1051
|
Chan KK, Yap SHK, Yong KT. Biogreen Synthesis of Carbon Dots for Biotechnology and Nanomedicine Applications. NANO-MICRO LETTERS 2018; 10:72. [PMID: 30417004 PMCID: PMC6208800 DOI: 10.1007/s40820-018-0223-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/02/2018] [Indexed: 05/14/2023]
Abstract
Over the past decade, carbon dots have ignited a burst of interest in many different fields, including nanomedicine, solar energy, optoelectronics, energy storage, and sensing applications, owing to their excellent photoluminescence properties and the easiness to modify their optical properties through doping and functionalization. In this review, the synthesis, structural and optical properties, as well as photoluminescence mechanisms of carbon dots are first reviewed and summarized. Then, we describe a series of designs for carbon dot-based sensors and the different sensing mechanisms associated with them. Thereafter, we elaborate on recent research advances on carbon dot-based sensors for the selective and sensitive detection of a wide range of analytes, including heavy metals, cations, anions, biomolecules, biomarkers, nitroaromatic explosives, pollutants, vitamins, and drugs. Lastly, we provide a concluding perspective on the overall status, challenges, and future directions for the use of carbon dots in real-life sensing.
Collapse
Affiliation(s)
- Kok Ken Chan
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Stephanie Hui Kit Yap
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Ken-Tye Yong
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore, 639798, Singapore.
| |
Collapse
|
1052
|
Vela D. The Dual Role of Hepcidin in Brain Iron Load and Inflammation. Front Neurosci 2018; 12:740. [PMID: 30374287 PMCID: PMC6196657 DOI: 10.3389/fnins.2018.00740] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 09/26/2018] [Indexed: 12/25/2022] Open
Abstract
Hepcidin is the major regulator of systemic iron metabolism, while the role of this peptide in the brain has just recently been elucidated. Studies suggest a dual role of hepcidin in neuronal iron load and inflammation. This is important since neuronal iron load and inflammation are pathophysiological processes frequently associated with neurodegeneration. Furthermore, manipulation of hepcidin activity has recently been used to recover neuronal damage due to brain inflammation in animal models and cultured cells. Therefore, understanding the mechanistic insights of hepcidin action in the brain is important to uncover its role in treating neuronal damage in neurodegenerative diseases.
Collapse
Affiliation(s)
- Driton Vela
- Department of Physiology, Faculty of Medicine, University of Pristina, Pristina, Kosovo
| |
Collapse
|
1053
|
Park M, Moon WJ, Moon Y, Choi JW, Han SH, Wang Y. Region-specific susceptibility change in cognitively impaired patients with diabetes mellitus. PLoS One 2018; 13:e0205797. [PMID: 30308069 PMCID: PMC6181414 DOI: 10.1371/journal.pone.0205797] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/02/2018] [Indexed: 12/22/2022] Open
Abstract
Emerging evidence suggests that diabetes mellitus (DM) is associated with iron and calcium metabolism. However, few studies have investigated the presence of DM in cognitively impaired patients and its effect on brain iron and calcium accumulation. Therefore, we assessed the effects of DM on cognitively impaired patients using quantitative susceptibility mapping (QSM). From June 2012 to Feb 2014, 92 eligible cognitively impaired patients underwent 3T magnetic resonance imaging (MRI). There were 46 patients with DM (DM+) and 46 aged matched patients without DM (DM-). QSM was obtained from gradient echo data and analyzed by drawing regions of interest around relevant anatomical structures. Clinical factors and vascular pathology were also evaluated. Measurement differences between DM+ and DM- patients were assessed by t tests. A multiple regression analysis was performed to identify independent predictors of magnetic susceptibility. DM+ patients showed lower susceptibility values, indicative of lower brain iron content, than DM- patients, which was significant in the hippocampus (4.80 ± 8.31 ppb versus 0.22 ± 10.60 ppb, p = 0.024) and pulvinar of the thalamus (36.30 ± 19.88 ppb versus 45.90 ± 20.02 ppb, p = 0.023). On multiple regression analysis, microbleed number was a predictor of susceptibility change in the hippocampus (F = 4.291, beta = 0.236, p = 0.042) and DM was a predictor of susceptibility change in the pulvinar of the thalamus (F = 4.900, beta = - 0.251, p = 0.030). In cognitively impaired patients, presence of DM was associated with lower susceptibility change in the pulvinar of the thalamus and hippocampus. This suggests that there may be region-specific alterations of calcium deposition in cognitively impaired subjects with DM.
Collapse
Affiliation(s)
- Mina Park
- Department of Radiology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Won-Jin Moon
- Department of Radiology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Yeonsil Moon
- Department of Neurology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Jin Woo Choi
- Department of Radiology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Seol-Heui Han
- Department of Neurology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Yi Wang
- Department of Radiology, Weill Cornell Medical College, New York, New York, United States of America
| |
Collapse
|
1054
|
A review on iron chelators as potential therapeutic agents for the treatment of Alzheimer’s and Parkinson’s diseases. Mol Divers 2018; 23:509-526. [DOI: 10.1007/s11030-018-9878-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/25/2018] [Indexed: 12/19/2022]
|
1055
|
Seibler P, Burbulla LF, Dulovic M, Zittel S, Heine J, Schmidt T, Rudolph F, Westenberger A, Rakovic A, Münchau A, Krainc D, Klein C. Iron overload is accompanied by mitochondrial and lysosomal dysfunction in WDR45 mutant cells. Brain 2018; 141:3052-3064. [PMID: 30169597 PMCID: PMC7190033 DOI: 10.1093/brain/awy230] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 07/08/2018] [Accepted: 07/15/2018] [Indexed: 01/10/2023] Open
Abstract
Beta-propeller protein-associated neurodegeneration is a subtype of monogenic neurodegeneration with brain iron accumulation caused by de novo mutations in WDR45. The WDR45 protein functions as a beta-propeller scaffold and plays a putative role in autophagy through its interaction with phospholipids and autophagy-related proteins. Loss of WDR45 function due to disease-causing mutations has been linked to defects in autophagic flux in patient and animal cells. However, the role of WDR45 in iron homeostasis remains elusive. Here we studied patient-specific WDR45 mutant fibroblasts and induced pluripotent stem cell-derived midbrain neurons. Our data demonstrated that loss of WDR45 increased cellular iron levels and oxidative stress, accompanied by mitochondrial abnormalities, autophagic defects, and diminished lysosomal function. Restoring WDR45 levels partially rescued oxidative stress and the susceptibility to iron treatment, and activation of autophagy reduced the observed iron overload in WDR45 mutant cells. Our data suggest that iron-containing macromolecules and organelles cannot effectively be degraded through the lysosomal pathway due to loss of WDR45 function.
Collapse
Affiliation(s)
- Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Lena F Burbulla
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Marija Dulovic
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Simone Zittel
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johanne Heine
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Thomas Schmidt
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | | | - Ana Westenberger
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | | | | | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
1056
|
The influence of brain iron and myelin on magnetic susceptibility and effective transverse relaxation - A biochemical and histological validation study. Neuroimage 2018; 179:117-133. [DOI: 10.1016/j.neuroimage.2018.06.007] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 05/25/2018] [Accepted: 06/04/2018] [Indexed: 12/22/2022] Open
|
1057
|
Jin L, Wan W, Wang L, Wang C, Xiao J, Zhang F, Zhao J, Wang J, Zhan C, Zhong C. Elevated microRNA-520d-5p in the serum of patients with Parkinson's disease, possibly through regulation of cereloplasmin expression. Neurosci Lett 2018; 687:88-93. [PMID: 30243884 DOI: 10.1016/j.neulet.2018.09.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 12/21/2022]
Abstract
Iron metabolism dysfunction and redox-active iron-induced oxidative stress in the brain may contribute to the pathogenesis of Parkinson's disease. We have previously demonstrated that reduced serum ceruloplasmin level exacerbates nigral iron deposition in Parkinson's disease, although the underlying cause of the low serum ceruloplasmin level in Parkinson's disease remains unknown. Fluorescent quantitative real-time polymerase chain reaction analysis revealed that patients with Parkinson's disease had higher serum levels of microRNA (miR)-520d-5p than controls (p = 0.0011). Patients with Alzheimer's disease or multiple system atrophy did not have significantly elevated miR-520d-5p levels. Expression of miR-520d-5p did not correlate with disease severity or the motor phenotype of Parkinson's disease. Luciferase assays confirmed that miR-520d-5p was associated with ceruloplasmin gene expression, as predicted by the TargetScan tool and miRBase. In vitro experiments showed that miR-520d-5p reduced ceruloplasmin gene expression in the U251 astrocyte cell line. Our data suggest that miR-520d-5p may be a potential regulator of ceruloplasmin gene expression in vitro.
Collapse
Affiliation(s)
- Lirong Jin
- Department of Neurology, Zhongshan Hospital, Fudan University, China.
| | - Wenbin Wan
- Department of Neurology, Zhongshan Hospital, Fudan University, China
| | - Lingyan Wang
- Biomedical Research Center, Zhongshan Hospital, Fudan University, China
| | - Changpeng Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, China
| | - Jianqiu Xiao
- School of Life Sciences, Fudan University, China
| | - Feng Zhang
- School of Life Sciences, Fudan University, China
| | - Jue Zhao
- Department of Neurology, Huashan Hospital, Fudan University, China
| | - Jian Wang
- Department of Neurology, Huashan Hospital, Fudan University, China
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Chunjiu Zhong
- Department of Neurology, Zhongshan Hospital, Fudan University, China.
| |
Collapse
|
1058
|
Pamphlett R, Bishop DP, Kum Jew S, Doble PA. Age-related accumulation of toxic metals in the human locus ceruleus. PLoS One 2018; 13:e0203627. [PMID: 30231068 PMCID: PMC6145836 DOI: 10.1371/journal.pone.0203627] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/23/2018] [Indexed: 12/28/2022] Open
Abstract
Damage to the locus ceruleus has been implicated in the pathogenesis of a number of neurological conditions. Locus ceruleus neurons accumulate toxic metals such as mercury selectively, however, the presence of toxic metals in locus ceruleus neurons of people of different ages, and with a variety of disorders, is not known. To demonstrate at what age toxic metals are first detectable in the locus ceruleus, and to evaluate whether their presence is more common in certain clinicopathological conditions, we looked for these metals in 228 locus ceruleus samples. Samples were taken at coronial autopsies from individuals with a wide range of ages, pre-existing conditions and causes of death. Paraffin sections of pons containing the locus ceruleus were stained with silver nitrate autometallography, which indicates inorganic mercury, silver and bismuth within cells (termed autometallography-detected toxic metals, or AMG™). No locus ceruleus AMG neurons were seen in 38 individuals aged under 20 years. 47% of the 190 adults (ie, aged 20 years and over) had AMG locus ceruleus neurons. The proportion of adults with locus ceruleus AMG neurons increased during aging, except for a decreased proportion in the 90-plus years age group. No differences were found in the proportions of locus ceruleus AMG neurons between groups with different neurological, psychiatric, or other clinicopathological conditions, or among various causes of death. Elemental analysis with laser ablation-inductively coupled plasma-mass spectrometry was used to cross-validate the metals detected by AMG, by looking for silver, gold, bismuth, cadmium, chromium, iron, mercury, nickel, and lead in the locus ceruleus of ten individuals. This confirmed the presence of mercury in locus ceruleus samples containing AMG neurons, and showed cadmium, silver, lead, iron, and nickel in the locus ceruleus of some individuals. In conclusion, toxic metals stained by AMG (most likely inorganic mercury) appear in locus ceruleus neurons in early adult life. About half of adults in this study had locus ceruleus neurons containing inorganic mercury, and elemental analysis found a range of other toxic metals in the locus ceruleus. Locus ceruleus inorganic mercury increased during aging, except for a decrease in advanced age, but was not found more often in any single clinicopathological condition or cause of death.
Collapse
Affiliation(s)
- Roger Pamphlett
- Discipline of Pathology, Sydney Medical School, Brain and Mind Centre, The University of Sydney, New South Wales, Australia
- Department of Neuropathology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - David P. Bishop
- Elemental Bio-imaging Facility, University of Technology, Sydney, New South Wales, Australia
| | - Stephen Kum Jew
- Discipline of Pathology, Sydney Medical School, Brain and Mind Centre, The University of Sydney, New South Wales, Australia
| | - Philip A. Doble
- Elemental Bio-imaging Facility, University of Technology, Sydney, New South Wales, Australia
| |
Collapse
|
1059
|
Ide K, Matsuoka N, Yamada H, Furushima D, Kawakami K. Effects of Tea Catechins on Alzheimer's Disease: Recent Updates and Perspectives. Molecules 2018; 23:molecules23092357. [PMID: 30223480 PMCID: PMC6225145 DOI: 10.3390/molecules23092357] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/10/2018] [Accepted: 09/13/2018] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the most common neurodegenerative disorders worldwide. Its incidence is gradually increasing because of an aging demographic. Therefore, AD prevention and modification is important to improve the health status of older adults. Oxidative stress is a component of the pathological mechanisms underlying AD. It is caused by a disruption of the balance between reactive oxygen species and antioxidant molecules. This imbalance also causes neuroinflammation. Catechins, which are bioactive components of tea, have antioxidative and anti-inflammatory effects. Moreover, other potential properties related to AD prevention and modification have been reported in in vitro and in vivo studies. Several clinical studies have also been conducted to date. The current review summarizes recent updates and perspectives of the effects of catechins on AD based on the molecular mechanisms and related clinical studies.
Collapse
Affiliation(s)
- Kazuki Ide
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto 606-8501, Japan.
- Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501, Japan.
- Department of Drug Evaluation and Informatics, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan.
| | - Norihiro Matsuoka
- Jyoto Hospital, 11-22 Hanatenhigashi 2-chome, Tsurumi-ku Osaka-shi, Osaka 538-0044, Japan.
| | - Hiroshi Yamada
- Department of Drug Evaluation and Informatics, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan.
| | - Daisuke Furushima
- Department of Drug Evaluation and Informatics, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan.
| | - Koji Kawakami
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto 606-8501, Japan.
- Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
1060
|
Liu JL, Fan YG, Yang ZS, Wang ZY, Guo C. Iron and Alzheimer's Disease: From Pathogenesis to Therapeutic Implications. Front Neurosci 2018; 12:632. [PMID: 30250423 PMCID: PMC6139360 DOI: 10.3389/fnins.2018.00632] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/22/2018] [Indexed: 12/28/2022] Open
Abstract
As people age, iron deposits in different areas of the brain may impair normal cognitive function and behavior. Abnormal iron metabolism generates hydroxyl radicals through the Fenton reaction, triggers oxidative stress reactions, damages cell lipids, protein and DNA structure and function, and ultimately leads to cell death. There is an imbalance in iron homeostasis in Alzheimer's disease (AD). Excessive iron contributes to the deposition of β-amyloid and the formation of neurofibrillary tangles, which in turn, promotes the development of AD. Therefore, iron-targeted therapeutic strategies have become a new direction. Iron chelators, such as desferoxamine, deferiprone, deferasirox, and clioquinol, have received a great deal of attention and have obtained good results in scientific experiments and some clinical trials. Given the limitations and side effects of the long-term application of traditional iron chelators, alpha-lipoic acid and lactoferrin, as self-synthesized naturally small molecules, have shown very intriguing biological activities in blocking Aβ-aggregation, tauopathy and neuronal damage. Despite a lack of evidence for any clinical benefits, the conjecture that therapeutic chelation, with a special focus on iron ions, is a valuable approach for treating AD remains widespread.
Collapse
Affiliation(s)
- Jun-Lin Liu
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yong-Gang Fan
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Zheng-Sheng Yang
- Department of Dermatology, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Zhan-You Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, China.,Key Laboratory of Medical Cell Biology of Ministry of Education, Institute of Health Sciences, China Medical University, Shenyang, China
| | - Chuang Guo
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| |
Collapse
|
1061
|
da Silva VK, de Freitas BS, Garcia RCL, Monteiro RT, Hallak JE, Zuardi AW, Crippa JAS, Schröder N. Antiapoptotic effects of cannabidiol in an experimental model of cognitive decline induced by brain iron overload. Transl Psychiatry 2018; 8:176. [PMID: 30177808 PMCID: PMC6120904 DOI: 10.1038/s41398-018-0232-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/03/2018] [Accepted: 07/24/2018] [Indexed: 12/26/2022] Open
Abstract
Iron accumulation in the brain has been recognized as a common feature of both normal aging and neurodegenerative diseases. Cognitive dysfunction has been associated to iron excess in brain regions in humans. We have previously described that iron overload leads to severe memory deficits, including spatial, recognition, and emotional memory impairments in adult rats. In the present study we investigated the effects of neonatal iron overload on proteins involved in apoptotic pathways, such as Caspase 8, Caspase 9, Caspase 3, Cytochrome c, APAF1, and PARP in the hippocampus of adult rats, in an attempt to establish a causative role of iron excess on cell death in the nervous system, leading to memory dysfunction. Cannabidiol (CBD), the main non-psychotropic component of Cannabis sativa, was examined as a potential drug to reverse iron-induced effects on the parameters analyzed. Male rats received vehicle or iron carbonyl (30 mg/kg) from the 12th to the 14th postnatal days and were treated with vehicle or CBD (10 mg/kg) for 14 days in adulthood. Iron increased Caspase 9, Cytochrome c, APAF1, Caspase 3 and cleaved PARP, without affecting cleaved Caspase 8 levels. CBD reversed iron-induced effects, recovering apoptotic proteins Caspase 9, APAF1, Caspase 3 and cleaved PARP to the levels found in controls. These results suggest that iron can trigger cell death pathways by inducing intrinsic apoptotic proteins. The reversal of iron-induced effects by CBD indicates that it has neuroprotective potential through its anti-apoptotic action.
Collapse
Affiliation(s)
- Vanessa Kappel da Silva
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil
| | - Betânia Souza de Freitas
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| | - Rebeca Carvalho Lacerda Garcia
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| | - Ricardo Tavares Monteiro
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| | - Jaime Eduardo Hallak
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, São Paulo, 14048-900, Brazil
| | - Antônio Waldo Zuardi
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, São Paulo, 14048-900, Brazil
| | - José Alexandre S Crippa
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, São Paulo, 14048-900, Brazil
| | - Nadja Schröder
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil.
- Departamento de Fisiologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90050-170, Brazil.
| |
Collapse
|
1062
|
Willkommen D, Lucio M, Schmitt-Kopplin P, Gazzaz M, Schroeter M, Sigaroudi A, Michalke B. Species fractionation in a case-control study concerning Parkinson's disease: Cu-amino acids discriminate CSF of PD from controls. J Trace Elem Med Biol 2018; 49:164-170. [PMID: 29472131 DOI: 10.1016/j.jtemb.2018.01.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/02/2018] [Accepted: 01/17/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Parkinson's disease is affecting about 1% of the population above 65 years. Improvements in medicine support prolonged lifetime which increases the total concentration of humans affected by the disease. It is suggested that occupational and environmental exposure to metals like iron (Fe), manganese (Mn), copper (Cu) and zinc (Zn) can influence the risk for Parkinson's disease. These metals play a key role as cofactors in many enzymes and proteins. METHODS In this case-control study, we investigated the Mn-, Fe-, Cu- and Zn-species in cerebrospinal fluid (CSF) by size-exclusion chromatography hyphenated to inductively coupled plasma mass spectrometry (SEC-ICP-MS) and the total concentration of these metals by inductively coupled plasma sector field mass spectrometry (ICP-sf-MS). RESULTS The investigation of total metal concentration and speciation provided only minor changes, but it produced strong significance for a number of ratios. The analysis revealed a strong change in the ratio between total concentration of Fe and the amino acid-fraction of Cu. This could be observed when analyzing both the respective element concentrations of the fraction (which also depends on individual variation of the total element concentration) as well as when being expressed as percentage of total concentration (normalization) which more clearly shows changes of distribution pattern independent of individual variation of total element concentrations. CONCLUSION Speciation analysis, therefore, is a powerful technique to investigate changes in a case-control study where ratios of different species play an important role.
Collapse
Affiliation(s)
- Desiree Willkommen
- Helmholtz Zentrum München, Analytical Biogeochemistry, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
| | - Marianna Lucio
- Helmholtz Zentrum München, Analytical Biogeochemistry, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Philippe Schmitt-Kopplin
- Helmholtz Zentrum München, Analytical Biogeochemistry, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany; TU München, Lehrstuhl für Analytische Lebensmittelchemie, Wissenschaftszentrum Weihenstephan, Alte Akademie 10, 85354 Freising, Germany
| | - Malaz Gazzaz
- Uniklinik Köln, Institut I für Pharmakologie, Zentrum für Pharmakologie, Gleueler Straße 24, 50931 Köln, Germany
| | - Michael Schroeter
- Uniklinik Köln, Klinik und Poliklinik für Neurologie und Psychiatrie, Kerpener Str. 62, 50924 Köln, Germany
| | - Ali Sigaroudi
- Uniklinik Köln, Institut I für Pharmakologie, Zentrum für Pharmakologie, Gleueler Straße 24, 50931 Köln, Germany; Universitätsspital Zürich, Klinik für Klinische Pharmakologie und Toxikologie, Rämistraße 100, 8091 Zürich, Switzerland
| | - Bernhard Michalke
- Helmholtz Zentrum München, Analytical Biogeochemistry, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| |
Collapse
|
1063
|
da Cruz Fernandes M, Severino AG, Zago AM, Tortorelli L, Carvalho FB, Schneider FL. Iron localization in the guinea pig choroid plexus: A light and transmission electron microscopy study. J Trace Elem Med Biol 2018; 49:128-133. [PMID: 29895361 DOI: 10.1016/j.jtemb.2018.05.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 05/04/2018] [Accepted: 05/07/2018] [Indexed: 10/16/2022]
Abstract
The role of the choroid plexus (CP) in iron (Fe) homeostasis has been suggested as the main mechanism of Fe uptake and storage in the mammalian central nervous system. Thus, the CP of the lateral and fourth ventricles was studied in guinea pigs with light and electron microscopy using methods including Perls' Prussian blue and Gomori acid phosphatase staining, immunoreactivity for ferritin and transferrin, as well as energy dispersive spectrometry microanalysis. The present study reveals the presence of endogenous Fe in CP epithelial cells. Under light microscopy, Prussian blue staining revealed dark blue precipitates (i.e., Fe3+) with a preferentially perinuclear localization. The Fe was also positive for such granules with similar cellular localization. Ultrastructural analysis demonstrated the presence of dense bodies and siderosomes with molecular ferritin. The spectra obtained by the microanalysis demonstrated emissions for Fe, both in dense bodies and siderosomes. This study suggests that guinea pig CP epithelial cells accumulate Fe in the form of ferritin, possibly in cytoplasmic organelles such as lysosomes.
Collapse
Affiliation(s)
- Marilda da Cruz Fernandes
- Laboratório de Pesquisa em Patologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Sarmento Leite, 245, CEP 90050-170, Porto Alegre, RS, Brazil.
| | - Antônio Generoso Severino
- Laboratório de Neuroanatomia, Universidade Federal do Rio Grande do Sul, Sarmento Leite, 500, CEP 90050-170, Porto Alegre, RS, Brazil
| | - Adriana Maria Zago
- Laboratório de Pesquisa em Patologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Sarmento Leite, 245, CEP 90050-170, Porto Alegre, RS, Brazil
| | - Lucas Tortorelli
- Laboratório de Pesquisa em Patologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Sarmento Leite, 245, CEP 90050-170, Porto Alegre, RS, Brazil
| | - Fabiano B Carvalho
- Laboratório de Pesquisa em Patologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Sarmento Leite, 245, CEP 90050-170, Porto Alegre, RS, Brazil
| | - Felipe Luis Schneider
- Laboratório de Neuroanatomia, Universidade Federal do Rio Grande do Sul, Sarmento Leite, 500, CEP 90050-170, Porto Alegre, RS, Brazil
| |
Collapse
|
1064
|
Zhang Q, Li C, Zhang T, Ge Y, Han X, Sun S, Ding J, Lu M, Hu G. Deletion of Kir6.2/SUR1 potassium channels rescues diminishing of DA neurons via decreasing iron accumulation in PD. Mol Cell Neurosci 2018; 92:164-176. [PMID: 30171894 DOI: 10.1016/j.mcn.2018.08.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 07/16/2018] [Accepted: 08/25/2018] [Indexed: 11/25/2022] Open
Abstract
ATP-sensitive potassium (K-ATP) channels express in the central nervous system extensively which coupling cell metabolism and cellular electrical activity. K-ATP channels in mature substantia nigra (SN) dopaminergic (DA) neurons are composed of inwardly rectifying potassium channel (Kir) subunit 6.2 and sulfonylurea receptor 1 (SUR1). Our previous study revealed that regulating K-ATP channel exerts the protective effect on DA neurons in a mouse model of Parkinson's disease (PD). However, the detailed mechanism underlying the role of Kir6.2/K-ATP remains unclear. In the present study, we found the deletion of Kir6.2 dramatically alleviated PD-like motor dysfunction of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) PD model. We further found that Kir6.2 knockout selectively restored the reduction of both DA neuronal number and dopamine transmitter level in the nigrostriatal of MPTP-treated PD mice. To gain some understanding on the molecular basis of this effect, we focused on the regulation of Kir6.2 deletion on iron metabolism which is tightly associated with DA neuron damage. We found that Kir6.2 knockout suppressed the excessive iron accumulation in MPTP-treated mouse midbrain and inhibited the upregulation of ferritin light chain (FTL), which is a main intracellular iron storage protein. We probed further and found out that the deletion of Kir6.2 inhibited the excessive production of FTL via IRP-IRE regulatory system, and thereby protecting SN DA neurons against MPTP challenge. Our findings suggest that Kir6.2 plays a crucial role in the pathogenesis of PD and regulating Kir6.2/K-ATP channel may be a promising strategy for PD treatment.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Pharmacology, School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Chengwu Li
- Department of Pharmacology, School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Ting Zhang
- Department of Pharmacology, School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Yaping Ge
- Department of Pharmacology, School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Xiaojuan Han
- Department of Pharmacology, School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Sifan Sun
- First Clinic Medical School, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Jianhua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China.
| | - Gang Hu
- Department of Pharmacology, School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China; Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China.
| |
Collapse
|
1065
|
Abstract
Appraising success in meeting the world's nutritional needs has largely focused on infant mortality and anthropometric measurements with an emphasis on the first 1,000 days (conception to approximately age 2 years). This ignores the unique nutritional needs of the human brain. Although the intrauterine environment and the early postnatal years are important, equally critical periods follow during which the brain's intricate wiring is established for a lifetime of experience-driven remodeling. At the peak of this process during childhood, the human brain may account for 50% of the body's basal nutritional requirement. Thus, the consequences of proper nutritional management of the brain play out over a lifetime. Our motivation in preparing this review was to move the human brain into a more central position in the planning of nutritional programs. Here we review the macro- and micronutrient requirements of the human brain and how they are delivered, from conception to adulthood.
Collapse
Affiliation(s)
- Manu S. Goyal
- Mallinckrodt Institute of Radiology and Department of Neurology, Washington University School of Medicine, Washington University, St. Louis, Missouri 63130, USA
| | - Lora L. Iannotti
- Brown School, Institute for Public Health, Washington University, St. Louis, Missouri 63130, USA
| | - Marcus E. Raichle
- Mallinckrodt Institute of Radiology and Department of Neurology, Washington University School of Medicine, Washington University, St. Louis, Missouri 63130, USA
| |
Collapse
|
1066
|
Alcalde LA, de Freitas BS, Machado GDB, de Freitas Crivelaro PC, Dornelles VC, Gus H, Monteiro RT, Kist LW, Bogo MR, Schröder N. Iron chelator deferiprone rescues memory deficits, hippocampal BDNF levels and antioxidant defenses in an experimental model of memory impairment. Biometals 2018; 31:927-940. [PMID: 30117045 DOI: 10.1007/s10534-018-0135-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/12/2018] [Indexed: 12/18/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) plays a key role in neural development and physiology, as well as in pathological states. Post-mortem studies demonstrate that BDNF is reduced in the brains of patients affected by neurodegenerative diseases. Iron accumulation has also been associated to the pathogenesis of neurodegenerative diseases. In rats, iron overload induces persistent memory deficits, increases oxidative stress and apoptotic markers, and decreases the expression of the synaptic marker, synaptophysin. Deferiprone (DFP) is an oral iron chelator used for the treatment of systemic iron overload disorders, and has recently been tested for Parkinson's disease. Here, we investigated the effects of iron overload on BDNF levels and on mRNA expression of genes encoding TrkB, p75NTR, catalase (CAT) and NQO1. We also aimed at investigating the effects of DFP on iron-induced impairments. Rats received iron or vehicle at postnatal days 12-14 and when adults, received chronic DFP or water (vehicle). Recognition memory was tested 19 days after the beginning of chelation therapy. BDNF measurements and expression analyses in the hippocampus were performed 24 h after the last day of DFP treatment. DFP restored memory and increased hippocampal BDNF levels, ameliorating iron-induced effects. Iron overload in the neonatal period reduced, while treatment with DFP was able to rescue, the expression of antioxidant enzymes CAT and NQO1.
Collapse
Affiliation(s)
- Luisa Azambuja Alcalde
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90619-900, Brazil
| | - Betânia Souza de Freitas
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90619-900, Brazil
| | - Gustavo Dalto Barroso Machado
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90619-900, Brazil
| | - Pedro Castilhos de Freitas Crivelaro
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90619-900, Brazil
| | - Victoria Campos Dornelles
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90619-900, Brazil
| | - Henrique Gus
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90619-900, Brazil
| | - Ricardo Tavares Monteiro
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90619-900, Brazil
| | - Luiza Wilges Kist
- Laboratory of Genomics and Molecular Biology, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90619-900, Brazil.,Graduate Program in Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90610-000, Brazil
| | - Mauricio Reis Bogo
- Laboratory of Genomics and Molecular Biology, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90619-900, Brazil.,Graduate Program in Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, 90610-000, Brazil
| | - Nadja Schröder
- Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, Porto Alegre, RS, 90050-170, Brazil. .,National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, 71605-001, Brazil.
| |
Collapse
|
1067
|
Pesch B, Casjens S, Weiss T, Kendzia B, Arendt M, Eisele L, Behrens T, Ulrich N, Pundt N, Marr A, Robens S, Van Thriel C, Van Gelder R, Aschner M, Moebus S, Dragano N, Brüning T, Jöckel KH. Occupational Exposure to Manganese and Fine Motor Skills in Elderly Men: Results from the Heinz Nixdorf Recall Study. Ann Work Expo Health 2018; 61:1118-1131. [PMID: 29136419 DOI: 10.1093/annweh/wxx076] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 08/23/2017] [Indexed: 12/22/2022] Open
Abstract
Objectives Exposure to manganese (Mn) may cause movement disorders, but less is known whether the effects persist after the termination of exposure. This study investigated the association between former exposure to Mn and fine motor deficits in elderly men from an industrial area with steel production. Methods Data on the occupational history and fine motor tests were obtained from the second follow-up of the prospective Heinz Nixdorf Recall Study (2011-2014). The study population included 1232 men (median age 68 years). Mn in blood (MnB) was determined in archived samples (2000-2003). The association between Mn exposure (working as welder or in other at-risk occupations, cumulative exposure to inhalable Mn, MnB) with various motor functions (errors in line tracing, steadiness, or aiming and tapping hits) was investigated with Poisson and logistic regression, adjusted for iron status and other covariates. Odds ratios (ORs) with 95% confidence intervals (CIs) were estimated for substantially impaired dexterity (errors >90th percentile, tapping hits <10th percentile). Results The median of cumulative exposure to inhalable Mn was 58 µg m-3 years in 322 men who ever worked in at-risk occupations. Although we observed a partly better motor performance of exposed workers at group level, we found fewer tapping hits in men with cumulative Mn exposure >184.8 µg m-3 years (OR 2.15, 95% CI 1.17-3.94). MnB ≥ 15 µg l-1, serum ferritin ≥ 400 µg l-1, and gamma-glutamyl transferase ≥74 U l-1 were associated with a greater number of errors in line tracing. Conclusions We found evidence that exposure to inhalable Mn may carry a risk for dexterity deficits. Whether these deficits can be exclusively attributed to Mn remains to be elucidated, as airborne Mn is strongly correlated with iron in metal fumes, and high ferritin was also associated with errors in line tracing. Furthermore, hand training effects must be taken into account when testing for fine motor skills.
Collapse
Affiliation(s)
- Beate Pesch
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-Universität Bochum (IPA), Bochum, Germany
| | - Swaantje Casjens
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-Universität Bochum (IPA), Bochum, Germany
| | - Tobias Weiss
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-Universität Bochum (IPA), Bochum, Germany
| | - Benjamin Kendzia
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-Universität Bochum (IPA), Bochum, Germany
| | - Marina Arendt
- Institute for Medical Informatics, Biometry and Epidemiology (IMIBE), University of Essen-Duisburg, Essen, Germany
| | - Lewin Eisele
- Institute for Medical Informatics, Biometry and Epidemiology (IMIBE), University of Essen-Duisburg, Essen, Germany
| | - Thomas Behrens
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-Universität Bochum (IPA), Bochum, Germany
| | - Nadin Ulrich
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-Universität Bochum (IPA), Bochum, Germany
| | - Noreen Pundt
- Institute for Medical Informatics, Biometry and Epidemiology (IMIBE), University of Essen-Duisburg, Essen, Germany
| | - Anja Marr
- Institute for Medical Informatics, Biometry and Epidemiology (IMIBE), University of Essen-Duisburg, Essen, Germany
| | - Sibylle Robens
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-Universität Bochum (IPA), Bochum, Germany
| | - Christoph Van Thriel
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Rainer Van Gelder
- Institute for Occupational Safety and Health of the German Social Accident Insurance (IFA), Sankt Augustin, Germany
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY, USA
| | - Susanne Moebus
- Institute for Medical Informatics, Biometry and Epidemiology (IMIBE), University of Essen-Duisburg, Essen, Germany
| | - Nico Dragano
- Institute for Medical Sociology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Thomas Brüning
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-Universität Bochum (IPA), Bochum, Germany
| | - Karl-Heinz Jöckel
- Institute for Medical Informatics, Biometry and Epidemiology (IMIBE), University of Essen-Duisburg, Essen, Germany
| |
Collapse
|
1068
|
Tang M, Chen Z, Wu D, Chen L. Ferritinophagy/ferroptosis: Iron-related newcomers in human diseases. J Cell Physiol 2018; 233:9179-9190. [PMID: 30076709 DOI: 10.1002/jcp.26954] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/12/2018] [Indexed: 02/06/2023]
Abstract
Nuclear receptor coactivator 4 mediated ferritinophagy is an autophagic phenomenon that specifically involves ferritin to release intracellular free iron. Ferritinophagy is implicated in maintaining efficient erythropoiesis. Notably, ferritinophagy also plays a central role in driving some pathological processes, including Parkinson's disease (PD) and urinary tract infections. Some evidence has demonstrated that ferritinophagy is critical to induce ferroptosis. Ferroptosis is a newly nonapoptotic form of cell death, characterized by the accumulation of iron-based lipid reactive oxygen species. Ferroptosis plays an important role in inhibiting some types of cancers, such as hepatocellular carcinoma, pancreatic carcinoma, prostate cancer, and breast cancer. Conversely, the activation of ferroptosis accelerates neurodegeneration diseases, including PD and Alzheimer's disease. Therefore, in this review, we summarize the regulatory mechanisms related to ferritinophagy and ferroptosis. Moreover, the distinctive effects of ferritinophagy in human erythropoiesis and some pathologies, coupled with the promotive or inhibitory role of tumorous and neurodegenerative diseases mediated by ferroptosis, are elucidated. Obviously, activating or inhibiting ferroptosis could be exploited to achieve desirable therapeutic effects on diverse cancers and neurodegeneration diseases. Interrupting ferritinophagy to control iron level might provide a potentially therapeutic avenue to suppress urinary tract infections.
Collapse
Affiliation(s)
- Mingzhu Tang
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Zhe Chen
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Di Wu
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
1069
|
Model Senescent Microglia Induce Disease Related Changes in α-Synuclein Expression and Activity. Biomolecules 2018; 8:biom8030067. [PMID: 30071596 PMCID: PMC6164966 DOI: 10.3390/biom8030067] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 12/13/2022] Open
Abstract
Aging is the most prominent risk factor for most neurodegenerative diseases. However, incorporating aging-related changes into models of neurodegeneration rarely occurs. One of the significant changes that occurs in the brain as we age is the shift in phenotype of the resident microglia population to one less able to respond to deleterious changes in the brain. These microglia are termed dystrophic microglia. In order to better model neurodegenerative diseases, we have developed a method to convert microglia into a senescent phenotype in vitro. Mouse microglia grown in high iron concentrations showed many characteristics of dystrophic microglia including, increased iron storage, increased expression of proteins, such as ferritin and the potassium channel, Kv1.3, increased reactive oxygen species production and cytokine release. We have applied this new model to the study of α-synuclein, a protein that is closely associated with a number of neurodegenerative diseases. We have shown that conditioned medium from our model dystrophic microglia increases α-synuclein transcription and expression via tumor necrosis factor alpha (TNFα) and mediated through nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). The conditioned medium also decreases the formation of α-synuclein tetramers, associated ferrireductase activity, and increases aggregates of α-synuclein. The results suggest that we have developed an interesting new model of aged microglia and that factors, including TNFα released from dystrophic microglia could have a significant influence on the pathogenesis of α-synuclein related diseases.
Collapse
|
1070
|
van Duijn S, Bulk M, van Duinen SG, Nabuurs RJA, van Buchem MA, van der Weerd L, Natté R. Cortical Iron Reflects Severity of Alzheimer's Disease. J Alzheimers Dis 2018; 60:1533-1545. [PMID: 29081415 PMCID: PMC5676973 DOI: 10.3233/jad-161143] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Abnormal iron distribution in the isocortex is increasingly recognized as an in vivo marker for Alzheimer’s disease (AD). However, the contribution of iron accumulation to the AD pathology is still poorly understood. In this study, we investigated: 1) frontal cortical iron distribution in AD and normal aging and 2) the relation between iron distribution and degree of AD pathology. We used formalin fixed paraffin embedded frontal cortex from 10 AD patients, 10 elder, 10 middle aged, and 10 young controls and visualized iron with a modified Perl’s histochemical procedure. AD and elderly subjects were not different with respect to age and sex distribution. Iron distribution in the frontal cortex was not affected by normal aging but was clearly different between AD and controls. AD showed accumulation of iron in plaques, activated microglia, and, in the most severe cases, in the mid-cortical layers along myelinated fibers. The degree of altered iron accumulations was correlated to the amount of amyloid-β plaques and tau pathology in the same block, as well as to Braak stage (p < 0.001). AD and normal aging show different iron and myelin distribution in frontal cortex. These changes appear to occur after the development of the AD pathological hallmarks. These findings may help the interpretation of high resolution in vivo MRI and suggest the potential of using changes in iron-based MRI contrast to indirectly determine the degree of AD pathology in the frontal cortex.
Collapse
Affiliation(s)
- Sara van Duijn
- Department of Pathology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Marjolein Bulk
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands.,Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands.,Percuros BV, Leiden, The Netherlands
| | - Sjoerd G van Duinen
- Department of Pathology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Rob J A Nabuurs
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Louise van der Weerd
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands.,Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Remco Natté
- Department of Pathology, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
1071
|
Hosking DE, Ayton S, Beckett N, Booth A, Peters R. More evidence is needed. Iron, incident cognitive decline and dementia: a systematic review. Ther Adv Chronic Dis 2018; 9:241-256. [PMID: 30719269 DOI: 10.1177/2040622318788485] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/07/2018] [Indexed: 12/22/2022] Open
Abstract
Background Our aim was to systematically review the relationship between iron and incident cognitive decline or dementia from midlife onwards. Methods Systematic review of eligible studies using Medline, Embase and PsycINFO® for the period from 1 January 1986 to 2 December 2016 (CRD42016023800), where study populations had a mean age of over 50 years and were free of cognitive impairment or dementia at baseline. Two authors independently extracted data according to eligibility criteria and assessed study characteristics, quality and outcomes. Disagreement was resolved by discussion. Results A total of 1185 relevant records were identified with 12 full-text articles eligible for review. Six studies were excluded, leaving six texts to be included. Sample size ranged from 90 to 7173, with an average follow up of approximately 11.5 years. Baseline iron measures included brain iron (n = 2), iron-related biomarkers in blood and plasma (n = 2), and iron intake estimates from dietary records (n = 2). Outcomes were dementia incidence (n = 2) and longitudinal outcomes on neuropsychological tests (n = 4). Bias was evident across studies in one or more of the following: recruitment, iron exposure, outcome assessments, potential confounders, missing data or attrition. Conclusions Diversity across the small number of identified studies precludes conclusions regarding the role of iron in cognitive decline or dementia. Our review highlights substantial gaps in the evidence base and the need for more comprehensive, higher quality studies in this area.
Collapse
Affiliation(s)
- Diane E Hosking
- Centre for Research into Ageing Health and Wellbeing, Australian National University, Canberra, Australia
| | - Scott Ayton
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Nigel Beckett
- Care of the Elderly, Imperial College London, London, UK
| | - Andrew Booth
- School of Health and Related Research, University of Sheffield, Sheffield, UK
| | - Ruth Peters
- Lifecourse Ageing Research Centre, Neuroscience Research Australia, University of New South Wales, Sydney NSW 2031, Australia
| |
Collapse
|
1072
|
Aaseth J, Dusek P, Roos PM. Prevention of progression in Parkinson's disease. Biometals 2018; 31:737-747. [PMID: 30030679 PMCID: PMC6133181 DOI: 10.1007/s10534-018-0131-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 07/11/2018] [Indexed: 01/01/2023]
Abstract
Environmental influences affecting genetically susceptible individuals seem to contribute significantly to the development of Parkinson’s disease (PD). Xenobiotic exposure including transitional metal deposition into vulnerable CNS regions appears to interact with PD genes. Such exposure together with mitochondrial dysfunction evokes a destructive cascade of biochemical events, including oxidative stress and degeneration of the sensitive dopamine (DA) production system in the basal ganglia. Recent research indicates that the substantia nigra degeneration can be decelerated by treatment with iron binding compounds such as deferiprone. Interestingly compounds known to decrease PD risk including caffeine, niacin, nicotine and salbutamol also possess iron binding properties. Adequate function of antioxidative mechanisms in the vulnerable brain cells can be restored by acetylcysteine supplementation to normalize intracellular glutathione activity. Other preventive measures to reduce deterioration of dopaminergic neurons may involve life-style changes such as intake of natural antioxidants and physical exercise. Further research is recommended to identify therapeutic targets of the proposed interventions, in particular protection of the DA biosynthesis by oxygen radical scavengers and iron binding agents.
Collapse
Affiliation(s)
- Jan Aaseth
- Research Department, Innlandet Hospital Trust, Brumunddal, Norway.,Inland Norway University of Applied Sciences, Elverum, Norway
| | - Petr Dusek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University, Praha 2, Czech Republic.,General University Hospital in Prague, Prague, Czech Republic.,Department of Radiology, First Faculty of Medicine, Charles University, Praha 2, Czech Republic
| | - Per M Roos
- Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden. .,Department of Clinical Physiology, Capio St. Görans Hospital, Stockholm, Sweden.
| |
Collapse
|
1073
|
Helgudottir SS, Lichota J, Burkhart A, Moos T. Hepcidin Mediates Transcriptional Changes in Ferroportin mRNA in Differentiated Neuronal-Like PC12 Cells Subjected to Iron Challenge. Mol Neurobiol 2018; 56:2362-2374. [DOI: 10.1007/s12035-018-1241-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 07/11/2018] [Indexed: 01/01/2023]
|
1074
|
Oxidative stress and neurodegeneration: the involvement of iron. Biometals 2018; 31:715-735. [PMID: 30014355 DOI: 10.1007/s10534-018-0126-2] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/04/2018] [Indexed: 12/14/2022]
Abstract
Many evidences indicate that oxidative stress plays a significant role in a variety of human disease states, including neurodegenerative diseases. Iron is an essential metal for almost all living organisms due to its involvement in a large number of iron-containing proteins and enzymes, though it could be also toxic. Actually, free iron excess generates oxidative stress, particularly in brain, where anti-oxidative defences are relatively low. Its accumulation in specific regions is associated with pathogenesis in a variety of neurodegenerative diseases (i.e., Parkinson's disease, Alzheimer's disease, Huntington's chorea, Amyotrophic Lateral Sclerosis and Neurodegeneration with Brain Iron Accumulation). Anyway, the extent of toxicity is dictated, in part, by the localization of the iron complex within the cell (cytosolic, lysosomal and mitochondrial), its biochemical form, i.e., ferritin or hemosiderin, as well as the ability of the cell to prevent the generation and propagation of free radical by the wide range of antioxidants and cytoprotective enzymes in the cell. Particularly, ferrous iron can act as a catalyst in the Fenton reaction that potentiates oxygen toxicity by generating a wide range of free radical species, including hydroxyl radicals (·OH). The observation that patients with neurodegenerative diseases show a dramatic increase in their brain iron content, correlated with the production of reactive oxigen species in these areas of the brain, conceivably suggests that disturbances in brain iron homeostasis may contribute to the pathogenesis of these disorders. The aim of this review is to describe the chemical features of iron in human beings and iron induced toxicity in neurodegenerative diseases. Furthermore, the attention is focused on metal chelating drugs therapeutic strategies.
Collapse
|
1075
|
Varga E, Pandur E, Abrahám H, Horváth A, Ács P, Komoly S, Miseta A, Sipos K. Cuprizone Administration Alters the Iron Metabolism in the Mouse Model of Multiple Sclerosis. Cell Mol Neurobiol 2018; 38:1081-1097. [PMID: 29464444 PMCID: PMC11482029 DOI: 10.1007/s10571-018-0578-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/13/2018] [Indexed: 01/01/2023]
Abstract
Cuprizone (CZ) is a widely used copper chelating agent to develop non-autoimmune animal model of multiple sclerosis, characterized by demyelination of the corpus callosum (CC) and other brain regions. The exact mechanisms of CZ action are still arguable, but it seems that the only affected cells are the mature oligodendrocytes, possibly via metabolic disturbances caused by copper deficiency. During the pathogenesis of multiple sclerosis, high amount of deposited iron can be found throughout the demyelinated areas of the brain in the form of extracellular iron deposits and intracellularly accumulated iron in microglia. In the present study, we used the accepted experimental model of 0.2% CZ-containing diet with standard iron concentration to induce demyelination in the brain of C57BL/6 mice. Our aim was to examine the changes of iron homeostasis in the CC and as a part of the systemic iron regulation, in the liver. Our data showed that CZ treatment changed the iron metabolism of both tissues; however, it had more impact on the liver. Besides the alterations in the expressions of iron storage and import proteins, we detected reduced serum iron concentration and iron stores in the liver, together with elevated hepcidin levels and feasible disturbances in the Fe-S cluster biosynthesis. Our results revealed that the CZ-containing diet influences the systemic iron metabolism in mice, particularly the iron homeostasis of the liver. This inadequate systemic iron regulation may affect the iron homeostasis of the brain, eventually indicating a relationship among CZ treatment, iron metabolism, and neurodegeneration.
Collapse
Affiliation(s)
- E Varga
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus str. 2., Pécs, 7624, Hungary
| | - E Pandur
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus str. 2., Pécs, 7624, Hungary
| | - H Abrahám
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, Szigeti str. 12., Pécs, 7624, Hungary
| | - A Horváth
- Department of Neurosurgery, Medical School, University of Pécs, Rét str. 2., Pécs, 7623, Hungary
| | - P Ács
- Department of Neurology, Medical School, University of Pécs, Rét str. 2., Pécs, 7623, Hungary
| | - S Komoly
- Department of Neurology, Medical School, University of Pécs, Rét str. 2., Pécs, 7623, Hungary
| | - A Miseta
- Department of Laboratory Medicine, Medical School, University of Pécs, Ifjúság str. 13., Pécs, 7624, Hungary
| | - K Sipos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus str. 2., Pécs, 7624, Hungary.
- , Szigeti str. 12., Pécs, 7624, Hungary.
| |
Collapse
|
1076
|
Lee DG, Kam MK, Kim KM, Kim HS, Kwon OS, Lee HS, Lee DS. Peroxiredoxin 5 prevents iron overload-induced neuronal death by inhibiting mitochondrial fragmentation and endoplasmic reticulum stress in mouse hippocampal HT-22 cells. Int J Biochem Cell Biol 2018; 102:10-19. [PMID: 29906559 DOI: 10.1016/j.biocel.2018.06.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/01/2018] [Accepted: 06/11/2018] [Indexed: 01/18/2023]
Abstract
Iron is an essential element for neuronal as well as cellular functions. However, Iron overload has been known to cause neuronal toxicity through mitochondrial fission, dysregulation of Ca2+, endoplasmic reticulum (ER) stress, and reactive oxygen species (ROS) production. Nevertheless, the precise mechanisms of iron-induced oxidative stress and mitochondria- and ER-related iron toxicity in neuronal cells are not fully understood. In this study, we demonstrated that iron overload induces ROS production earlier in the ER than in the mitochondria, and peroxiredoxin 5 (Prx5), which is a kind of antioxidant induced by iron overload, prevents iron overload-induced mitochondrial fragmentation mediated by contact with ER and translocation of Drp1, by inhibiting ROS production and calcium/calcineurin pathway in HT-22 mouse hippocampal neuronal cells. Moreover, Prx5 also prevented iron overload-induced ER-stress and cleavage of caspase-3, which consequently attenuated neuronal cell death. Therefore, we suggested that iron overload induces oxidative stress in the ER earlier than in the mitochondria, thereby increasing ER stress and calcium levels, and consequently causing mitochondrial fragmentation and neuronal cell death. So we thought that this study is essential for understanding iron toxicity in neurons, and Prx5 may serve as a new therapeutic target to prevent iron overload-induced diseases and neurodegenerative disorders.
Collapse
Affiliation(s)
- Dong Gil Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Min Kyoung Kam
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Kyung Min Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Han Seop Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Oh-Shin Kwon
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Hyun-Shik Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Dong-Seok Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
1077
|
Gai Y, Zhao X, Chen Y, Yang S, Xia X, Liu S, Wan X, Xiong K. A cyano-bridged Cu(i)-based organic framework coupled with the C-C bond cleavage of acetonitrile for selective and sensitive sensing of Fe 3+ ions. Dalton Trans 2018; 47:6888-6892. [PMID: 29708255 DOI: 10.1039/c8dt00979a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A cyano-bridged Cu(i) organic framework based on [Cu6(CN)6]n was synthesized, in which cyano anions were generated in situ from the C-C bond cleavage of acetonitrile. The as-synthesized compound displays orange-red luminescence and is further proven to be a promising Fe3+ luminescent sensor with high selectivity and sensitivity.
Collapse
Affiliation(s)
- Yanli Gai
- School of Chemistry and Materials Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
1078
|
Shen Y, Li X, Dong D, Zhang B, Xue Y, Shang P. Transferrin receptor 1 in cancer: a new sight for cancer therapy. Am J Cancer Res 2018; 8:916-931. [PMID: 30034931 PMCID: PMC6048407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 05/29/2018] [Indexed: 06/08/2023] Open
Abstract
Iron as an important element plays crucial roles in various physiological and pathological processes. Iron metabolism behaves in systemic and cellular two levels that usually are in balance conditions. The disorders of the iron metabolism balances relate with many kinds of diseases including Alzheimer's disease, osteoporosis and various cancers. In systemic iron metabolism that is regulated by hepcidin-ferroportin axis, plasma iron is bound with transferrin (TF) which has two high-affinity binding sites for ferric iron. The generic cellular iron metabolism consists of iron intake, utilization and efflux. During the iron intake process in generic cells, transferrin receptors (TFRs) act as the most important receptor mediated controls. TFR1 and TFR2 are two subtypes of TFRs those bind with iron-transferrin complex to facilitate iron into cells. TFR1 is ubiquitously expressed on the surfaces of generic cells, whereas TFR2 is specially expressed in liver cells. TFR1 has attracted more attention than TFR2 by having diverse functions in both invertebrates and vertebrates. Recently reports showed that TFR1 involved in many kinds of diseases including anemia, neurodegenerative diseases and cancers. Most importantly, TFR1 has been verified to be abnormally expressed in various cancers. Some experimental and clinical drugs and antibodies targeting TFR1 have showed strong anti-tumor effects, herein TFR1 probably become a potential molecular target for diagnosis and treatment for cancer therapy. This paper reviewed the research progresses of the roles of TFR1 in the tumorigenesis and cancer progression, the regulations of TFR1, and the therapeutic effects of targeting TFR1 on many kinds of cancers.
Collapse
Affiliation(s)
- Ying Shen
- School of Life Science, Northwestern Polytechnical UniversityXi’an 710072, Shaanxi, China
- Research and Development Institute in Shenzhen, Northwestern Polytechnical UniversityShenzhen 518057, China
| | - Xin Li
- School of Life Science, Northwestern Polytechnical UniversityXi’an 710072, Shaanxi, China
- Research and Development Institute in Shenzhen, Northwestern Polytechnical UniversityShenzhen 518057, China
| | - Dandan Dong
- School of Life Science, Northwestern Polytechnical UniversityXi’an 710072, Shaanxi, China
- Research and Development Institute in Shenzhen, Northwestern Polytechnical UniversityShenzhen 518057, China
| | - Bin Zhang
- School of Life Science, Northwestern Polytechnical UniversityXi’an 710072, Shaanxi, China
- Research and Development Institute in Shenzhen, Northwestern Polytechnical UniversityShenzhen 518057, China
| | - Yanru Xue
- School of Life Science, Northwestern Polytechnical UniversityXi’an 710072, Shaanxi, China
- Research and Development Institute in Shenzhen, Northwestern Polytechnical UniversityShenzhen 518057, China
| | - Peng Shang
- Research and Development Institute in Shenzhen, Northwestern Polytechnical UniversityShenzhen 518057, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, School of Life Science, Northwestern Polytechnical UniversityXi’an 710072, Shaanxi, China
| |
Collapse
|
1079
|
Trist BG, Hare DJ, Double KL. A Proposed Mechanism for Neurodegeneration in Movement Disorders Characterized by Metal Dyshomeostasis and Oxidative Stress. Cell Chem Biol 2018; 25:807-816. [PMID: 29861271 DOI: 10.1016/j.chembiol.2018.05.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/09/2018] [Accepted: 05/01/2018] [Indexed: 02/07/2023]
Abstract
Shared molecular pathologies between distinct neurodegenerative disorders offer unique opportunities to identify common mechanisms of neuron death, and apply lessons learned from one disease to another. Neurotoxic superoxide dismutase 1 (SOD1) proteinopathy in SOD1-associated familial amyotrophic lateral sclerosis (fALS) is recapitulated in idiopathic Parkinson disease (PD), suggesting that these two phenotypically distinct disorders share an etiological pathway, and tractable therapeutic target(s). Despite 25 years of research, the molecular determinants underlying SOD1 misfolding and toxicity in fALS remain poorly understood. The absence of SOD1 mutations in PD highlights mounting evidence that SOD1 mutations are not the sole cause of SOD1 protein misfolding occasioning oligomerization and toxicity, reinforcing the importance of non-genetic factors, including protein metallation and post-translational modification in determining SOD1 stability and function. We propose that these non-genetic factors underlie the misfolding and dysfunction of SOD1 and other proteins in both PD and fALS, constituting a shared and tractable pathway to neurodegeneration.
Collapse
Affiliation(s)
- Benjamin Guy Trist
- Discipline of Biomedical Science and Brain and Mind Centre, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia
| | - Dominic James Hare
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia; Department of Pathology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Kay Lorraine Double
- Discipline of Biomedical Science and Brain and Mind Centre, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia.
| |
Collapse
|
1080
|
Takahashi H, Watanabe Y, Tanaka H, Mihara M, Mochizuki H, Liu T, Wang Y, Tomiyama N. Quantifying changes in nigrosomes using quantitative susceptibility mapping and neuromelanin imaging for the diagnosis of early-stage Parkinson's disease. Br J Radiol 2018; 91:20180037. [PMID: 29498540 DOI: 10.1259/bjr.20180037] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To quantify nigral changes with a focus on their spatial variation within the substantia nigra pars compacta (SNpc) for diagnosing early-stage Parkinson's disease (PD). METHODS The study participants were 18 patients with early-stage PD (PD group) and 18 healthy controls (HC group) who underwent quantitative susceptibility mapping (QSM) and neuromelanin imaging. The QSM and neuromelanin values in each whole SNpc containing the entire nigrosome and dorsolateral SNpc containing nigrosome 1 were calculated. The neuromelanin area was defined as the volume with a signal-to-noise ratio higher than that of the background region. The significance of intergroup differences in the QSM value and neuromelanin area in each SNpc region was tested. Logit (p) was used to estimate the probability of PD in relation to the QSM value and the neuromelanin area, and receiver operating characteristic analyses were performed for each value. RESULTS In both SNpc, QSM values were significantly higher and neuromelanin areas were significantly lower in the PD group compared with the HC group (p < 0.05). The respective areas under the receiver operating characteristic curve for the two groups were 0.70/0.73 for the QSM value, 0.81/0.78 for the neuromelanin area in the whole/dorsolateral SNpc, and 0.86 for logit (p) in relation to the QSM value of the dorsolateral SNpc and the neuromelanin area of the whole SNpc. CONCLUSION Comprehensive MRI assessment of the abnormality involving the nigrosomes can yield a high diagnostic performance for early-stage PD. Advances in knowledge: Focusing on spatial differences in nigral changes within the SNpc can increase the sensitivity of the detection of PD-related neurodegenerative changes.
Collapse
Affiliation(s)
- Hiroto Takahashi
- 1 Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine , Osaka , Japan
| | - Yoshiyuki Watanabe
- 1 Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine , Osaka , Japan
| | - Hisashi Tanaka
- 1 Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine , Osaka , Japan
| | - Masahito Mihara
- 2 Department of Neurology, Osaka University Graduate School of Medicine , Osaka , Japan
| | - Hideki Mochizuki
- 2 Department of Neurology, Osaka University Graduate School of Medicine , Osaka , Japan
| | - Tian Liu
- 3 Departments of Biomedical Engineering and Radiology, Cornell University, MedImageMetric LLC , New York, NY , USA
| | - Yi Wang
- 4 Departments of Radiology and Biomedical Engineering, Cornell University , New York, NY , USA
| | - Noriyuki Tomiyama
- 1 Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine , Osaka , Japan
| |
Collapse
|
1081
|
Tang X, Cai F, Ding DX, Zhang LL, Cai XY, Fang Q. Magnetic resonance imaging relaxation time in Alzheimer's disease. Brain Res Bull 2018; 140:176-189. [PMID: 29738781 DOI: 10.1016/j.brainresbull.2018.05.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/18/2018] [Accepted: 05/04/2018] [Indexed: 12/26/2022]
Abstract
The magnetic resonance imaging (MRI) relaxation time constants, T1 and T2, are sensitive to changes in brain tissue microstructure integrity. Quantitative T1 and T2 relaxation times have been proposed to serve as non-invasive biomarkers of Alzheimer's disease (AD), in which alterations are believed to not only reflect AD-related neuropathology but also cognitive impairment. In this review, we summarize the applications and key findings of MRI techniques in the context of both AD subjects and AD transgenic mouse models. Furthermore, the possible mechanisms of relaxation time alterations in AD will be discussed. Future studies could focus on relaxation time alterations in the early stage of AD, and longitudinal studies are needed to further explore relaxation time alterations during disease progression.
Collapse
Affiliation(s)
- Xiang Tang
- Department of Neurology, The First Affiliated Hospital of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| | - Feng Cai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| | - Dong-Xue Ding
- Department of Neurology, The First Affiliated Hospital of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| | - Lu-Lu Zhang
- Department of Neurology, The First Affiliated Hospital of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China
| | - Xiu-Ying Cai
- Department of Neurology, The First Affiliated Hospital of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China.
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, No. 899, Pinghai Road, Suzhou, Jiangsu 215006, China.
| |
Collapse
|
1082
|
Quantitative comparison of different iron forms in the temporal cortex of Alzheimer patients and control subjects. Sci Rep 2018; 8:6898. [PMID: 29720594 PMCID: PMC5932027 DOI: 10.1038/s41598-018-25021-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/10/2018] [Indexed: 12/25/2022] Open
Abstract
We present a quantitative study of different molecular iron forms found in the temporal cortex of Alzheimer (AD) patients. Applying the methodology we developed in our previous work, we quantify the concentrations of non-heme Fe(III) by Electron Paramagnetic Resonance (EPR), magnetite/maghemite and ferrihydrite by SQUID magnetometry, together with the MRI transverse relaxation rate [Formula: see text], to obtain a systematic view of molecular iron in the temporal cortex. Significantly higher values of [Formula: see text], a larger concentration of ferrihydrite, and a larger magnetic moment of magnetite/maghemite particles are found in the brain of AD patients. Moreover, we found correlations between the concentration of the iron detected by EPR, the concentration of the ferrihydrite mineral and the average iron loading of ferritin. We discuss these findings in the framework of iron dis-homeostasis, which has been proposed to occur in the brain of AD patients.
Collapse
|
1083
|
Specific visualization of neuromelanin-iron complex and ferric iron in the human post-mortem substantia nigra using MR relaxometry at 7T. Neuroimage 2018; 172:874-885. [DOI: 10.1016/j.neuroimage.2017.11.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/19/2017] [Accepted: 11/17/2017] [Indexed: 11/22/2022] Open
|
1084
|
Kakkar AK, Singh H, Medhi B. Old wines in new bottles: Repurposing opportunities for Parkinson's disease. Eur J Pharmacol 2018; 830:115-127. [PMID: 29689247 DOI: 10.1016/j.ejphar.2018.04.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 04/17/2018] [Accepted: 04/20/2018] [Indexed: 11/30/2022]
Abstract
Parkinson's disease (PD) is a chronic progressive neurological disorder characterized by accumulation of Lewy bodies and profound loss of substantia nigra dopaminergic neurons. PD symptomatology is now recognized to include both cardinal motor as well as clinically significant non-motor symptoms. Despite intensive research, the current understanding of molecular mechanisms underlying neurodegeneration in PD is limited and has hampered the development of novel symptomatic and disease modifying therapies. The currently available treatment options are only partially or transiently effective and fail to restore the lost dopaminergic neurons or retard disease progression. Given the escalating drug development costs, lengthening timelines and declining R&D efficiency, industry and academia are increasingly focusing on ways to repurpose existing molecules as an accelerated route for drug discovery. The field of PD therapeutics is witnessing vigorous repurposing activity supported by big data analytics, computational models, and high-throughput drug screening systems. Here we review the mechanisms, efficacy, and safety of several emerging drugs currently aspiring to be repositioned for PD pharmacotherapy.
Collapse
Affiliation(s)
- Ashish Kumar Kakkar
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India.
| | - Harmanjit Singh
- Department of Pharmacology, Government Medical College and Hospital Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
1085
|
Watson J, Lee M, Garcia-Casal MN. Consequences of Inadequate Intakes of Vitamin A, Vitamin B 12, Vitamin D, Calcium, Iron, and Folate in Older Persons. CURRENT GERIATRICS REPORTS 2018; 7:103-113. [PMID: 29721404 PMCID: PMC5918526 DOI: 10.1007/s13670-018-0241-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Purpose of Review This review broadly discusses the consequences of inadequate consumption, by deficit or excess, of selected micronutrients on the quality of life and morbidity during aging, specifically considering increases in life expectancy and the costs of care in the older persons. Recent Findings A literature review of the periods 2012 to 2018, focusing on vitamins A, B12, and D, calcium, iron and folate, was completed as these micronutrients are found to significantly affect the aging process. Causation and application of these micronutrients to disorders related to aging are controversial and mixed. This review highlights research needs and controversial points on the role of these micronutrients. Summary Micronutrient deficiencies are a common and avoidable contributor to decreased quality of life and healthcare costs in the older persons. Further research is needed to determine adequate intakes and innovative uses, including appropriate thresholds for improved health outcomes for this population.
Collapse
Affiliation(s)
- Jessica Watson
- 1Morsani College of Medicine, University of South Florida, Tampa, Florida USA
| | - Marissa Lee
- 2Loma Linda University, School of Public Health, Loma Linda, CA USA
| | - Maria Nieves Garcia-Casal
- 3Evidence and Programme Guidance Unit, Department of Nutrition for Health and Development, World Health Organization, 20 Av Appia, 1211 Geneva, Switzerland
| |
Collapse
|
1086
|
Sulzer D, Cassidy C, Horga G, Kang UJ, Fahn S, Casella L, Pezzoli G, Langley J, Hu XP, Zucca FA, Isaias IU, Zecca L. Neuromelanin detection by magnetic resonance imaging (MRI) and its promise as a biomarker for Parkinson's disease. NPJ PARKINSONS DISEASE 2018; 4:11. [PMID: 29644335 PMCID: PMC5893576 DOI: 10.1038/s41531-018-0047-3] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 03/05/2018] [Accepted: 03/08/2018] [Indexed: 11/10/2022]
Abstract
The diagnosis of Parkinson’s disease (PD) occurs after pathogenesis is advanced and many substantia nigra (SN) dopamine neurons have already died. Now that therapies to block this neuronal loss are under development, it is imperative that the disease be diagnosed at earlier stages and that the response to therapies is monitored. Recent studies suggest this can be accomplished by magnetic resonance imaging (MRI) detection of neuromelanin (NM), the characteristic pigment of SN dopaminergic, and locus coeruleus (LC) noradrenergic neurons. NM is an autophagic product synthesized via oxidation of catecholamines and subsequent reactions, and in the SN and LC it increases linearly during normal aging. In PD, however, the pigment is lost when SN and LC neurons die. As shown nearly 25 years ago by Zecca and colleagues, NM’s avid binding of iron provides a paramagnetic source to enable electron and nuclear magnetic resonance detection, and thus a means for safe and noninvasive measure in living human brain. Recent technical improvements now provide a means for MRI to differentiate between PD patients and age-matched healthy controls, and should be able to identify changes in SN NM with age in individuals. We discuss how MRI detects NM and how this approach might be improved. We suggest that MRI of NM can be used to confirm PD diagnosis and monitor disease progression. We recommend that for subjects at risk for PD, and perhaps generally for older people, that MRI sequences performed at regular intervals can provide a pre-clinical means to detect presymptomatic PD.
Collapse
Affiliation(s)
- David Sulzer
- 1Department of Psychiatry, Columbia University Medical Center , New York State Psychiatric Institute, New York, NY USA.,2Department of Neurology, Columbia University Medical Center, New York, NY USA.,3Department of Pharmacology, Columbia University Medical Center, New York, NY USA
| | - Clifford Cassidy
- 4The Royal's Institute of Mental Health Research, Affiliated with the University of Ottawa, Ottawa, ON Canada
| | - Guillermo Horga
- 1Department of Psychiatry, Columbia University Medical Center , New York State Psychiatric Institute, New York, NY USA
| | - Un Jung Kang
- 2Department of Neurology, Columbia University Medical Center, New York, NY USA
| | - Stanley Fahn
- 2Department of Neurology, Columbia University Medical Center, New York, NY USA
| | - Luigi Casella
- 5Department of Chemistry, University of Pavia, Pavia, Italy
| | - Gianni Pezzoli
- Parkinson Institute, ASST "Gaetano Pini-CTO", Milan, Italy
| | - Jason Langley
- 7Center for Advanced NeuroImaging, University of California Riverside, Riverside, CA USA
| | - Xiaoping P Hu
- 8Department of Bioengineering, University of California Riverside, Riverside, CA USA
| | - Fabio A Zucca
- 9Institute of Biomedical Technologies, National Research Council of Italy, Milan, Italy
| | - Ioannis U Isaias
- Department of Neurology, University Hospital and Julius-Maximillian-University, Wuerzburg, Germany
| | - Luigi Zecca
- 9Institute of Biomedical Technologies, National Research Council of Italy, Milan, Italy
| |
Collapse
|
1087
|
Puy V, Darwiche W, Trudel S, Gomila C, Lony C, Puy L, Lefebvre T, Vitry S, Boullier A, Karim Z, Ausseil J. Predominant role of microglia in brain iron retention in Sanfilippo syndrome, a pediatric neurodegenerative disease. Glia 2018; 66:1709-1723. [DOI: 10.1002/glia.23335] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/03/2018] [Accepted: 03/16/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Vincent Puy
- Unité INSERM U1088, CURS-Université de Picardie Jules Verne; Amiens F-80054 France
- Laboratoire de Biochimie Métabolique, CHU Amiens Picardie; Amiens F-80054 France
| | - Walaa Darwiche
- Unité INSERM U1088, CURS-Université de Picardie Jules Verne; Amiens F-80054 France
| | - Stéphanie Trudel
- Laboratoire d'Oncobiologie Moléculaire, CHU Amiens Picardie, F-80054 Amiens, France and EA4666 Lymphocyte Normal, Pathologique et Cancers (LNPC); CURS-Université de Picardie Jules Verne; Amiens F-80054 France
| | - Cathy Gomila
- Unité INSERM U1088, CURS-Université de Picardie Jules Verne; Amiens F-80054 France
- Laboratoire de Biochimie Métabolique, CHU Amiens Picardie; Amiens F-80054 France
| | - Christelle Lony
- Unité INSERM U1088, CURS-Université de Picardie Jules Verne; Amiens F-80054 France
| | - Laurent Puy
- Département de Neurologie et Laboratoire de Neuroscience Fonctionnelle EA-4559; CHU Amiens Picardie; Amiens F-80054, France
| | - Thibaud Lefebvre
- INSERM U1149, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, F-75018 Paris, France, DHU UNITY, Laboratory of Excellence, GR-Ex; Paris France
| | - Sandrine Vitry
- Unité de NeuroImmunologie Virale, Institut Pasteur; Paris F-75015 France
| | - Agnès Boullier
- Unité INSERM U1088, CURS-Université de Picardie Jules Verne; Amiens F-80054 France
- Laboratoire de Biochimie Métabolique, CHU Amiens Picardie; Amiens F-80054 France
| | - Zoubida Karim
- INSERM U1149, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, F-75018 Paris, France, DHU UNITY, Laboratory of Excellence, GR-Ex; Paris France
| | - Jérôme Ausseil
- Unité INSERM U1088, CURS-Université de Picardie Jules Verne; Amiens F-80054 France
- Laboratoire de Biochimie Métabolique, CHU Amiens Picardie; Amiens F-80054 France
| |
Collapse
|
1088
|
Shin C, Lee S, Lee JY, Rhim JH, Park SW. Non-Motor Symptom Burdens Are Not Associated with Iron Accumulation in Early Parkinson's Disease: a Quantitative Susceptibility Mapping Study. J Korean Med Sci 2018; 33:e96. [PMID: 29573246 PMCID: PMC5865060 DOI: 10.3346/jkms.2018.33.e96] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 01/21/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Quantitative susceptibility mapping (QSM) has been used to measure iron accumulation in the deep nuclei of patients with Parkinson's disease (PD). This study examined the relationship between non-motor symptoms (NMSs) and iron accumulation in the deep nuclei of patients with PD. METHODS The QSM data were acquired from 3-Tesla magnetic resonance imaging (MRI) in 29 patients with early PD and 19 normal controls. The Korean version of the NMS scale (K-NMSS) was used for evaluation of NMSs in patients. The patients were divided into high NMS and low NMS groups. The region-of-interest analyses were performed in the following deep nuclei: red nucleus, substantia nigra pars compacta, substantia nigra pars reticulata, dentate nucleus, globus pallidus, putamen, and head of the caudate nucleus. RESULTS Thirteen patients had high NMS scores (total K-NMSS score, mean = 32.1), and 16 had low NMS scores (10.6). The QSM values in the deep were not different among the patients with high NMS scores, low NMS scores, and controls. The QSM values were not correlated linearly with K-NMSS total score after adjusting the age at acquisition of brain MRI. CONCLUSION The study demonstrated that the NMS burdens are not associated with iron accumulation in the deep nuclei of patients with PD. These results suggest that future neuroimaging studies on the pathology of NMSs in PD should use more specific and detailed clinical tools and recruit PD patients with severe NMSs.
Collapse
Affiliation(s)
- Chaewon Shin
- Department of Neurology, Kyung Hee University Medical Center, Seoul, Korea
| | - Seon Lee
- Department of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Korea
| | - Jee Young Lee
- Department of Neurology, Seoul National University-Seoul Metropolitan Government Boramae Medical Center, Seoul, Korea
| | - Jung Hyo Rhim
- Department of Radiology, Seoul National University-Seoul Metropolitan Government Boramae Medical Center, Seoul, Korea
| | - Sun Won Park
- Department of Radiology, Seoul National University-Seoul Metropolitan Government Boramae Medical Center, Seoul, Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
1089
|
Abstract
Brain iron is tightly regulated by a multitude of proteins to ensure homeostasis. Iron dyshomeostasis has become a molecular signature associated with aging which is accompanied by progressive decline in cognitive processes. A common theme in neurodegenerative diseases where age is the major risk factor, iron dyshomeostasis coincides with neuroinflammation, abnormal protein aggregation, neurodegeneration, and neurobehavioral deficits. There is a great need to determine the mechanisms governing perturbations in iron metabolism, in particular to distinguish between physiological and pathological aging to generate fruitful therapeutic targets for neurodegenerative diseases. The aim of the present review is to focus on the age-related alterations in brain iron metabolism from a cellular and molecular biology perspective, alongside genetics, and neuroimaging aspects in man and rodent models, with respect to normal aging and neurodegeneration. In particular, the relationship between iron dyshomeostasis and neuroinflammation will be evaluated, as well as the effects of systemic iron overload on the brain. Based on the evidence discussed here, we suggest a synergistic use of iron-chelators and anti-inflammatories as putative anti-brain aging therapies to counteract pathological aging in neurodegenerative diseases.
Collapse
Affiliation(s)
- Azhaar Ashraf
- Institute of Psychiatry, Psychology and Neuroscience, Department of Neuroimaging, King's College London, London, United Kingdom
| | - Maryam Clark
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Po-Wah So
- Institute of Psychiatry, Psychology and Neuroscience, Department of Neuroimaging, King's College London, London, United Kingdom
| |
Collapse
|
1090
|
Sadhukhan P, Saha S, Dutta S, Mahalanobish S, Sil PC. Nutraceuticals: An emerging therapeutic approach against the pathogenesis of Alzheimer's disease. Pharmacol Res 2018; 129:100-114. [PMID: 29183770 DOI: 10.1016/j.phrs.2017.11.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 11/22/2017] [Accepted: 11/22/2017] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease (AD) is regarded as a progressive and devastating neurodegenerative disorder. In aged individuals, it is the most prevalent cause of dementia and is characterized by gradual loss of cognitive functions. In the last decade, numerous research works were undertaken to investigate the pathogenesis of AD. Although the etiology of AD is still not clear, several histopathological studies confirm prominent changes in the AD affected brains. The major changes include the formation of senile plaques and neurofibrillary tangles primarily owing to the deposition of amyloid β plaques (Aβ) and hyper-phosphorylation of tau protein. Disruption of the redox homeostasis in the brain is a major triggering factor for the development of such pathophysiological conditions. Chemical formulations usually act by inhibiting activities of the enzymes responsible for the development of AD. But with time, these pharmacotherapies develop many side effects including toxicity in different organs. Recent researches are henceforth focused on the identification of novel therapeutic molecules from the nature's basket. This review aims to emphasize the therapeutic effects and regulation of molecular targets of different natural products such as curcumin, resveratrol, genistein and others. These prophylactic multipotent natural compounds have the potency to interfere with the formation as well as deposition of the Aβ peptides. These natural compounds have also been found in modulating different intracellular signalling molecules and enzymes including β-secretase and γ-secretase. This review article is expected to be helpful in understanding the recent progress in natural product research as a therapeutic approach in amelioration and/or delaying the detrimental effects of AD.
Collapse
Affiliation(s)
- Pritam Sadhukhan
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII-M, Kolkata, 700054, India
| | - Sukanya Saha
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII-M, Kolkata, 700054, India
| | - Sayanta Dutta
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII-M, Kolkata, 700054, India
| | - Sushweta Mahalanobish
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII-M, Kolkata, 700054, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII-M, Kolkata, 700054, India.
| |
Collapse
|
1091
|
Birkl C, Carassiti D, Hussain F, Langkammer C, Enzinger C, Fazekas F, Schmierer K, Ropele S. Assessment of ferritin content in multiple sclerosis brains using temperature-induced R* 2 changes. Magn Reson Med 2018; 79:1609-1615. [PMID: 28618066 DOI: 10.1002/mrm.26780] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/13/2017] [Accepted: 05/13/2017] [Indexed: 12/23/2022]
Abstract
PURPOSE Current MRI techniques cannot reliably assess iron content in white matter due to the confounding diamagnetic effect of myelin. The purpose of this study was to validate with histology a novel iron mapping technique that uses the temperature dependency of the paramagnetic susceptibility in multiple sclerosis (MS) brains, where white matter has been reported to show significant variations in iron content. METHODS We investigated post mortem brain tissue from three MS patients and one control subject. Temperature-dependent R2* relaxometry was performed between 4°C and 37°C. The resulting temperature coefficient ( TcR2*) maps were compared with immunohistochemical stains for ferritin light chain. RESULTS Good agreement between TcR2* maps and ferritin staining was found by way of visual comparison and quantitative analysis. The highest iron concentrations were detected at the edge of MS lesions and in the basal ganglia. For all regions, except the subcortical U-fibers, there was a significant negative correlation between the TcR2* values and the ferritin count. CONCLUSION This study provides further evidence that TcR2* may be a reliable measure of white matter iron content due to the elimination of myelin-induced susceptibility changes and is well suited for further research into neurological diseases with distortions of the iron homeostasis. Magn Reson Med 79:1609-1615, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Christoph Birkl
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Daniele Carassiti
- Blizard Institute (Neuroscience), Queen Mary, University of London, London, United Kingdom
| | - Fariha Hussain
- Blizard Institute (Neuroscience), Queen Mary, University of London, London, United Kingdom
| | | | | | - Franz Fazekas
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Klaus Schmierer
- Blizard Institute (Neuroscience), Queen Mary, University of London, London, United Kingdom.,Barts Health NHS Trust, Emergency Care and Acute Medicine Neuroscience Clinical Academic Group, London, United Kingdom
| | - Stefan Ropele
- Department of Neurology, Medical University of Graz, Graz, Austria
| |
Collapse
|
1092
|
Duyn JH. Studying brain microstructure with magnetic susceptibility contrast at high-field. Neuroimage 2018; 168:152-161. [PMID: 28242317 PMCID: PMC5569005 DOI: 10.1016/j.neuroimage.2017.02.046] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 02/03/2017] [Accepted: 02/16/2017] [Indexed: 12/14/2022] Open
Abstract
A rapidly developing application of high field MRI is the study of brain anatomy and function with contrast based on the magnetic susceptibility of tissues. To study the subtle variations in susceptibility contrast between tissues and with changes in brain activity, dedicated scan techniques such as susceptibility-weighted MRI and blood-oxygen level dependent functional MRI have been developed. Particularly strong susceptibility contrast has been observed with systems that operate at 7T and above, and their recent widespread use has led to an improved understanding of contributing sources and mechanisms. To interpret magnetic susceptibility contrast, analysis approaches have been developed with the goal of extracting measures that report on local tissue magnetic susceptibility, a physical quantity that, under certain conditions, allows estimation of blood oxygenation, local tissue iron content, and quantification of its changes with disease. Interestingly, high field studies have also brought to light that not only the makeup of tissues affects MRI susceptibility contrast, but that also a tissue's sub-voxel structure at scales all the way down to the molecular level plays an important role as well. In this review, various ways will be discussed by which sub-voxel structure can affect the MRI signal in general, and magnetic susceptibility in particular, sometimes in a complex fashion. In the light of this complexity, it appears likely that accurate, brain-wide quantification of iron will require the combination of multiple contrasts that may include diffusion and magnetization transfer information with susceptibility-weighted contrast. On the other hand, this complexity also offers opportunities to use magnetic susceptibility contrast to inform about specific microstructural aspects of brain tissue. Details and several examples will be presented in this review.
Collapse
Affiliation(s)
- Jeff H Duyn
- Advanced MRI Section, Laboratory of Functional and Molecular Imaging, National Institutes of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
1093
|
Madan CR, Kensinger EA. Predicting age from cortical structure across the lifespan. Eur J Neurosci 2018; 47:399-416. [PMID: 29359873 PMCID: PMC5835209 DOI: 10.1111/ejn.13835] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 01/12/2018] [Accepted: 01/15/2018] [Indexed: 01/22/2023]
Abstract
Despite interindividual differences in cortical structure, cross-sectional and longitudinal studies have demonstrated a large degree of population-level consistency in age-related differences in brain morphology. This study assessed how accurately an individual's age could be predicted by estimates of cortical morphology, comparing a variety of structural measures, including thickness, gyrification and fractal dimensionality. Structural measures were calculated across up to seven different parcellation approaches, ranging from one region to 1000 regions. The age prediction framework was trained using morphological measures obtained from T1-weighted MRI volumes collected from multiple sites, yielding a training dataset of 1056 healthy adults, aged 18-97. Age predictions were calculated using a machine-learning approach that incorporated nonlinear differences over the lifespan. In two independent, held-out test samples, age predictions had a median error of 6-7 years. Age predictions were best when using a combination of cortical metrics, both thickness and fractal dimensionality. Overall, the results reveal that age-related differences in brain structure are systematic enough to enable reliable age prediction based on metrics of cortical morphology.
Collapse
Affiliation(s)
- Christopher R. Madan
- School of Psychology, University of Nottingham, Nottingham, UK
- Department of Psychology, Boston College, Chestnut Hill, MA, USA
| | | |
Collapse
|
1094
|
How to choose the right MR sequence for your research question at 7 T and above? Neuroimage 2018; 168:119-140. [DOI: 10.1016/j.neuroimage.2017.04.044] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 12/29/2022] Open
|
1095
|
Beesley S, Olcese J, Saunders C, Bienkiewicz EA. Combinatorial Treatment Effects in a Cell Culture Model of Alzheimer's Disease. J Alzheimers Dis 2018; 55:1155-1166. [PMID: 27814295 DOI: 10.3233/jad-160459] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the leading cause of dementia, and as its prevalence increases, so does its detrimental impact on society. The currently available therapies have limited efficacy, leaving AD patients on an irrevocably fatal path of this disease. OBJECTIVE The purpose of this study was to test efficacy of a novel combinatorial treatment approach to alleviate AD-like pathology. METHODS We selected four naturally occurring compounds and used them in different combinations to test their effect on AD-like pathology. Employing a well-established cell culture AD model system, we evaluated levels of several diverse biomarkers associated with a number of cellular pathways associated with AD. The readouts included: amyloid-β peptides, anti-inflammatory and anti-apoptotic proteins, oxidative enzymes, and reactive oxygen species. RESULTS Using this approach, we demonstrated that the compounds delivered in combination had higher efficacy than individual treatments. Specifically, we observed significant reduction in levels of the amyloid-β peptides, as well as pro-inflammatory proteins and reactive oxygen species. Similarly, delivery of compounds in combination resulted in an increased expression of anti-apoptotic proteins and anti-oxidative enzymes. Collectively, these modifications in AD pathology biomarkers reflect a promising therapeutic and preventive strategy to combat this disease. CONCLUSION The above findings support a novel therapeutic approach to address a currently unmet medical need, which would benefit not only AD patients and their caregivers, but also society as a whole.
Collapse
Affiliation(s)
- Stephen Beesley
- Department of Biomedical Sciences, College of Medicine, Florida State University, FL, USA
| | - James Olcese
- Department of Biomedical Sciences, College of Medicine, Florida State University, FL, USA.,Center for Brain Repair, College of Medicine, Florida State University, FL, USA
| | - Charles Saunders
- Department of Behavioral Sciences and Social Medicine, College of Medicine, Florida State University, FL, USA
| | - Ewa A Bienkiewicz
- Department of Biomedical Sciences, College of Medicine, Florida State University, FL, USA.,Center for Brain Repair, College of Medicine, Florida State University, FL, USA
| |
Collapse
|
1096
|
Choi JG, Kim SY, Jeong M, Oh MS. Pharmacotherapeutic potential of ginger and its compounds in age-related neurological disorders. Pharmacol Ther 2018; 182:56-69. [PMID: 28842272 DOI: 10.1016/j.pharmthera.2017.08.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Age-related neurological disorders (ANDs), including neurodegenerative diseases, are multifactorial disorders with a risk that increases with aging. ANDs are generally characterized by common neuropathological conditions of the central nervous system, such as oxidative stress, neuroinflammation, and protein misfolding. Recently, efforts have been made to overcome ANDs because of the increase in age-dependent prevalence. Ginger, the rhizome of Zingiber officinale Roscoe, is a popular food spice and has a long history of use in traditional medicine for treating various disease symptoms. The structure-activity relationships of ginger phytochemicals show that ginger can be used to treat ANDs by targeting different ligand sites. This review shows that ginger and its constituents, such as 6-gingerol, 6-shogaol, 6-paradol, zingerone, and dehydrozingerone, are effective for ameliorating the neurological symptoms and pathological conditions of ANDs through by modulating cell death or cell survival signaling molecules. From this review, we conclude that the active ingredients in ginger have therapeutic potential in ANDs.
Collapse
Affiliation(s)
- Jin Gyu Choi
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sun Yeou Kim
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, Republic of Korea
| | - Minsun Jeong
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, Republic of Korea
| | - Myung Sook Oh
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; College of Pharmacy and Kyung Hee East-West Pharmaceutical Research Institute, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
1097
|
Vázquez‐Costa JF, Mazón M, Carreres‐Polo J, Hervás D, Pérez‐Tur J, Martí‐Bonmatí L, Sevilla T. Brain signal intensity changes as biomarkers in amyotrophic lateral sclerosis. Acta Neurol Scand 2018; 137:262-271. [PMID: 29082510 DOI: 10.1111/ane.12863] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2017] [Indexed: 01/31/2023]
Abstract
OBJECTIVES To evaluate the contribution of the demographical, clinical, analytical and genetic factors to brain signal intensity changes in T2-weighted MR images in amyotrophic lateral sclerosis (ALS) patients and controls. METHODS Susceptibility-weighted and FLAIR sequences were obtained in a 3T MR scanner. Iron-related hypointensities in the motor cortex (IRhMC) and hyperintensities of the corticospinal tract (HCT) were qualitatively scored. Age, gender, family history and clinical variables were recorded. Baseline levels of ferritin were measured. C9orf72 was tested in all patients and SOD1 only in familial ALS patients not carrying a C9orf72 expansion. Patients who carried a mutation were categorized as genetic. Associations of these variables with visual scores were assessed with multivariable analysis. RESULTS A total of 102 ALS patients (92 non-genetic and 10 genetic) and 48 controls (28 ALS mimics and 20 healthy controls) were recruited. In controls, IRhMC associated with age, but HCT did not. In ALS patients, both HTC and IRhMC strongly associated with clinical UMN impairment and bulbar onset. The intensity/extent of IRhMC in the different motor homunculus regions (lower limbs, upper limbs and bulbar) were linked to the symptoms onset site. Between genetic and sporadic patients, no difference in IRhMC and HCT was found. CONCLUSIONS IRhMC and HCT are reliable markers of UMN degeneration in ALS patients and are more frequent in bulbar onset patients, independently of the mutation status. Age should be considered when evaluating IRhMC. The regional measurement of IRhMC following the motor homunculus could be used as a measure of disease progression.
Collapse
Affiliation(s)
- Juan F. Vázquez‐Costa
- Neuromuscular Research Unit Instituto de Investigación Sanitaria la Fe Valencia Spain
- ALS Unit Department of Neurology Hospital Universitario y Politécnico La Fe Valencia Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Valencia Spain
| | - Miguel Mazón
- Department of Radiology and Biomedical Imaging Research Group GIBI2 Hospital Universitario y Politécnico La Fe and Instituto de Investigación Sanitaria la Fe Valencia Spain
| | - Joan Carreres‐Polo
- Department of Radiology and Biomedical Imaging Research Group GIBI2 Hospital Universitario y Politécnico La Fe and Instituto de Investigación Sanitaria la Fe Valencia Spain
| | - David Hervás
- Biostatistics Unit Instituto de Investigación Sanitaria la Fe Valencia Spain
| | - Jordi Pérez‐Tur
- Laboratory of Molecular Genetics Institut de Biomedicina de València‐CSIC Valencia Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) Valencia Spain
- Unidad mixta de Neurología y Genética Instituto de Investigación Sanitaria la Fe (IIS La Fe) Valencia Spain
| | - Luis Martí‐Bonmatí
- Department of Radiology and Biomedical Imaging Research Group GIBI2 Hospital Universitario y Politécnico La Fe and Instituto de Investigación Sanitaria la Fe Valencia Spain
| | - Teresa Sevilla
- Neuromuscular Research Unit Instituto de Investigación Sanitaria la Fe Valencia Spain
- ALS Unit Department of Neurology Hospital Universitario y Politécnico La Fe Valencia Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Valencia Spain
- Department of Medicine University of Valencia Valencia Spain
| |
Collapse
|
1098
|
Iron dysregulates APP processing accompanying with sAPPα cellular retention and β-secretase inhibition in rat cortical neurons. Acta Pharmacol Sin 2018; 39:177-183. [PMID: 28836584 DOI: 10.1038/aps.2017.113] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 04/19/2017] [Indexed: 12/14/2022]
Abstract
Amyloid precursor protein (APP) and iron both play pivotal roles in the central nervous system, but whether and how iron influences the processing of endogenous APP in neurons remain unclear. Here, we investigated the regulatory effects and underlying mechanisms of iron on non-amyloidogenic and amyloidogenic processing of APP in rat primary cortical neurons. Treatment of the neurons with ferric ammonium citrate (FAC, 100 μmol/L) markedly facilitated the non-amyloidogenic processing of APP, as evidenced by a robust increase in α-secretase-derived carboxy-terminal fragment α (CTFα). Furthermore, the distribution of sAPPα was altered after iron treatment, and sAPPα remained in the cellular lysates instead of being secreted into the extracellular milieu. Moreover, the levels of APP amyloidogenic products, including sAPPβ and Aβ were both decreased. We further revealed that FAC did not alter the expression of β-secretase, but significantly suppressed its enzymatic activity in iron-treated neurons. In a cell-free β-secretase activity assay, FAC dose-dependently inhibited the activity of purified β-secretase with an IC50 value of 21.67 μmol/L. Our data provide the first evidence that iron overload alters the neuronal sAPPα distribution and directly inhibits β-secretase activity. These findings shed light on the regulatory mechanism of bio-metals on APP processing.
Collapse
|
1099
|
Unraveling the Burden of Iron in Neurodegeneration: Intersections with Amyloid Beta Peptide Pathology. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2850341. [PMID: 29581821 PMCID: PMC5831758 DOI: 10.1155/2018/2850341] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 12/17/2017] [Indexed: 12/14/2022]
Abstract
Iron overload is a hallmark of many neurodegenerative processes such as Alzheimer's, Parkinson's, and Huntington's diseases. Unbound iron accumulated as a consequence of brain aging is highly reactive with water and oxygen and produces reactive oxygen species (ROS) or free radicals. ROS are toxic compounds able to damage cell membranes, DNA, and mitochondria. Which are the mechanisms involved in neuronal iron homeostasis and in neuronal response to iron-induced oxidative stress constitutes a cutting-edge topic in metalloneurobiology. Increasing our knowledge about the underlying mechanisms that operate in iron accumulation and their consequences would shed light on the comprehension of the molecular events that participate in the pathophysiology of the abovementioned neurodegenerative diseases. In this review, current evidences about iron accumulation in the brain, the signaling mechanisms triggered by metal overload, as well as the interaction between amyloid β (Aβ) and iron, will be summarized.
Collapse
|
1100
|
Moreau C, Duce JA, Rascol O, Devedjian JC, Berg D, Dexter D, Cabantchik ZI, Bush AI, Devos D. Iron as a therapeutic target for Parkinson's disease. Mov Disord 2018; 33:568-574. [PMID: 29380903 DOI: 10.1002/mds.27275] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/24/2017] [Accepted: 11/27/2017] [Indexed: 12/12/2022] Open
Affiliation(s)
- Caroline Moreau
- Université de Lille, CHU de Lille, INSERM UMRS_1171, Service de Neurologie NS-Park/FCRIN Network LICEND COEN Center, Lille, France
| | - James A Duce
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, UK, and Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Olivier Rascol
- Université de Toulouse, UPS, CHU de Toulouse, INSERM; Centre d'Investigation Clinique CIC1436, Services de Neurologie et de Pharmacologie Clinique, UMR TONIC, NS-Park/FCRIN Network, NeuroToul COEN Center, Toulouse, France
| | - Jean-Christophe Devedjian
- University de Lille, CHU de Lille, INSERM UMRS_1171, NS-Park/FCRIN Network LICEND COEN Center, Lille, France
| | - Daniela Berg
- Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany and Hertie-Institute of Clinical Brain Research, Department of Neurodegeneration, Tübingen, Germany
| | | | - Z Ioav Cabantchik
- Della Pergola Chair, Alexander Silberman Institute of Life Sciences, Hebrew University, Jerusalem, Israel
| | - Ashley I Bush
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - David Devos
- Université de Lille, CHU de Lille, INSERM UMRS_1171, Service de Pharmacologie Clinique et Service de Neurologie NS-Park/FCRIN Network LICEND COEN Center, Lille, France
| | | |
Collapse
|