Murakami K, Irie K, Hoseki J, Sakai Y. Synthesized Aβ42 Caused Intracellular Oxidative Damage, Leading to Cell Death, via Lysosome Rupture. Cell Struct Funct 2017; 42:71-79. [PMID: 28413178 DOI: 10.1247/csf.17006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Neuronal cellular accumulation of amyloid beta peptide (Aβ) has been implicated in the pathogenesis of Alzheimer's disease (AD). Intracellular accumulation of Aβ42, a toxic form of Aβ, was observed as an early event in AD patients. However, its contribution and the cellular mechanism of cell death remained unclear. We herein revealed the mechanism by which Aβ42 incorporated into cells leads to cell death by using chemically synthesized Aβ42 variants. The Aβ42 variant Aβ42 (E22P) which has an increased tendency to oligomerize, accumulated in lysosomes at an earlier stage than wild-type Aβ42, leading to higher ROS production and lysosomal membrane oxidation, and resulting in cell death. On the other hand, Aβ42 (E22V), which is incapable of oligomerization, did not accumulate in cells or affect the cell viability. Moreover, intracellular localization of EGFP-Galectin-3, a β-galactoside binding lectin, showed that accumulation of oligomerized Aβ42 in lysosomes caused lysosomal membrane permeabilization (LMP). Overexpression of lysosome-localized LAMP1-fused peroxiredoxin 1 and treatment with U18866A, an inhibitor of cholesterol export from lysosomes that causes an increase in lysosomal membrane stability, attenuated Aβ42-mediated LMP and cell death. Our findings show that lysosomal ROS generation by toxic conformer of Aβ led to cell death via LMP, and suggest that these events are potential targets for AD prevention.Key words: Amyloid-beta (Aβ), Cell death, Lysosome, Lysosomal membrane permeabilization, Reactive oxygen species (ROS).
Collapse
Affiliation(s)
- Yuki Oku
- Graduate School of Advanced Integrated Studies, Kyoto University
| | - Kazuma Murakami
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University.,Research Unit for Physiological Chemistry, Kyoto University
| | - Kazuhiro Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University.,Research Unit for Physiological Chemistry, Kyoto University
| | - Jun Hoseki
- Research Unit for Physiological Chemistry, Kyoto University.,Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University
| | - Yasuyoshi Sakai
- Research Unit for Physiological Chemistry, Kyoto University.,Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University
| |
Collapse
|
112
|
Yang R, Wei L, Fu QQ, You H, Yu HR. SOD3 Ameliorates Aβ 25-35-Induced Oxidative Damage in SH-SY5Y Cells by Inhibiting the Mitochondrial Pathway. Cell Mol Neurobiol 2017; 37:513-525. [PMID: 27272114 DOI: 10.1007/s10571-016-0390-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 05/30/2016] [Indexed: 02/06/2023]
Abstract
This study was designed to investigate the protective effects of extracellular superoxide dismutase (SOD3) against amyloid beta (Aβ25-35)-induced damage in human neuroblastoma SH-SY5Y cells and to elucidate the mechanisms responsible for this beneficial effect. SH-SY5Y cells overexpressing SOD3 were generated by adenoviral vector-mediated infection and Aβ25-35 was then added to the cell culture system to establish an in vitro model of oxidative stress. Cell viability, the generation of intracellular reactive oxygen species (ROS), the expression and activity of antioxidant enzymes, the levels of lipid peroxidation malondialdehyde (MDA), the expression of mitochondrial apoptosis-related genes and calcium images were examined. Following Aβ25-35 exposure, SOD3 overexpression promoted the survival of SH-SY5Y cells, decreased the production of ROS, decreased MDA and calcium levels, and decreased cytochrome c, caspase-3, caspase-9 and Bax gene expression. Furthermore, SOD3 overexpression increased the expression and activity of antioxidant enzyme genes and Bcl-2 expression. Together, our data demonstrate that SOD3 ameliorates Aβ25-35-induced oxidative damage in neuroblastoma SH-SY5Y cells by inhibiting the mitochondrial pathway. These data provide new insights into the functional actions of SOD3 on oxidative stress-induced cell damage.
Collapse
Affiliation(s)
- Rong Yang
- Research Center of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
| | - Li Wei
- Key Laboratory of Birth Defects and Reproductive Health of the National Health and Family Planning Commission, Chongqing Population and Family Planning Science and Technology Research Institute, Chongqing, 400020, China
| | - Qing-Qing Fu
- Research Center of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
| | - Hua You
- Affiliated Hospital of the Academy of Military Medical Sciences, Beijing, 100071, China.
| | - Hua-Rong Yu
- Research Center of Neuroscience, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
113
|
Fang WL, Zhao DQ, Wang F, Li M, Fan SN, Liao W, Zheng YQ, Liao SW, Xiao SH, Luan P, Liu J. Neurotropin® alleviates hippocampal neuron damage through a HIF-1α/MAPK pathway. CNS Neurosci Ther 2017; 23:428-437. [PMID: 28271615 DOI: 10.1111/cns.12689] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/09/2017] [Accepted: 02/09/2017] [Indexed: 01/13/2023] Open
Abstract
AIMS The main purpose was to verify the potent capacity of Neurotropin® against neuronal damage in hippocampus and to explore its underlying mechanisms. METHODS HT22 cells were treated with 40 μmol/L Aβ25-35 in the presence of various concentrations of Neurotropin® or in its absence. The cell viability was assessed with a CCK-8 assay, and flow cytometry was used to measure cell apoptosis, intracellular ROS levels, and mitochondrial membrane potential. Aβ plaques were examined by Bielschowsky silver staining, and the activities of antioxidants were detected in hippocampus of APP/PS1 mice after Neurotropin® treatment. The expression of proteins, including HIF-1α, Bcl-2, Bax, and MAPKs signaling molecules was evaluated by Western blot. RESULTS Neurotropin® significantly reversed the cell injury induced by Aβ25-35 through increasing cell viability and mitochondrial membrane potential, decreasing intracellular ROS and cell apoptosis of HT22 cells (P<.05). Furthermore, Neurotropin® markedly reduced the formation of Aβ plaques and upregulated the activities of antioxidants (P<.05). Additionally, the protein expression of HIF-1α, p-ERK1/2, p-JNK, and p-P38 was significantly inhibited in hippocampus of APP/PS1 mice. CONCLUSIONS Neurotropin® exhibited a potent neuroprotective effect on inhibiting Aβ-induced oxidative damage and alleviating Aβ deposition in hippocampus via modulation of HIF-1α/MAPK signaling pathway.
Collapse
Affiliation(s)
- Wen-Li Fang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - De-Qiang Zhao
- Department of Neurology, Nanfang Hospital Huiqiao Medical Center, Guangzhou, Guangdong, China
| | - Fei Wang
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mei Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Sheng-Nuo Fan
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wang Liao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yu-Qiu Zheng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shao-Wei Liao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Song-Hua Xiao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ping Luan
- Medicine School, Shenzhen University, Shenzhen, Guangdong, China
| | - Jun Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Laboratory of RNA and Major Diseases of Brain and Heart, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
114
|
Choi H, Kim HJ, Kim J, Kim S, Yang J, Lee W, Park Y, Hyeon SJ, Lee DS, Ryu H, Chung J, Mook-Jung I. Increased acetylation of Peroxiredoxin1 by HDAC6 inhibition leads to recovery of Aβ-induced impaired axonal transport. Mol Neurodegener 2017; 12:23. [PMID: 28241840 PMCID: PMC5330132 DOI: 10.1186/s13024-017-0164-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 02/22/2017] [Indexed: 02/07/2023] Open
Abstract
Background Reduction or inhibition of histone deacetylase 6 (HDAC6) has been shown to rescue memory in mouse models of Alzheimer’s disease (AD) and is recently being considered a possible therapeutic strategy. However, the restoring mechanism of HDAC6 inhibition has not been fully understood. Methods and results Here, we found that an anti-oxidant protein Peroxdiredoxin1 (Prx1), a substrate of HDAC6, malfunctions in Aβ treated cells, the brains of 5xFAD AD model mice and AD patients. Malfunctioning Prx1, caused by reduced Prx1 acetylation levels, was recovered by HDAC6 inhibition. Increasing acetylation levels of Prx1 by HDAC6 inhibition recovered elevated reactive oxygen species (ROS) levels, elevated Ca2+ levels and impaired mitochondrial axonal transport, sequentially, even in the presence of Aβ. Prx1 mutant studies on the K197 site for an acetylation mimic or silencing mutation support the results showing that HDAC6 inhibitor restores Aβ-induced disruption of ROS, Ca2+ and axonal transport. Conclusions Taken together, increasing acetylation of Prx1 by HDAC6 inhibition has several beneficial effects in AD pathology. Here, we present the novel mechanism by which elevated acetylation of Prx1 rescues mitochondrial axonal transport impaired by Aβ. Therefore, our results suggest that modulation of Prx1 acetylation by HDAC6 inhibition has great therapeutic potential for AD and has further therapeutic possibilities for other neurodegenerative diseases as well. Electronic supplementary material The online version of this article (doi:10.1186/s13024-017-0164-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Heesun Choi
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Korea
| | - Haeng Jun Kim
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Korea
| | - Jisoo Kim
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Korea
| | - Soohyun Kim
- Department of Biochemistry and Molecular Biology, Seoul National University, College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jinhee Yang
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Korea
| | - Wonik Lee
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Korea
| | - Yeonju Park
- Department of Biomedical Sciences, Laboratory of Immunology and Cancer Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Jae Hyeon
- Center for Neuromedicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Korea
| | - Dong-Sup Lee
- Department of Biomedical Sciences, Laboratory of Immunology and Cancer Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Hoon Ryu
- VA Boston Healthcare System, Boston University Alzheimer's Disease Center, and Department of Neurology, Boston University School of Medicine, Boston, MA02130, USA.,Center for Neuromedicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Korea
| | - Junho Chung
- Department of Biochemistry and Molecular Biology, Seoul National University, College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Inhee Mook-Jung
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Korea.
| |
Collapse
|
115
|
Pickart L, Vasquez-Soltero JM, Margolina A. The Effect of the Human Peptide GHK on Gene Expression Relevant to Nervous System Function and Cognitive Decline. Brain Sci 2017; 7:E20. [PMID: 28212278 PMCID: PMC5332963 DOI: 10.3390/brainsci7020020] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/07/2017] [Accepted: 02/08/2017] [Indexed: 12/25/2022] Open
Abstract
Neurodegeneration, the progressive death of neurons, loss of brain function, and cognitive decline is an increasing problem for senior populations. Its causes are poorly understood and therapies are largely ineffective. Neurons, with high energy and oxygen requirements, are especially vulnerable to detrimental factors, including age-related dysregulation of biochemical pathways caused by altered expression of multiple genes. GHK (glycyl-l-histidyl-l-lysine) is a human copper-binding peptide with biological actions that appear to counter aging-associated diseases and conditions. GHK, which declines with age, has health promoting effects on many tissues such as chondrocytes, liver cells and human fibroblasts, improves wound healing and tissue regeneration (skin, hair follicles, stomach and intestinal linings, boney tissue), increases collagen, decorin, angiogenesis, and nerve outgrowth, possesses anti-oxidant, anti-inflammatory, anti-pain and anti-anxiety effects, increases cellular stemness and the secretion of trophic factors by mesenchymal stem cells. Studies using the Broad Institute Connectivity Map show that GHK peptide modulates expression of multiple genes, resetting pathological gene expression patterns back to health. GHK has been recommended as a treatment for metastatic cancer, Chronic Obstructive Lung Disease, inflammation, acute lung injury, activating stem cells, pain, and anxiety. Here, we present GHK's effects on gene expression relevant to the nervous system health and function.
Collapse
Affiliation(s)
- Loren Pickart
- Research & Development Department, Skin Biology, 4122 Factoria Boulevard SE Suite No. 200 Bellevue, WA 98006, USA.
| | | | - Anna Margolina
- Research & Development Department, Skin Biology, 4122 Factoria Boulevard SE Suite No. 200 Bellevue, WA 98006, USA.
| |
Collapse
|
116
|
Pate KM, Rogers M, Reed JW, van der Munnik N, Vance SZ, Moss MA. Anthoxanthin Polyphenols Attenuate Aβ Oligomer-induced Neuronal Responses Associated with Alzheimer's Disease. CNS Neurosci Ther 2017; 23:135-144. [PMID: 27864869 PMCID: PMC5239747 DOI: 10.1111/cns.12659] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/27/2016] [Accepted: 10/17/2016] [Indexed: 12/27/2022] Open
Abstract
AIMS Epidemiological evidence implicates polyphenols as potential natural therapeutics for Alzheimer's disease (AD). To investigate this prospect, five anthoxanthin polyphenols were characterized for their ability to reduce amyloid-β (Aβ) oligomer-induced neuronal responses by two mechanisms of action, modulation of oligomerization and antioxidant activity, as well as the synergy between these two mechanisms. METHODS Anthoxanthin oligomerization modulation and antioxidant capabilities were evaluated and correlated with anthoxanthin attenuation of oligomer-induced intracellular reactive oxygen species (ROS) and caspase activation using human neuroblastoma cell treatments designed to isolate these mechanisms of action and to achieve dual-action. RESULTS While modulation of oligomerization resulted in only minor reductions to neuronal responses, anthoxanthin antioxidant action significantly attenuated oligomer-induced intracellular ROS and caspase activation. Kaempferol uniquely exhibited synergism when the two mechanisms functioned in concert, leading to a pronounced reduction in both ROS and caspase activation. CONCLUSIONS Together, these findings identify the dominant mechanism by which these anthoxanthins attenuate Aβ oligomer-induced neuronal responses, elucidate their prospective synergy, and demonstrate the potential of anthoxanthin polyphenols as natural AD therapeutics.
Collapse
Affiliation(s)
- Kayla M. Pate
- Department of Chemical EngineeringUniversity of South CarolinaColumbiaSCUSA
- Present address:
Department of Chemical and Biological EngineeringUniversity of WisconsinMadisonWI53706USA
| | - McCall Rogers
- Department of Chemical EngineeringUniversity of South CarolinaColumbiaSCUSA
| | - John Will Reed
- Department of Chemical EngineeringUniversity of South CarolinaColumbiaSCUSA
- Present address:
Data AnalyticsThe Home DepotAtlantaGA30318USA
| | | | | | - Melissa A. Moss
- Department of Chemical EngineeringUniversity of South CarolinaColumbiaSCUSA
- Biomedical Engineering ProgramUniversity of South CarolinaColumbiaSCUSA
| |
Collapse
|
117
|
Kountouras J, Boziki M, Polyzos SA, Katsinelos P, Gavalas E, Zeglinas C, Tzivras D, Romiopoulos I, Giorgakis N, Anastasiadou K, Vardaka E, Kountouras C, Kazakos E, Xiromerisiou G, Dardiotis E, Deretzi G. Impact of reactive oxygen species generation on Helicobacter pylori-related extragastric diseases: a hypothesis. Free Radic Res 2017; 51:73-79. [DOI: 10.1080/10715762.2016.1271122] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Jannis Kountouras
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Marina Boziki
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Stergios A. Polyzos
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Panagiotis Katsinelos
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Emmanouel Gavalas
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Christos Zeglinas
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Dimitri Tzivras
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Iordanis Romiopoulos
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Nikolaos Giorgakis
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Kyriaki Anastasiadou
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Elizabeth Vardaka
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Constantinos Kountouras
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Evangelos Kazakos
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Georgia Xiromerisiou
- Department of Neurology, Multiple Sclerosis Unit, Papageorgiou General Hospital, Thessaloniki, Greece
| | - Efthimios Dardiotis
- Laboratory of Neurogenetics, Department of Neurology, University of Thessaly, University Hospital of Larissa, Greece
| | - Georgia Deretzi
- Department of Neurology, Multiple Sclerosis Unit, Papageorgiou General Hospital, Thessaloniki, Greece
| |
Collapse
|
118
|
Kondo Y, Ishigami A. Involvement of senescence marker protein-30 in glucose metabolism disorder and non-alcoholic fatty liver disease. Geriatr Gerontol Int 2017; 16 Suppl 1:4-16. [PMID: 27018279 DOI: 10.1111/ggi.12722] [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] [Accepted: 11/27/2015] [Indexed: 12/16/2022]
Abstract
Senescence marker protein-30 (SMP30) was found to decrease in the liver, kidneys and lungs of mice during aging. SMP30 is a pleiotropic protein that acts to protect cells from apoptosis by enhancing plasma membrane Ca(2+) -pump activity and is bona fide gluconolactonase (EC 3.1.1.17) that participates in the penultimate step of the vitamin C biosynthetic pathway. For the past several years, we have obtained strong evidence showing the close relationship between SMP30, glucose metabolism disorder and non-alchoholic fatty liver disease in experiments with SMP30 knockout mice. Emerging proof links the following abnormalities: (i) the reduction of SMP30 by aging and/or excessive dietary fat or genetic deficiency causes a loss of Ca(2+) pumping activity, which impairs acute insulin release in pancreatic β-cells, initiates inflammatory responses with oxidative stress and endoplasmic reticulum stress in non-alchoholic steatohepatitis, exacerbates renal tubule damage, and introduces tubulointerstitial inflammation and fibrosis in diabetic nephropathy; (ii) vitamin C insufficiency also impairs acute insulin secretion in pancreatic β-cells by a mechanism distinct from that of the SMP30 deficiency; and (iii) the increased oxidative stress by concomitant deficiencies of SMP30, superoxide dismutase 1 and vitamin C similarly causes hepatic steatosis. Here, we review recent advances in our understanding of SMP30 in glucose metabolism disorder and non-alchoholic fatty liver disease.
Collapse
Affiliation(s)
- Yoshitaka Kondo
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
119
|
Kaliszewski M, Kennedy AK, Blaes SL, Shaffer RS, Knott AB, Song W, Hauser HA, Bossy B, Huang TT, Bossy-Wetzel E. SOD1 Lysine 123 Acetylation in the Adult Central Nervous System. Front Cell Neurosci 2016; 10:287. [PMID: 28066183 PMCID: PMC5167747 DOI: 10.3389/fncel.2016.00287] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 12/01/2016] [Indexed: 01/09/2023] Open
Abstract
Superoxide dismutase 1 (SOD1) knockout (Sod1−/−) mice exhibit an accelerated aging phenotype. In humans, SOD1 mutations are linked to familial amyotrophic lateral sclerosis (ALS), and post-translational modification (PTM) of wild-type SOD1 has been associated with sporadic ALS. Reversible acetylation regulates many enzymes and proteomic studies have identified SOD1 acetylation at lysine 123 (K123). The function and distribution of K123-acetylated SOD1 (Ac-K123 SOD1) in the nervous system is unknown. Here, we generated polyclonal rabbit antibodies against Ac-K123 SOD1. Sod1 deletion in Sod1−/− mice, K123 mutation or preabsorption with Ac-K123 peptide all abolished antibody binding. Using immunohistochemistry, we assessed Ac-K123 SOD1 distribution in the normal adult mouse nervous system. In the cerebellum, Ac-K123 SOD1 staining was prominent in cell bodies of the granular cell layer (GCL) and Purkinje cell dendrites and interneurons of the molecular cell layer. In the hippocampus, Ac-K123 SOD1 staining was strong in the fimbria, subiculum, pyramidal cells and Schaffer collateral fibers of the cornus ammonis field 1 (CA1) region and granule and neuronal progenitor cells of the dentate gyrus. In addition, labeling was observed in the choroid plexus (CP) and the ependyma of the brain ventricles and central canal of the spinal cord. In the olfactory bulb, Ac-K123 SOD1 staining was prominent in axons of sensory neurons, in cell bodies of interneurons and neurites of the mitral and tufted cells. In the retina, labeling was strong in the retinal ganglion cell layer (RGCL) and axons of retinal ganglion cells (RGCs), the inner nuclear layer (INL) and cone photoreceptors of the outer nuclear layer (ONL). In summary, our findings describe Ac-K123 SOD1 distribution to distinct regions and cell types of the normal nervous system.
Collapse
Affiliation(s)
- Michael Kaliszewski
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida Orlando, FL, USA
| | - Austin K Kennedy
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida Orlando, FL, USA
| | - Shelby L Blaes
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida Orlando, FL, USA
| | - Robert S Shaffer
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida Orlando, FL, USA
| | - Andrew B Knott
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida Orlando, FL, USA
| | - Wenjun Song
- Burnett School of Biomedical Sciences, College of Medicine, University of Central FloridaOrlando, FL, USA; Yale School of Forestry and Environmental Studies, Yale UniversityNew Haven, CT, USA
| | - Henry A Hauser
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida Orlando, FL, USA
| | - Blaise Bossy
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida Orlando, FL, USA
| | - Ting-Ting Huang
- Department of Neurology and Neurological Sciences, Stanford University School of MedicineStanford, CA, USA; Geriatric Research, Education, and Clinical Center, VA Palo Alto Health Care SystemPalo Alto, CA, USA
| | - Ella Bossy-Wetzel
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida Orlando, FL, USA
| |
Collapse
|
120
|
Yoshihara D, Fujiwara N, Kitanaka N, Kitanaka J, Sakiyama H, Eguchi H, Takemura M, Suzuki K. The absence of the SOD1 gene causes abnormal monoaminergic neurotransmission and motivational impairment-like behavior in mice. Free Radic Res 2016; 50:1245-1256. [PMID: 27629432 DOI: 10.1080/10715762.2016.1234048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Copper/zinc superoxide dismutase (SOD1), a primary anti-oxidative enzyme, protects cells against oxidative stress. We report herein on a comparison of behavioral and neurobiological changes between SOD1 knockout (KO) and wild-type mice, in an attempt to assess the role of SOD1 in brain functions. SOD1 KO mice exhibited impaired motivational behavior in both shuttle-box learning and three-chamber social interaction tests. High levels of dopamine transporter protein and an acceleration of serotonin turnover were also detected in the cerebrums of the SOD1 KO mice. These findings suggest that SOD1 deficiency disturbs monoaminergic neurotransmission leading to a decrease in motivational behavior.
Collapse
Affiliation(s)
- Daisaku Yoshihara
- a Department of Biochemistry , Hyogo College of Medicine , Nishinomiya , Japan
| | - Noriko Fujiwara
- a Department of Biochemistry , Hyogo College of Medicine , Nishinomiya , Japan
| | - Nobue Kitanaka
- b Department of Pharmacology , Hyogo College of Medicine , Nishinomiya , Japan
| | - Junichi Kitanaka
- b Department of Pharmacology , Hyogo College of Medicine , Nishinomiya , Japan
| | - Haruhiko Sakiyama
- a Department of Biochemistry , Hyogo College of Medicine , Nishinomiya , Japan
| | - Hironobu Eguchi
- a Department of Biochemistry , Hyogo College of Medicine , Nishinomiya , Japan
| | - Motohiko Takemura
- b Department of Pharmacology , Hyogo College of Medicine , Nishinomiya , Japan
| | - Keiichiro Suzuki
- a Department of Biochemistry , Hyogo College of Medicine , Nishinomiya , Japan
| |
Collapse
|
121
|
Chaves RS, Kazi AI, Silva CM, Almeida MF, Lima RS, Carrettiero DC, Demasi M, Ferrari MFR. Presence of insoluble Tau following rotenone exposure ameliorates basic pathways associated with neurodegeneration. IBRO Rep 2016; 1:32-45. [PMID: 30135926 PMCID: PMC6084878 DOI: 10.1016/j.ibror.2016.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/23/2016] [Accepted: 09/24/2016] [Indexed: 01/24/2023] Open
Abstract
Protein aggregation is an important feature of neurodegenerative disorders. In Alzheimer's disease (AD) protein aggregates are composed of hyperphosphorylated Tau and amyloid beta peptide (Aβ). Despite the involvement and identification of the molecular composition of these aggregates, their role in AD pathophysiology is not fully understood. However, depositions of these insoluble aggregates are typically reported as pathogenic and toxic for cell homeostasis. New evidences suggest that the deposition of these aggregates is a protective mechanism that preserves cell from toxic insults associated with the early stages of neurodegenerative diseases. To better understand the biological role of the protein aggregation with regard its effects in cellular homeostasis, the present study investigated the role of insoluble Tau and Tau aggregates on crucial cellular parameters such as redox homeostasis, proteasome activity and autophagy in hippocampal cell cultures and hippocampus of aged Lewis rats using a rotenone-induced aggregation model. Neurons were exposed to rotenone in different concentrations and exposure times aiming to determine the interval required for Tau aggregation. Our experimental design allowed us to demonstrate that rotenone exposure induces Tau hyperphosphorylation and aggregation in a concentration and time-dependent manner. Oxidative stress triggered by rotenone exposure was observed with the absence of Tau aggregates and was reduced or absent when Tau aggregates were present. This reduction of oxidative stress along with the presence of insoluble Tau was independent of alterations in antioxidant enzymes activities or cell death. In addition, rotenone induced oxidative stress was mainly associated with decrease in proteasome activity and autophagy flux. Conversely, when insoluble Tau appeared, autophagy turns to be overactivated while proteasome activity remained low. Our studies significantly advance the understanding that Tau aggregation might exert protective cellular effects, at least briefly, when neurons are facing neurodegeneration stimulus. We believe that our data add more complexity for the understanding of protein aggregation role in AD etiology.
Collapse
Affiliation(s)
- Rodrigo S Chaves
- Department of Genetics and Evolutionary Biology - Institute for Biosciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Amajad I Kazi
- Department of Genetics and Evolutionary Biology - Institute for Biosciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Carolliny M Silva
- Department of Genetics and Evolutionary Biology - Institute for Biosciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Michael F Almeida
- Department of Genetics and Evolutionary Biology - Institute for Biosciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Raquel S Lima
- Department of Genetics and Evolutionary Biology - Institute for Biosciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | | | - Marilene Demasi
- Laboratory of Biochemistry and Biophysics - Butantan Institute, Sao Paulo, SP, Brazil
| | - Merari F R Ferrari
- Department of Genetics and Evolutionary Biology - Institute for Biosciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
122
|
Hamadi N, Sheikh A, Madjid N, Lubbad L, Amir N, Shehab SADS, Khelifi-Touhami F, Adem A. Increased pro-inflammatory cytokines, glial activation and oxidative stress in the hippocampus after short-term bilateral adrenalectomy. BMC Neurosci 2016; 17:61. [PMID: 27586269 PMCID: PMC5009504 DOI: 10.1186/s12868-016-0296-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 08/25/2016] [Indexed: 12/22/2022] Open
Abstract
Background Bilateral adrenalectomy has been shown to damage the hippocampal neurons. Although the effects of long-term adrenalectomy have been studied extensively there are few publications on the effects of short-term adrenalectomy. In the present study we aimed to investigate the effects of short-term bilateral adrenalectomy on the levels of pro-inflammatory cytokines IL-1β, IL-6 and TNF-α; the response of microglia and astrocytes to neuronal cell death as well as oxidative stress markers GSH, SOD and MDA over the course of time (4 h, 24 h, 3 days, 1 week and 2 weeks) in the hippocampus of Wistar rats. Results Our results showed a transient significant elevation of pro-inflammatory cytokines IL-1β and IL-6 from 4 h to 3 days in the adrenalectomized compared to sham operated rats. After 1 week, the elevation of both cytokines returns to the sham levels. Surprisingly, TNF-α levels were significantly elevated at 4 h only in adrenalectomized compared to sham operated rats. The occurrence of neuronal cell death in the hippocampus following adrenalectomy was confirmed by Fluoro-Jade B staining. Our results showed a time dependent increase in degenerated neurons in the dorsal blade of the dentate gyrus from 3 days to 2 weeks after adrenalectomy. Our results revealed an early activation of microglia on day three whereas activation of astroglia in the hippocampus was observed at 1 week postoperatively. A progression of microglia and astroglia activation all over the dentate gyrus and their appearance for the first time in CA3 of adrenalectomized rats hippocampi compared to sham operated was seen after 2 weeks of surgery. Quantitative analysis revealed a significant increase in the number of microglia (3, 7 and 14 days) and astrocytes (7 and 14 days) of ADX compared to sham operated rats. Our study revealed no major signs of oxidative stress until 2 weeks after adrenalectomy when a significant decrease of GSH levels and SOD activity as well as an increase in MDA levels were found in adrenalectomized compared to sham rats. Conclusion Our study showed an early increase in the pro-inflammatory cytokines followed by neurodegeneration and activation of glial cells as well as oxidative stress. Taking these findings together it could be speculated that the early inflammatory components might contribute to the initiation of the biological cascade responsible for subsequent neuronal death in the current neurodegenerative animal model. These findings suggest that inflammatory mechanisms precede neurodegeneration and glial activation.
Collapse
Affiliation(s)
- Naserddine Hamadi
- Department of Pharmacology, College of Medicine and Health Science, United Arab Emirates University, 17666, Maqam, Al Ain, United Arab Emirates.,Ethnobotany-Palynology and Ethnopharmacology-Toxicology Laboratory, Department of Animal Biology, Constantine-1 University, 25000, Constantine, Algeria
| | - Azimullah Sheikh
- Department of Pharmacology, College of Medicine and Health Science, United Arab Emirates University, 17666, Maqam, Al Ain, United Arab Emirates
| | - Nather Madjid
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Loai Lubbad
- Department of Surgery, College of Medicine and Health Science, United Arab Emirates University, 17666, Maqam, Al Ain, United Arab Emirates
| | - Naheed Amir
- Department of Pharmacology, College of Medicine and Health Science, United Arab Emirates University, 17666, Maqam, Al Ain, United Arab Emirates
| | - Safa Al-Deen Saudi Shehab
- Department of Anatomy, College of Medicine and Health Science, United Arab Emirates University, 17666, Maqam, Al Ain, United Arab Emirates
| | - Fatima Khelifi-Touhami
- Ethnobotany-Palynology and Ethnopharmacology-Toxicology Laboratory, Department of Animal Biology, Constantine-1 University, 25000, Constantine, Algeria
| | - Abdu Adem
- Department of Pharmacology, College of Medicine and Health Science, United Arab Emirates University, 17666, Maqam, Al Ain, United Arab Emirates.
| |
Collapse
|
123
|
Bae JR, Kim SH. Impairment of SOD1-G93A motility is linked to mitochondrial movement in axons of hippocampal neurons. Arch Pharm Res 2016; 39:1144-50. [PMID: 27464601 DOI: 10.1007/s12272-016-0798-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 07/18/2016] [Indexed: 12/25/2022]
Abstract
Superoxide dismutase 1 (SOD1) is a well-known antioxidant enzyme. Mutation of SOD1 is closely associated with the pathogenesis of neurodegenerative disorders, such as amyotrophic lateral sclerosis and Alzheimer's disease. However, the pathologic pathways linking neurodegenerative diseases with mutation of SOD1 remain elusive. Here, we investigated the motility of SOD1-WT and -G93A (a pathogenic mutant of SOD1), and observed correlation of axonal transport of the mutant protein with mitochondria in primary cultured hippocampal neurons. The SOD1-G93A mutant showed significant accumulation at vGlut1-positive synaptic boutons and in cell bodies, compared to SOD1-WT. The proportions of motile WT and G93A proteins were similar (~30 %) while the motility velocity of SOD1-G93A was significantly slower (~40 %) than that of the WT counterpart. This motility defect of SOD1-G93A was highly correlated with mitochondrial movement. Our results collectively suggest that the SOD1-G93A mutant has a defect in motility that is linked to mitochondrial transport in axons.
Collapse
Affiliation(s)
- Jae Ryul Bae
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 130-701, South Korea
| | - Sung Hyun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 130-701, South Korea. .,Department of Physiology, School of Medicine, Neurodegeneration Control Research Center, Kyung Hee University, Seoul, 130-701, South Korea.
| |
Collapse
|
124
|
Lei XG, Zhu JH, Cheng WH, Bao Y, Ho YS, Reddi AR, Holmgren A, Arnér ESJ. Paradoxical Roles of Antioxidant Enzymes: Basic Mechanisms and Health Implications. Physiol Rev 2016; 96:307-64. [PMID: 26681794 DOI: 10.1152/physrev.00010.2014] [Citation(s) in RCA: 274] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are generated from aerobic metabolism, as a result of accidental electron leakage as well as regulated enzymatic processes. Because ROS/RNS can induce oxidative injury and act in redox signaling, enzymes metabolizing them will inherently promote either health or disease, depending on the physiological context. It is thus misleading to consider conventionally called antioxidant enzymes to be largely, if not exclusively, health protective. Because such a notion is nonetheless common, we herein attempt to rationalize why this simplistic view should be avoided. First we give an updated summary of physiological phenotypes triggered in mouse models of overexpression or knockout of major antioxidant enzymes. Subsequently, we focus on a series of striking cases that demonstrate "paradoxical" outcomes, i.e., increased fitness upon deletion of antioxidant enzymes or disease triggered by their overexpression. We elaborate mechanisms by which these phenotypes are mediated via chemical, biological, and metabolic interactions of the antioxidant enzymes with their substrates, downstream events, and cellular context. Furthermore, we propose that novel treatments of antioxidant enzyme-related human diseases may be enabled by deliberate targeting of dual roles of the pertaining enzymes. We also discuss the potential of "antioxidant" nutrients and phytochemicals, via regulating the expression or function of antioxidant enzymes, in preventing, treating, or aggravating chronic diseases. We conclude that "paradoxical" roles of antioxidant enzymes in physiology, health, and disease derive from sophisticated molecular mechanisms of redox biology and metabolic homeostasis. Simply viewing antioxidant enzymes as always being beneficial is not only conceptually misleading but also clinically hazardous if such notions underpin medical treatment protocols based on modulation of redox pathways.
Collapse
Affiliation(s)
- Xin Gen Lei
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jian-Hong Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Wen-Hsing Cheng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Yongping Bao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ye-Shih Ho
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Amit R Reddi
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Arne Holmgren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Elias S J Arnér
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing,China; Department of Animal Science, Cornell University, Ithaca, New York; Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Mississippi State, Mississippi; Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom; Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan; Georgia Institute of Technology, School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Atlanta, Georgia; and Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
125
|
Shi Y, Sun X, Sun Y, Hou L, Yao M, Lian K, Li J, Lu X, Jiang L. Elevation of cortical C26:0 due to the decline of peroxisomal β-oxidation potentiates amyloid β generation and spatial memory deficits via oxidative stress in diabetic rats. Neuroscience 2016; 315:125-35. [DOI: 10.1016/j.neuroscience.2015.11.067] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 11/07/2015] [Accepted: 11/30/2015] [Indexed: 01/23/2023]
|
126
|
The Therapeutic Potential of Rosemary (Rosmarinus officinalis) Diterpenes for Alzheimer's Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:2680409. [PMID: 26941822 PMCID: PMC4749867 DOI: 10.1155/2016/2680409] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 12/28/2015] [Indexed: 02/07/2023]
Abstract
Rosemary (Rosmarinus officinalis L.) is one of the most economically important species of the family Lamiaceae. Native to the Mediterranean region, the plant is now widely distributed all over the world mainly due to its culinary, medicinal, and commercial uses including in the fragrance and food industries. Among the most important group of compounds isolated from the plant are the abietane-type phenolic diterpenes that account for most of the antioxidant and many pharmacological activities of the plant. Rosemary diterpenes have also been shown in recent years to inhibit neuronal cell death induced by a variety of agents both in vitro and in vivo. The therapeutic potential of these compounds for Alzheimer's disease (AD) is reviewed in this communication by giving special attention to the chemistry of the compounds along with the various pharmacological targets of the disease. The multifunctional nature of the compounds from the general antioxidant-mediated neuronal protection to other specific mechanisms including brain inflammation and amyloid beta (Aβ) formation, polymerisation, and pathologies is discussed.
Collapse
|
127
|
Reybier K, Ayala S, Alies B, Rodrigues JV, Bustos Rodriguez S, La Penna G, Collin F, Gomes CM, Hureau C, Faller P. Free Superoxide is an Intermediate in the Production of H2O2 by Copper(I)-Aβ Peptide and O2. Angew Chem Int Ed Engl 2015; 55:1085-9. [PMID: 26629876 DOI: 10.1002/anie.201508597] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Indexed: 11/06/2022]
Abstract
Oxidative stress is considered as an important factor and an early event in the etiology of Alzheimer's disease (AD). Cu bound to the peptide amyloid-β (Aβ) is found in AD brains, and Cu-Aβ could contribute to this oxidative stress, as it is able to produce in vitro H2O2 and HO˙ in the presence of oxygen and biological reducing agents such as ascorbate. The mechanism of Cu-Aβ-catalyzed H2O2 production is however not known, although it was proposed that H2O2 is directly formed from O2 via a 2-electron process. Here, we implement an electrochemical setup and use the specificity of superoxide dismutase-1 (SOD1) to show, for the first time, that H2O2 production by Cu-Aβ in the presence of ascorbate occurs mainly via a free O2˙(-) intermediate. This finding radically changes the view on the catalytic mechanism of H2O2 production by Cu-Aβ, and opens the possibility that Cu-Aβ-catalyzed O2˙(-) contributes to oxidative stress in AD, and hence may be of interest.
Collapse
Affiliation(s)
- Karine Reybier
- University of Toulouse, UPS; UMR 152 PHARMA-DEV, 118 route de Narbonne, 31062, Toulouse cedex 9, France. .,IRD, UMR 152, 31062, Toulouse cedex 9, France.
| | - Sara Ayala
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099, 31077, Toulouse Cedex 4, France.,University of Toulouse, UPS, INPT, 31077, Toulouse Cedex 4, France
| | - Bruno Alies
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099, 31077, Toulouse Cedex 4, France.,University of Toulouse, UPS, INPT, 31077, Toulouse Cedex 4, France
| | - João V Rodrigues
- Instituto Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal.,Harvard University, Department of Chemistry and Chemical Biology, Cambridge, MA, USA
| | - Susana Bustos Rodriguez
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099, 31077, Toulouse Cedex 4, France.,University of Toulouse, UPS, INPT, 31077, Toulouse Cedex 4, France
| | - Giovanni La Penna
- CNR - National Research Council of Italy, ICCOM - Institute for Chemistry of Organo-Metallic Compounds, via Madonna del Piano 10, 50019 Sesto Fiorentino, Firenze, Italy
| | - Fabrice Collin
- University of Toulouse, UPS; UMR 152 PHARMA-DEV, 118 route de Narbonne, 31062, Toulouse cedex 9, France.,IRD, UMR 152, 31062, Toulouse cedex 9, France.,CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099, 31077, Toulouse Cedex 4, France.,University of Toulouse, UPS, INPT, 31077, Toulouse Cedex 4, France
| | - Cláudio M Gomes
- Instituto Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal.,Faculdade de Ciências, Biosystems and Integrative Sciences Institute, Department of Chemistry and Biochemistry, Universidade de Lisboa, Campo Grande, Lisboa, Portugal
| | - Christelle Hureau
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099, 31077, Toulouse Cedex 4, France.,University of Toulouse, UPS, INPT, 31077, Toulouse Cedex 4, France
| | - Peter Faller
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099, 31077, Toulouse Cedex 4, France. .,University of Toulouse, UPS, INPT, 31077, Toulouse Cedex 4, France. .,Institute de Chimie (UMR 7177), 4 rue B. Pascal, 67081, Strasbourg, France.
| |
Collapse
|
128
|
Reybier K, Ayala S, Alies B, Rodrigues JV, Bustos Rodriguez S, La Penna G, Collin F, Gomes CM, Hureau C, Faller P. Free Superoxide is an Intermediate in the Production of H
2
O
2
by Copper(I)‐Aβ Peptide and O
2. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201508597] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Karine Reybier
- University of Toulouse, UPS; UMR 152 PHARMA-DEV 118 route de Narbonne 31062 Toulouse cedex 9 France
- IRD, UMR 152 31062 Toulouse cedex 9 France
| | - Sara Ayala
- CNRS, LCC (Laboratoire de Chimie de Coordination) 205 route de Narbonne, BP 44099 31077 Toulouse Cedex 4 France
- University of Toulouse, UPS, INPT 31077 Toulouse Cedex 4 France
| | - Bruno Alies
- CNRS, LCC (Laboratoire de Chimie de Coordination) 205 route de Narbonne, BP 44099 31077 Toulouse Cedex 4 France
- University of Toulouse, UPS, INPT 31077 Toulouse Cedex 4 France
| | - João V. Rodrigues
- Instituto Tecnologia Química e Biológica Universidade Nova de Lisboa Oeiras Portugal
- Harvard University Department of Chemistry and Chemical Biology Cambridge MA USA
| | - Susana Bustos Rodriguez
- CNRS, LCC (Laboratoire de Chimie de Coordination) 205 route de Narbonne, BP 44099 31077 Toulouse Cedex 4 France
- University of Toulouse, UPS, INPT 31077 Toulouse Cedex 4 France
| | - Giovanni La Penna
- CNR – National Research Council of Italy ICCOM – Institute for Chemistry of Organo-Metallic Compounds via Madonna del Piano 10 50019 Sesto Fiorentino Firenze Italy
| | - Fabrice Collin
- University of Toulouse, UPS; UMR 152 PHARMA-DEV 118 route de Narbonne 31062 Toulouse cedex 9 France
- IRD, UMR 152 31062 Toulouse cedex 9 France
- CNRS, LCC (Laboratoire de Chimie de Coordination) 205 route de Narbonne, BP 44099 31077 Toulouse Cedex 4 France
- University of Toulouse, UPS, INPT 31077 Toulouse Cedex 4 France
| | - Cláudio M. Gomes
- Instituto Tecnologia Química e Biológica Universidade Nova de Lisboa Oeiras Portugal
- Faculdade de Ciências, Biosystems and Integrative Sciences Institute Department of Chemistry and Biochemistry Universidade de Lisboa, Campo Grande Lisboa Portugal
| | - Christelle Hureau
- CNRS, LCC (Laboratoire de Chimie de Coordination) 205 route de Narbonne, BP 44099 31077 Toulouse Cedex 4 France
- University of Toulouse, UPS, INPT 31077 Toulouse Cedex 4 France
| | - Peter Faller
- CNRS, LCC (Laboratoire de Chimie de Coordination) 205 route de Narbonne, BP 44099 31077 Toulouse Cedex 4 France
- University of Toulouse, UPS, INPT 31077 Toulouse Cedex 4 France
- Institute de Chimie (UMR 7177) 4 rue B. Pascal 67081 Strasbourg France
| |
Collapse
|
129
|
Muresan V, Ladescu Muresan Z. Shared Molecular Mechanisms in Alzheimer's Disease and Amyotrophic Lateral Sclerosis: Neurofilament-Dependent Transport of sAPP, FUS, TDP-43 and SOD1, with Endoplasmic Reticulum-Like Tubules. NEURODEGENER DIS 2015; 16:55-61. [PMID: 26605911 DOI: 10.1159/000439256] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/07/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS), a debilitating neurodegenerative disorder of the motor neurons, leads to the disorganization of the neurofilament (NF) cytoskeleton and - ultimately - the deterioration of the neuromuscular junction. Some familial cases of ALS are caused by mutated FUS, TDP-43 or SOD1; it is thought that the mutated proteins inflict pathology either by gain or loss of function. The proper function of the neuromuscular junction requires sAPP, a soluble proteolytic fragment of the amyloid-β precursor protein (APP) - a transmembrane protein implicated in the pathology of Alzheimer's disease (AD). Whether sAPP, FUS, TDP-43 and SOD1 are mechanistically linked in a common pathway deregulated in both AD and ALS is not known. SUMMARY We show that sAPP, TDP-43, FUS and SOD1 are transported to neurite terminals by a mechanism that involves endoplasmic reticulum (ER)-like tubules and requires peripherin NFs. The transport of these proteins, and the translocation of the ER protein reticulon 4 (Rtn4) into neurites was studied in CAD cells, a brainstem-derived neuronal cell line highly relevant to AD and ALS. We show that a significant fraction of sAPP is generated in the soma and accumulates in a juxtanuclear ER subdomain. In neurites, sAPP localizes to Rtn4-positive ER-like tubules that extend from the soma into the growth cone and colocalizes with peripherin NFs. Knocking down peripherin disrupts the NF network and diminishes the accumulation of sAPP, TDP-43, FUS, SOD1 and Rtn4 at terminals. KEY MESSAGES We propose that the impediment of a common, ER-mediated mechanism of transport of sAPP, TDP-43, FUS and SOD1, caused by a disrupted NF network, could be part of the mechanisms leading to AD and ALS.
Collapse
Affiliation(s)
- Virgil Muresan
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, N.J., USA
| | | |
Collapse
|
130
|
Foley AM, Ammar ZM, Lee RH, Mitchell CS. Systematic review of the relationship between amyloid-β levels and measures of transgenic mouse cognitive deficit in Alzheimer's disease. J Alzheimers Dis 2015; 44:787-95. [PMID: 25362040 PMCID: PMC4346318 DOI: 10.3233/jad-142208] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Amyloid-β (Aβ) is believed to directly affect memory and learning in Alzheimer’s disease (AD). It is widely suggested that there is a relationship between Aβ40 and Aβ42 levels and cognitive performance. In order to explore the validity of this relationship, we performed a meta-analysis of 40 peer-reviewed, published AD transgenic mouse studies that quantitatively measured Aβ levels in brain tissue after assessing cognitive performance. We examined the relationship between Aβ levels (Aβ40, Aβ42, or the ratio of Aβ42 to Aβ40) and cognitive function as measured by escape latency times in the Morris water maze or exploratory preference percentage in the novel object recognition test. Our systematic review examined five mouse models (Tg2576, APP, PS1, 3xTg, APP(OSK)-Tg), gender, and age. The overall result revealed no statistically significant correlation between quantified Aβ levels and experimental measures of cognitive function. However, enough of the trends were of the same sign to suggest that there probably is a very weak qualitative trend visible only across many orders of magnitude. In summary, the results of the systematic review revealed that mice bred to show elevated levels of Aβ do not perform significantly worse in cognitive tests than mice that do not have elevated Aβ levels. Our results suggest two lines of inquiry: 1) Aβ is a biochemical “side effect” of the AD pathology; or 2) learning and memory deficits in AD are tied to the presence of qualitatively “high” levels of Aβ but are not quantitatively sensitive to the levels themselves.
Collapse
Affiliation(s)
- Avery M Foley
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Zeena M Ammar
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Robert H Lee
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Cassie S Mitchell
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| |
Collapse
|
131
|
Kim GH, Kim JE, Rhie SJ, Yoon S. The Role of Oxidative Stress in Neurodegenerative Diseases. Exp Neurobiol 2015; 24:325-40. [PMID: 26713080 PMCID: PMC4688332 DOI: 10.5607/en.2015.24.4.325] [Citation(s) in RCA: 930] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 09/14/2015] [Accepted: 09/14/2015] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress is induced by an imbalanced redox states, involving either excessive generation of reactive oxygen species (ROS) or dysfunction of the antioxidant system. The brain is one of organs especially vulnerable to the effects of ROS because of its high oxygen demand and its abundance of peroxidation-susceptible lipid cells. Previous studies have demonstrated that oxidative stress plays a central role in a common pathophysiology of neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Antioxidant therapy has been suggested for the prevention and treatment of neurodegenerative diseases, although the results with regard to their efficacy of treating neurodegenerative disease have been inconsistent. In this review, we will discuss the role of oxidative stress in the pathophysiology of neurodegenerative diseases and in vivo measurement of an index of damage by oxidative stress. Moreover, the present knowledge on antioxidant in the treatment of neurodegenerative diseases and future directions will be outlined.
Collapse
Affiliation(s)
- Geon Ha Kim
- Ewha Brain Institute, Ewha Womans University, Seoul 03760, Korea. ; Department of Neurology, Ewha Womans University Mokdong Hospital, Ewha Womans University School of Medicine, Seoul 03760, Korea
| | - Jieun E Kim
- Ewha Brain Institute, Ewha Womans University, Seoul 03760, Korea. ; Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Sandy Jeong Rhie
- Ewha Brain Institute, Ewha Womans University, Seoul 03760, Korea. ; College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Sujung Yoon
- Ewha Brain Institute, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
132
|
The Role of Oxidative Stress-Induced Epigenetic Alterations in Amyloid-β Production in Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:604658. [PMID: 26543520 PMCID: PMC4620382 DOI: 10.1155/2015/604658] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 12/15/2014] [Indexed: 11/17/2022]
Abstract
An increasing number of studies have proposed a strong correlation between reactive oxygen species (ROS)-induced oxidative stress (OS) and the pathogenesis of Alzheimer's disease (AD). With over five million people diagnosed in the United States alone, AD is the most common type of dementia worldwide. AD includes progressive neurodegeneration, followed by memory loss and reduced cognitive ability. Characterized by the formation of amyloid-beta (Aβ) plaques as a hallmark, the connection between ROS and AD is compelling. Analyzing the ROS response of essential proteins in the amyloidogenic pathway, such as amyloid-beta precursor protein (APP) and beta-secretase (BACE1), along with influential signaling programs of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and c-Jun N-terminal kinase (JNK), has helped visualize the path between OS and Aβ overproduction. In this review, attention will be paid to significant advances in the area of OS, epigenetics, and their influence on Aβ plaque assembly. Additionally, we aim to discuss available treatment options for AD that include antioxidant supplements, Asian traditional medicines, metal-protein-attenuating compounds, and histone modifying inhibitors.
Collapse
|
133
|
Violet M, Chauderlier A, Delattre L, Tardivel M, Chouala MS, Sultan A, Marciniak E, Humez S, Binder L, Kayed R, Lefebvre B, Bonnefoy E, Buée L, Galas MC. Prefibrillar Tau oligomers alter the nucleic acid protective function of Tau in hippocampal neurons in vivo. Neurobiol Dis 2015; 82:540-551. [PMID: 26385829 DOI: 10.1016/j.nbd.2015.09.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 07/09/2015] [Accepted: 09/13/2015] [Indexed: 01/05/2023] Open
Abstract
The accumulation of DNA and RNA oxidative damage is observed in cortical and hippocampal neurons from Alzheimer's disease (AD) brains at early stages of pathology. We recently reported that Tau is a key nuclear player in the protection of neuronal nucleic acid integrity in vivo under physiological conditions and hyperthermia, a strong inducer of oxidative stress. In a mouse model of tauopathy (THY-Tau22), we demonstrate that hyperthermia selectively induces nucleic acid oxidative damage and nucleic acid strand breaks in the nucleus and cytoplasm of hippocampal neurons that display early Tau phosphorylation but no Tau fibrils. Nucleic acid-damaged neurons were exclusively immunoreactive for prefibrillar Tau oligomers. A similar association between prefibrillar Tau oligomers and nucleic acid oxidative damage was observed in AD brains. Pretreatment with Methylene Blue (MB), a Tau aggregation inhibitor and a redox cycler, reduced hyperthermia-induced Tau oligomerization as well as nucleic acid damage. This study clearly highlights the existence of an early and critical time frame for hyperthermia-induced Tau oligomerization, which most likely occurs through increased oxidative stress, and nucleic acid vulnerability during the progression of Tau pathology. These results suggest that at early stages of AD, Tau oligomerization triggers the loss of the nucleic acid protective function of monomeric Tau. This study highlights the existence of a short therapeutic window in which to prevent the formation of pathological forms of Tau and their harmful consequences on nucleic acid integrity during the progression of Tau pathology.
Collapse
Affiliation(s)
- Marie Violet
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Alban Chauderlier
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Lucie Delattre
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Meryem Tardivel
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Meliza Sendid Chouala
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Audrey Sultan
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Elodie Marciniak
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Sandrine Humez
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Lester Binder
- Department of Translational Science & Molecular Medicine, College of Human Medicine, Michigan State University, 333 Bostwick Ave. NE, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Rakez Kayed
- Department of Neurology, George and Cynthia Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Medical Research Building, Room 10.138C, Galveston, TX 77555-1045, USA; Department of Neuroscience & Cell Biology, George and Cynthia Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Medical Research Building, Room 10.138C, Galveston, TX 77555-1045, USA
| | - Bruno Lefebvre
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Eliette Bonnefoy
- Inserm UMRS 1007, Université Paris Descartes, 45 rue des Saints Pères, 75006 Paris Cedex 06, France
| | - Luc Buée
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Marie-Christine Galas
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France.
| |
Collapse
|
134
|
Wang SW, Yang SG, Liu W, Zhang YX, Xu PX, Wang T, Ling TJ, Liu RT. Alpha-tocopherol quinine ameliorates spatial memory deficits by reducing beta-amyloid oligomers, neuroinflammation and oxidative stress in transgenic mice with Alzheimer's disease. Behav Brain Res 2015; 296:109-117. [PMID: 26358659 DOI: 10.1016/j.bbr.2015.09.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/31/2015] [Accepted: 09/02/2015] [Indexed: 11/18/2022]
Abstract
The pathologies of Alzheimer's disease (AD) is associated with soluble beta-amyloid (Aβ) oligomers, neuroinflammation and oxidative stress. Decreasing the levels of Aβ oligomer, glial activation and oxidative stress are potential therapeutic approaches for AD treatment. We previously found alpha-tocopherol quinine (α-TQ) inhibited Aβ aggregation and cytotoxicity, decreased the release of inflammatory cytokines and reactive oxygen species (ROS) in vitro. However, whether α-TQ ameliorates memory deficits and other neuropathologies in mice or patients with AD remains unknown. In this study, we reported that orally administered α-TQ ameliorated memory impairment in APPswe/PS1dE9 transgenic mice, decreased oxidative stress and the levels of Aβ oligomer in the brains of mice, prevented the production of inducible nitric oxide synthase and inflammatory mediators, such as interleukin-6 and interleukin-1β, and inhibited microglial activation by inhibiting NF-κB signaling pathway. These findings suggest that α-TQ has potential therapeutic value for AD treatment.
Collapse
Affiliation(s)
- Shao-Wei Wang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Shi-Gao Yang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Wen Liu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; School of Life Science, Anhui Agricultural University, Hefei 230036, China
| | - Yang-Xin Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; School of Life Science, Anhui Agricultural University, Hefei 230036, China
| | - Peng-Xin Xu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; School of Life Science, Ningxia University, Yinchuan 750021, China
| | - Teng Wang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; School of Life Science, Anhui Agricultural University, Hefei 230036, China
| | - Tie-Jun Ling
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei 230036, China.
| | - Rui-Tian Liu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
135
|
Wiseman FK, Al-Janabi T, Hardy J, Karmiloff-Smith A, Nizetic D, Tybulewicz VLJ, Fisher EMC, Strydom A. A genetic cause of Alzheimer disease: mechanistic insights from Down syndrome. Nat Rev Neurosci 2015; 16:564-74. [PMID: 26243569 PMCID: PMC4678594 DOI: 10.1038/nrn3983] [Citation(s) in RCA: 357] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Down syndrome, which arises in individuals carrying an extra copy of chromosome 21, is associated with a greatly increased risk of early-onset Alzheimer disease. It is thought that this risk is conferred by the presence of three copies of the gene encoding amyloid precursor protein (APP)--an Alzheimer disease risk factor--although the possession of extra copies of other chromosome 21 genes may also play a part. Further study of the mechanisms underlying the development of Alzheimer disease in people with Down syndrome could provide insights into the mechanisms that cause dementia in the general population.
Collapse
Affiliation(s)
- Frances K Wiseman
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Tamara Al-Janabi
- Division of Psychiatry, University College London, Maple House, 149 Tottenham Court Road, London W1T 7NF, UK
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Annette Karmiloff-Smith
- Centre for Brain and Cognitive Development, Birkbeck, University of London, Malet Street, London WC1E 7HX, UK
| | - Dean Nizetic
- Lee Kong Chian School of Medicine, Nanyang Technological University, Novena Campus, 11 Mandalay Road, Singapore 308232; and the Blizard Institute, Barts and the London School of Medicine, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
| | | | - Elizabeth M C Fisher
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - André Strydom
- Division of Psychiatry, University College London, Maple House, 149 Tottenham Court Road, London W1T 7NF, UK
| |
Collapse
|
136
|
The Ambiguous Relationship of Oxidative Stress, Tau Hyperphosphorylation, and Autophagy Dysfunction in Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:352723. [PMID: 26171115 PMCID: PMC4485995 DOI: 10.1155/2015/352723] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 12/30/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. The pathological hallmarks of AD are amyloid plaques [aggregates of amyloid-beta (Aβ)] and neurofibrillary tangles (aggregates of tau). Growing evidence suggests that tau accumulation is pathologically more relevant to the development of neurodegeneration and cognitive decline in AD patients than Aβ plaques. Oxidative stress is a prominent early event in the pathogenesis of AD and is therefore believed to contribute to tau hyperphosphorylation. Several studies have shown that the autophagic pathway in neurons is important under physiological and pathological conditions. Therefore, this pathway plays a crucial role for the degradation of endogenous soluble tau. However, the relationship between oxidative stress, tau protein hyperphosphorylation, autophagy dysregulation, and neuronal cell death in AD remains unclear. Here, we review the latest progress in AD, with a special emphasis on oxidative stress, tau hyperphosphorylation, and autophagy. We also discuss the relationship of these three factors in AD.
Collapse
|
137
|
Murakami K. Conformation-specific antibodies to target amyloid β oligomers and their application to immunotherapy for Alzheimer's disease. Biosci Biotechnol Biochem 2015; 78:1293-305. [PMID: 25130729 DOI: 10.1080/09168451.2014.940275] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Amyloid β-protein (Aβ) oligomers, intermediates of Aβ aggregation, cause cognitive impairment and synaptotoxicity in the pathogenesis of Alzheimer's disease (AD). Immunotherapy using anti-Aβ antibody is one of the most promising approaches for AD treatment. However, most clinical trials using conventional sequence-specific antibodies have proceeded with difficulty. This is probably due to the unintended removal of the non-pathological monomer and fibrils of Aβ as well as the pathological oligomers by these antibodies that recognize Aβ sequence, which is not involved in synaptotoxicity. Several efforts have been made recently to develop conformation-specific antibodies that target the tertiary structure of Aβ oligomers. Here, we review the recent findings of Aβ oligomers and anti-Aβ antibodies including our own, and discuss their potential as therapeutic and diagnostic tools.
Collapse
Affiliation(s)
- Kazuma Murakami
- a Division of Food Science and Biotechnology , Graduate School of Agriculture, Kyoto University , Kyoto , Japan
| |
Collapse
|
138
|
Oxidative Stress during the Progression of β-Amyloid Pathology in the Neocortex of the Tg2576 Mouse Model of Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:967203. [PMID: 25973140 PMCID: PMC4418010 DOI: 10.1155/2015/967203] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 03/26/2015] [Accepted: 03/27/2015] [Indexed: 12/05/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by progressive neurodegeneration. Pathogenetic mechanisms, triggered by β-amyloid (Aβ) accumulation, include oxidative stress, derived from energy homeostasis deregulation and involving mitochondria and peroxisomes. We here addressed the oxidative stress status and the elicited cellular response at the onset and during the progression of Aβ pathology, studying the neocortex of Tg2576 model of AD. Age-dependent changes of oxidative damage markers, antioxidant enzymes, and related transcription factors were analysed in relation to the distribution of Aβ peptide and oligomers, by a combined molecular/morphological approach. Nucleic acid oxidative damage, accompanied by defective antioxidant defences, and decreased PGC1α expression are already detected in 3-month-old Tg2576 neurons. Conversely, PPARα is increased in these cells, with its cytoplasmic localization suggesting nongenomic, anti-inflammatory actions. At 6 months, when intracellular Aβ accumulates, PMP70 is downregulated, indicating impairment of fatty acids peroxisomal translocation and their consequent harmful accumulation. In 9-month-old Tg2576 neocortex, Aβ oligomers and acrolein deposition correlate with GFAP, GPX1, and PMP70 increases, supporting a compensatory response, involving astroglial peroxisomes. At severe pathological stages, when senile plaques disrupt cortical cytoarchitecture, antioxidant capacity is gradually lost. Overall, our data suggest early therapeutic intervention in AD, also targeting peroxisomes.
Collapse
|
139
|
Wang ZX, Tan L, Liu J, Yu JT. The Essential Role of Soluble Aβ Oligomers in Alzheimer's Disease. Mol Neurobiol 2015; 53:1905-1924. [PMID: 25833098 DOI: 10.1007/s12035-015-9143-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/18/2015] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease characterized by amyloid plaque and neurofibrillary tangles (NFT). With the finding that soluble nonfibrillar Aβ levels actually correlate strongly with the severity of the disease, the initial focus on amyloid plaques shifted to the contemporary concept that AD memory failure is caused by soluble Aβ oligomers. The soluble Aβ are known to be more neurotoxicthan fibrillar Aβ species. In this paper, we summarize the essential role of soluble Aβ oligomers in AD and discuss therapeutic strategies that target soluble Aβ oligomers.
Collapse
Affiliation(s)
- Zi-Xuan Wang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China.
| | - Jinyuan Liu
- Columbia College, Columbia University, New York, NY, USA
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China. .,Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA.
| |
Collapse
|
140
|
Kim D, Park GB, Hur DY. Apoptotic signaling through reactive oxygen species in cancer cells. World J Immunol 2014; 4:158-173. [DOI: 10.5411/wji.v4.i3.158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 06/03/2014] [Accepted: 10/16/2014] [Indexed: 02/05/2023] Open
Abstract
Reactive oxygen species (ROS) take part in diverse biological processes like cell growth, programmed cell death, cell senescence, and maintenance of the transformed state through regulation of signal transduction. Cancer cells adapt to new higher ROS circumstance. Sometimes, ROS induce cancer cell proliferation. Meanwhile, elevated ROS render cancer cells vulnerable to oxidative stress-induced cell death. However, this prominent character of cancer cells allows acquiring a resistance to oxidative stress conditions relative to normal cells. Activated signaling pathways that increase the level of intracellular ROS in cancer cells not only render up-regulation of several genes involved in cellular proliferation and evasion of apoptosis but also cause cancer cells and cancer stem cells to develop a high metabolic rate. In over the past several decades, many studies have indicated that ROS play a critical role as the secondary messenger of tumorigenesis and metastasis in cancer from both in vitro and in vivo. Here we summarize the role of ROS and anti-oxidants in contributing to or preventing cancer. In addition, we review the activated signaling pathways that make cancer cells susceptible to death.
Collapse
|
141
|
Campos PB, Paulsen BS, Rehen SK. Accelerating neuronal aging in in vitro model brain disorders: a focus on reactive oxygen species. Front Aging Neurosci 2014; 6:292. [PMID: 25386139 PMCID: PMC4209886 DOI: 10.3389/fnagi.2014.00292] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/06/2014] [Indexed: 12/20/2022] Open
Abstract
In this review, we discuss insights gained through the use of stem cell preparations regarding the modeling of neurological diseases, the need for aging neurons derived from pluripotent stem cells to further advance the study of late-onset adult neurological diseases, and the extent to which mechanisms linked to the mismanagement of reactive oxygen species (ROS). The context of these issues can be revealed using the three disease states of Parkinson’s (PD), Alzheimer’s (AD), and schizophrenia, as considerable insights have been gained into these conditions through the use of stem cells in terms of disease etiologies and the role of oxidative stress. The latter subject is a primary area of interest of our group. After discussing the molecular models of accelerated aging, we highlight the role of ROS for the three diseases explored here. Importantly, we do not seek to provide an extensive account of all genetic mutations for each of the three disorders discussed in this review, but we aim instead to provide a conceptual framework that could maximize the gains from merging the approaches of stem cell microsystems and the study of oxidative stress in disease in order to optimize therapeutics and determine new molecular targets against oxidative stress that spare stem cell proliferation and development.
Collapse
Affiliation(s)
- Priscila Britto Campos
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Bruna S Paulsen
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Stevens K Rehen
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro Rio de Janeiro, Brazil ; D'Or Institute for Research and Education (IDOR) Rio de Janeiro, Brazil
| |
Collapse
|
142
|
Palladium and platinum nanoparticles attenuate aging-like skin atrophy via antioxidant activity in mice. PLoS One 2014; 9:e109288. [PMID: 25333617 PMCID: PMC4198089 DOI: 10.1371/journal.pone.0109288] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 08/30/2014] [Indexed: 12/29/2022] Open
Abstract
Cu-Zn superoxide dismutase (Sod1) loss causes a redox imbalance as it leads to excess superoxide generation, which results in the appearance of various aging-related phenotypes, including skin atrophy. Noble metal nanoparticles, such as palladium (Pd) and platinum (Pt) nanoparticles, are considered to function as antioxidants due to their strong catalytic activity. In Japan, a mixture of Pd and Pt nanoparticles called PAPLAL has been used to treat chronic diseases over the past 60 years. In the present study, we investigated the protective effects of PAPLAL against aging-related skin pathologies in mice. Transdermal PAPLAL treatment reversed skin thinning associated with increased lipid peroxidation in Sod1−/− mice. Furthermore, PAPLAL normalized the gene expression levels of Col1a1, Mmp2, Has2, Tnf-α, Il-6, and p53 in the skin of the Sod1−/− mice. Pt nanoparticles exhibited marked SOD and catalase activity, while Pd nanoparticles only displayed weak SOD and catalase activity in vitro. Although the SOD and catalase activity of the Pt nanoparticles significantly declined after they had been oxidized in air, a mixture of Pd and Pt nanoparticles continued to exhibit SOD and catalase activity after oxidation. Importantly, a mixture of Pd and Pt nanoparticles with a molar ratio of 3 or 4 to 1 continued to exhibit SOD and catalase activity after oxidation, indicating that Pd nanoparticles prevent the oxidative deterioration of Pt nanoparticles. These findings indicate that PAPLAL stably suppresses intrinsic superoxide generation both in vivo and in vitro via SOD and catalase activity. PAPLAL is a potentially powerful tool for the treatment of aging-related skin diseases caused by oxidative damage.
Collapse
|
143
|
Ser70 phosphorylation of Bcl-2 by selective tyrosine nitration of PP2A-B56δ stabilizes its antiapoptotic activity. Blood 2014; 124:2223-34. [DOI: 10.1182/blood-2014-03-563296] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Key Points
O2− modifies B56δ at Y289 to block the PP2A holoenzyme assembly. This results in S70 Bcl-2 phosphorylation and promotes tumor chemoresistance. Primary lymphomas with low SOD1 have high B56δ tyrosine nitration and S70pBcl-2.
Collapse
|
144
|
Watanabe K, Shibuya S, Ozawa Y, Nojiri H, Izuo N, Yokote K, Shimizu T. Superoxide dismutase 1 loss disturbs intracellular redox signaling, resulting in global age-related pathological changes. BIOMED RESEARCH INTERNATIONAL 2014; 2014:140165. [PMID: 25276767 PMCID: PMC4170698 DOI: 10.1155/2014/140165] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/29/2014] [Accepted: 08/06/2014] [Indexed: 01/14/2023]
Abstract
Aging is characterized by increased oxidative stress, chronic inflammation, and organ dysfunction, which occur in a progressive and irreversible manner. Superoxide dismutase (SOD) serves as a major antioxidant and neutralizes superoxide radicals throughout the body. In vivo studies have demonstrated that copper/zinc superoxide dismutase-deficient (Sod1(-/-)) mice show various aging-like pathologies, accompanied by augmentation of oxidative damage in organs. We found that antioxidant treatment significantly attenuated the age-related tissue changes and oxidative damage-associated p53 upregulation in Sod1(-/-) mice. This review will focus on various age-related pathologies caused by the loss of Sod1 and will discuss the molecular mechanisms underlying the pathogenesis in Sod1(-/-) mice.
Collapse
Affiliation(s)
- Kenji Watanabe
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Shuichi Shibuya
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Yusuke Ozawa
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Hidetoshi Nojiri
- Department of Orthopaedics, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Naotaka Izuo
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Koutaro Yokote
- Department of Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Takahiko Shimizu
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| |
Collapse
|
145
|
Torres-Lista V, Parrado-Fernández C, Alvarez-Montón I, Frontiñán-Rubio J, Durán-Prado M, Peinado JR, Johansson B, Alcaín FJ, Giménez-Llort L. Neophobia, NQO1 and SIRT1 as premorbid and prodromal indicators of AD in 3xTg-AD mice. Behav Brain Res 2014; 271:140-6. [DOI: 10.1016/j.bbr.2014.04.055] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 04/25/2014] [Accepted: 04/30/2014] [Indexed: 10/25/2022]
|
146
|
Nazeri A, Ganjgahi H, Roostaei T, Nichols T, Zarei M. Imaging proteomics for diagnosis, monitoring and prediction of Alzheimer's disease. Neuroimage 2014; 102 Pt 2:657-65. [PMID: 25173418 DOI: 10.1016/j.neuroimage.2014.08.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 08/18/2014] [Accepted: 08/22/2014] [Indexed: 01/18/2023] Open
Abstract
Proteomic and imaging markers have been widely studied as potential biomarkers for diagnosis, monitoring and prognosis of Alzheimer's disease. In this study, we used Alzheimer Disease Neuroimaging Initiative dataset and performed parallel independent component analysis on cross sectional and longitudinal proteomic and imaging data in order to identify the best proteomic model for diagnosis, monitoring and prediction of Alzheimer disease (AD). We used plasma proteins measurement and imaging data from AD and healthy controls (HC) at the baseline and 1 year follow-up. Group comparisons at baseline and changes over 1 year were calculated for proteomic and imaging data. The results were fed into parallel independent component analysis in order to identify proteins that were associated with structural brain changes cross sectionally and longitudinally. Regression model was used to find the best model that can discriminate AD from HC, monitor AD and to predict MCI converters from non-converters. We showed that five proteins are associated with structural brain changes in the brain. These proteins could discriminate AD from HC with 57% specificity and 89% sensitivity. Four proteins whose change over 1 year were associated with brain structural changes could discriminate AD from HC with sensitivity of 93%, and specificity of 92%. This model predicted MCI conversion to AD in 2 years with 94% accuracy. This model has the highest accuracy in prediction of MCI conversion to AD within the ADNI-1 dataset. This study shows that combination of selected plasma protein levels and MR imaging is a useful method in identifying potential biomarker.
Collapse
Affiliation(s)
- Arash Nazeri
- Interdisciplinary Neuroscience Research Program, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Habib Ganjgahi
- National Brain Mapping Centre, and Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran 4739, Iran; Department of Statistics, University of Warwick, Coventry CV4 7AL, UK
| | - Tina Roostaei
- Interdisciplinary Neuroscience Research Program, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Thomas Nichols
- Department of Statistics, University of Warwick, Coventry CV4 7AL, UK
| | - Mojtaba Zarei
- National Brain Mapping Centre, and Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran 4739, Iran.
| | | |
Collapse
|
147
|
Wang BY, Xu XS, Cui YX, Wang H, Liu G, Zhao ZJ, Ma JF, Fu XQ. Caenorhabditis elegans eyes absent ortholog EYA-1 Is required for stress resistance. BIOCHEMISTRY (MOSCOW) 2014; 79:653-62. [DOI: 10.1134/s0006297914070074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
148
|
Potter-Baker KA, Ravikumar M, Burke AA, Meador WD, Householder KT, Buck AC, Sunil S, Stewart WG, Anna JP, Tomaszewski WH, Capadona JR. A comparison of neuroinflammation to implanted microelectrodes in rat and mouse models. Biomaterials 2014; 35:5637-46. [PMID: 24755527 PMCID: PMC4071936 DOI: 10.1016/j.biomaterials.2014.03.076] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/27/2014] [Indexed: 12/31/2022]
Abstract
Rat models have emerged as a common tool to study neuroinflammation to intracortical microelectrodes. While a number of studies have attempted to understand the factors resulting in neuroinflammation using rat models, a complete understanding of key mechanistic pathways remains elusive. Transgenic mouse models, however, could facilitate a deeper understanding of mechanistic pathways due to an ease of genetic alteration. Therefore, the goal of the present study is to compare neuroinflammation following microelectrode implantation between the rat and the mouse model. Our study suggests that subtle differences in the classic neuroinflammatory markers exist between the animal models at both two and sixteen weeks post implantation. Most notably, neuronal densities surrounding microelectrodes were significantly lower in the rat model at two weeks, while similar densities were observed between the animal models at sixteen weeks. Physiological differences between the species and slight alterations in surgical methods are likely key contributors to the observed differences. Moving forward, we propose that differences in the time course of neuroinflammation between the animal models should be considered when trying to understand and prevent intracortical microelectrode failure.
Collapse
Affiliation(s)
- Kelsey A Potter-Baker
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA; Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702, USA
| | - Madhumitha Ravikumar
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA; Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702, USA
| | - Alan A Burke
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA
| | - William D Meador
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA
| | - Kyle T Householder
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA; Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702, USA
| | - Amy C Buck
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA; Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702, USA
| | - Smrithi Sunil
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA; Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702, USA
| | - Wade G Stewart
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA
| | - Jake P Anna
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA
| | - William H Tomaszewski
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA; Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702, USA.
| |
Collapse
|
149
|
Cerium oxide nanoparticles protect against Aβ-induced mitochondrial fragmentation and neuronal cell death. Cell Death Differ 2014; 21:1622-32. [PMID: 24902900 DOI: 10.1038/cdd.2014.72] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 04/21/2014] [Accepted: 04/22/2014] [Indexed: 12/17/2022] Open
Abstract
Evidence indicates that nitrosative stress and mitochondrial dysfunction participate in the pathogenesis of Alzheimer's disease (AD). Amyloid beta (Aβ) and peroxynitrite induce mitochondrial fragmentation and neuronal cell death by abnormal activation of dynamin-related protein 1 (DRP1), a large GTPase that regulates mitochondrial fission. The exact mechanisms of mitochondrial fragmentation and DRP1 overactivation in AD remain unknown; however, DRP1 serine 616 (S616) phosphorylation is likely involved. Although it is clear that nitrosative stress caused by peroxynitrite has a role in AD, effective antioxidant therapies are lacking. Cerium oxide nanoparticles, or nanoceria, switch between their Ce(3+) and Ce(4+) states and are able to scavenge superoxide anions, hydrogen peroxide and peroxynitrite. Therefore, nanoceria might protect against neurodegeneration. Here we report that nanoceria are internalized by neurons and accumulate at the mitochondrial outer membrane and plasma membrane. Furthermore, nanoceria reduce levels of reactive nitrogen species and protein tyrosine nitration in neurons exposed to peroxynitrite. Importantly, nanoceria reduce endogenous peroxynitrite and Aβ-induced mitochondrial fragmentation, DRP1 S616 hyperphosphorylation and neuronal cell death.
Collapse
|
150
|
Di Domenico F, Pupo G, Tramutola A, Giorgi A, Schininà ME, Coccia R, Head E, Butterfield DA, Perluigi M. Redox proteomics analysis of HNE-modified proteins in Down syndrome brain: clues for understanding the development of Alzheimer disease. Free Radic Biol Med 2014; 71:270-280. [PMID: 24675226 PMCID: PMC4686229 DOI: 10.1016/j.freeradbiomed.2014.03.027] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/04/2014] [Accepted: 03/18/2014] [Indexed: 01/18/2023]
Abstract
Down syndrome (DS) is the most common genetic cause of intellectual disability, due to partial or complete triplication of chromosome 21. DS subjects are characterized by a number of abnormalities including premature aging and development of Alzheimer disease (AD) neuropathology after approximately 40 years of age. Several studies show that oxidative stress plays a crucial role in the development of neurodegeneration in the DS population. Increased lipid peroxidation is one of the main events causing redox imbalance within cells through the formation of toxic aldehydes that easily react with DNA, lipids, and proteins. In this study we used a redox proteomics approach to identify specific targets of 4-hydroxynonenal modifications in the frontal cortex from DS cases with and without AD pathology. We suggest that a group of identified proteins followed a specific pattern of oxidation in DS vs young controls, probably indicating characteristic features of the DS phenotype; a second group of identified proteins showed increased oxidation in DS/AD vs DS, thus possibly playing a role in the development of AD. The third group of comparison, DS/AD vs old controls, identified proteins that may be considered specific markers of AD pathology. All the identified proteins are involved in important biological functions including intracellular quality control systems, cytoskeleton network, energy metabolism, and antioxidant response. Our results demonstrate that oxidative damage is an early event in DS, as well as dysfunctions of protein-degradation systems and cellular protective pathways, suggesting that DS subjects are more vulnerable to oxidative damage accumulation that might contribute to AD development. Further, considering that the majority of proteins have been already demonstrated to be oxidized in AD brain, our results strongly support similarities with AD in DS.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Gilda Pupo
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Alessandra Giorgi
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | | | - Raffaella Coccia
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Elizabeth Head
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA
| | - D Allan Butterfield
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA; Department of Chemistry and Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506-0055, USA
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy.
| |
Collapse
|