1
|
Gromadzka G, Wilkaniec A, Tarnacka B, Hadrian K, Bendykowska M, Przybyłkowski A, Litwin T. The Role of Glia in Wilson's Disease: Clinical, Neuroimaging, Neuropathological and Molecular Perspectives. Int J Mol Sci 2024; 25:7545. [PMID: 39062788 PMCID: PMC11276698 DOI: 10.3390/ijms25147545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Wilson's disease (WD) is inherited in an autosomal recessive manner and is caused by pathogenic variants of the ATP7B gene, which are responsible for impaired copper transport in the cell, inhibition of copper binding to apoceruloplasmin, and biliary excretion. This leads to the accumulation of copper in the tissues. Copper accumulation in the CNS leads to the neurological and psychiatric symptoms of WD. Abnormalities of copper metabolism in WD are associated with impaired iron metabolism. Both of these elements are redox active and may contribute to neuropathology. It has long been assumed that among parenchymal cells, astrocytes have the greatest impact on copper and iron homeostasis in the brain. Capillary endothelial cells are separated from the neuropil by astrocyte terminal legs, putting astrocytes in an ideal position to regulate the transport of iron and copper to other brain cells and protect them if metals breach the blood-brain barrier. Astrocytes are responsible for, among other things, maintaining extracellular ion homeostasis, modulating synaptic transmission and plasticity, obtaining metabolites, and protecting the brain against oxidative stress and toxins. However, excess copper and/or iron causes an increase in the number of astrocytes and their morphological changes observed in neuropathological studies, as well as a loss of the copper/iron storage function leading to macromolecule peroxidation and neuronal loss through apoptosis, autophagy, or cuproptosis/ferroptosis. The molecular mechanisms explaining the possible role of glia in copper- and iron-induced neurodegeneration in WD are largely understood from studies of neuropathology in Parkinson's disease and Alzheimer's disease. Understanding the mechanisms of glial involvement in neuroprotection/neurotoxicity is important for explaining the pathomechanisms of neuronal death in WD and, in the future, perhaps for developing more effective diagnostic/treatment methods.
Collapse
Affiliation(s)
- Grażyna Gromadzka
- Department of Biomedical Sciences, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University, Wóycickiego 1/3, 01-938 Warsaw, Poland
| | - Anna Wilkaniec
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland
| | - Beata Tarnacka
- Department of Rehabilitation, Medical University of Warsaw, Spartańska 1, 02-637 Warsaw, Poland
| | - Krzysztof Hadrian
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland (A.P.)
| | - Maria Bendykowska
- Students Scientific Association “Immunis”, Cardinal Stefan Wyszynski University, Dewajtis 5, 01-815 Warsaw, Poland
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland (A.P.)
| | - Tomasz Litwin
- Second Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957 Warsaw, Poland
| |
Collapse
|
2
|
Bjørklund G, Oliinyk P, Khavrona O, Lozynska I, Lysiuk R, Darmohray R, Antonyak H, Dub N, Zayachuk V, Antoniv O, Rybak O, Peana M. The Effects of Fisetin and Curcumin on Oxidative Damage Caused by Transition Metals in Neurodegenerative Diseases. Mol Neurobiol 2024:10.1007/s12035-024-04321-2. [PMID: 38970766 DOI: 10.1007/s12035-024-04321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/19/2024] [Indexed: 07/08/2024]
Abstract
Neurodegenerative diseases pose a significant health challenge for the elderly. The escalating presence of toxic metals and chemicals in the environment is a potential contributor to central nervous system dysfunction and the onset of neurodegenerative conditions. Transition metals play a crucial role in various pathophysiological mechanisms associated with prevalent neurodegenerative diseases such as Alzheimer's and Parkinson's. Given the ubiquitous exposure to metals from diverse sources in everyday life, the workplace, and the environment, most of the population faces regular contact with different forms of these metals. Disturbances in the levels and homeostasis of certain transition metals are closely linked to the manifestation of neurodegenerative disorders. Oxidative damage further exacerbates the progression of neurological consequences. Presently, there exists no curative therapy for individuals afflicted by neurodegenerative diseases, with treatment approaches primarily focusing on alleviating pathological symptoms. Within the realm of biologically active compounds derived from plants, flavonoids and curcuminoids stand out for their extensively documented antioxidant, antiplatelet, and neuroprotective properties. The utilization of these compounds holds the potential to formulate highly effective therapeutic strategies for managing neurodegenerative diseases. This review provides a comprehensive overview of the impact of abnormal metal levels, particularly copper, iron, and zinc, on the initiation and progression of neurodegenerative diseases. Additionally, it aims to elucidate the potential of fisetin and curcumin to inhibit or decelerate the neurodegenerative process.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Toften 24, 8610, Mo I Rana, Norway.
| | - Petro Oliinyk
- Department of Disaster Medicine and Military Medicine, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Oksana Khavrona
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
- Department of Biological Chemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Iryna Lozynska
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
- Department of Biological Chemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Roman Lysiuk
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
- Department of Pharmacognosy and Botany, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Roman Darmohray
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
- Department of Pharmacognosy and Botany, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Halyna Antonyak
- Department of Ecology, Ivan Franko National University of Lviv, Lviv, 79005, Ukraine
| | - Natalia Dub
- Andrei Krupynskyi Lviv Medical Academy, Lviv, 79000, Ukraine
| | - Vasyl Zayachuk
- Department of Botany, Ukrainian National Forestry University, Wood Science and Non-Wood Forest Products, Lviv, 79057, Ukraine
| | - Olha Antoniv
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
- Department of Pharmacology, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Oksana Rybak
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
- Department of Pharmacognosy and Botany, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, 07100, Sassari, Italy.
| |
Collapse
|
3
|
Ghaderi S, Mohammadi S, Nezhad NJ, Karami S, Sayehmiri F. Iron quantification in basal ganglia: quantitative susceptibility mapping as a potential biomarker for Alzheimer's disease - a systematic review and meta-analysis. Front Neurosci 2024; 18:1338891. [PMID: 38469572 PMCID: PMC10925682 DOI: 10.3389/fnins.2024.1338891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/13/2024] [Indexed: 03/13/2024] Open
Abstract
Introduction Alzheimer's disease (AD), characterized by distinctive pathologies such as amyloid-β plaques and tau tangles, also involves deregulation of iron homeostasis, which may accelerate neurodegeneration. This meta-analysis evaluated the use of quantitative susceptibility mapping (QSM) to detect iron accumulation in the deep gray matter (DGM) of the basal ganglia in AD, contributing to a better understanding of AD progression, and potentially leading to new diagnostic and therapeutic approaches. Methods Using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we systematically searched the PubMed, Scopus, Web of Sciences, and Google Scholar databases up to October 2023 for studies employing QSM in AD research. Eligibility criteria were based on the PECO framework, and we included studies assessing alterations in magnetic susceptibility indicative of iron accumulation in the DGM of patients with AD. After initial screening and quality assessment using the Newcastle-Ottawa Scale, a meta-analysis was conducted to compare iron levels between patients with AD and healthy controls (HCs) using a random-effects model. Results The meta-analysis included nine studies comprising 267 patients with AD and 272 HCs. There were significantly higher QSM values, indicating greater iron deposition, in the putamen (standardized mean difference (SMD) = 1.23; 95% CI: 0.62 to 1.84; p = 0.00), globus pallidus (SMD = 0.79; 95% CI: 0.07 to 1.52; p = 0.03), and caudate nucleus (SMD = 0.72; 95% CI: 0.39 to 1.06; p = 0.00) of AD patients compared to HCs. However, no significant differences were found in the thalamus (SMD = 1.00; 95% CI: -0.42 to 2.43; p = 0.17). The sensitivity analysis indicated that no single study impacted the overall results. Age was identified as a major contributor to heterogeneity across all basal ganglia nuclei in subgroup analysis. Older age (>69 years) and lower male percentage (≤30%) were associated with greater putamen iron increase in patients with AD. Conclusion The study suggests that excessive iron deposition is linked to the basal ganglia in AD, especially the putamen. The study underscores the complex nature of AD pathology and the accumulation of iron, influenced by age, sex, and regional differences, necessitating further research for a comprehensive understanding.
Collapse
Affiliation(s)
- Sadegh Ghaderi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sana Mohammadi
- Department of Medical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Jashire Nezhad
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Shaghayegh Karami
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sayehmiri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Science, Tehran, Iran
| |
Collapse
|
4
|
Schildroth S, Kordas K, White RF, Friedman A, Placidi D, Smith D, Lucchini RG, Wright RO, Horton M, Claus Henn B. An Industry-Relevant Metal Mixture, Iron Status, and Reported Attention-Related Behaviors in Italian Adolescents. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:27008. [PMID: 38363634 PMCID: PMC10871126 DOI: 10.1289/ehp12988] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 12/01/2023] [Accepted: 01/17/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND Exposure to environmental metals has been consistently associated with attention and behavioral deficits in children, and these associations may be modified by coexposure to other metals or iron (Fe) status. However, few studies have investigated Fe status as a modifier of a metal mixture, particularly with respect to attention-related behaviors. METHODS We used cross-sectional data from the Public Health Impact of Metals Exposure study, which included 707 adolescents (10-14 years of age) from Brescia, Italy. Manganese, chromium, and copper were quantified in hair samples, and lead was quantified in whole blood, using inductively coupled plasma mass spectrometry. Concentrations of Fe status markers (ferritin, hemoglobin, transferrin) were measured using immunoassays or luminescence assays. Attention-related behaviors were assessed using the Conners Rating Scales Self-Report Scale-Long Form, Parent Rating Scales Revised-Short Form, and Teacher Rating Scales Revised-Short Form. We employed Bayesian kernel machine regression to examine associations of the metal mixture with these outcomes and evaluate Fe status as a modifier. RESULTS Higher concentrations of the metals and ferritin were jointly associated with worse self-reported attention-related behaviors: metals and ferritin set to their 90th percentiles were associated with 3.0% [β = 0.03 ; 95% credible interval (CrI): - 0.01 , 0.06], 4.1% (β = 0.04 ; 95% CrI: 0.00, 0.08), and 4.1% (β = 0.04 ; 95% CrI: 0.00, 0.08) higher T -scores for self-reported attention deficit/hyperactivity disorder (ADHD) index, inattention, and hyperactivity, respectively, compared with when metals and ferritin were set to their 50th percentiles. These associations were driven by hair manganese, which exhibited nonlinear associations with all self-reported scales. There was no evidence that Fe status modified the neurotoxicity of the metal mixture. The metal mixture was not materially associated with any parent-reported or teacher-reported scale. CONCLUSIONS The overall metal mixture, driven by manganese, was adversely associated with self-reported attention-related behavior. These findings suggest that exposure to multiple environmental metals impacts adolescent neurodevelopment, which has significant public health implications. https://doi.org/10.1289/EHP12988.
Collapse
Affiliation(s)
- Samantha Schildroth
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Katarzyna Kordas
- Department of Epidemiology and Environmental Health, University at Buffalo, Buffalo, New York, USA
| | - Roberta F. White
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts, USA
- Department of Neurology, Boston University, Boston, Massachusetts, USA
| | - Alexa Friedman
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Donatella Placidi
- Department of Occupational Health, University of Brescia, Brescia, Italy
| | - Donald Smith
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, Santa Cruz, California, USA
| | - Roberto G. Lucchini
- Department of Occupational Health, University of Brescia, Brescia, Italy
- Department of Environmental Health Sciences, Florida International University, Miami, Florida, USA
| | - Robert O. Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Megan Horton
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Birgit Claus Henn
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Lee EY, Kim J, Prado-Rico JM, Du G, Lewis MM, Kong L, Yanosky JD, Eslinger P, Kim BG, Hong YS, Mailman RB, Huang X. Effects of mixed metal exposures on MRI diffusion features in the medial temporal lobe. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.07.18.23292828. [PMID: 37503124 PMCID: PMC10371112 DOI: 10.1101/2023.07.18.23292828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Background Environmental exposure to metal mixtures is common and may be associated with increased risk for neurodegenerative disorders including Alzheimer's disease. Objective This study examined associations of mixed metal exposures with medial temporal lobe (MTL) MRI structural metrics and neuropsychological performance. Methods Metal exposure history, whole blood metal, and neuropsychological tests were obtained from subjects with/without a history of mixed metal exposure from welding fumes (42 exposed subjects; 31 controls). MTL structures (hippocampus, entorhinal and parahippocampal cortices) were assessed by morphologic (volume, cortical thickness) and diffusion tensor imaging [mean (MD), axial (AD), radial diffusivity (RD), and fractional anisotropy (FA)] metrics. In exposed subjects, correlation, multiple linear, Bayesian kernel machine regression, and mediation analyses were employed to examine effects of single- or mixed-metal predictor(s) and their interactions on MTL structural and neuropsychological metrics; and on the path from metal exposure to neuropsychological consequences. Results Compared to controls, exposed subjects had higher blood Cu, Fe, K, Mn, Pb, Se, and Zn levels (p's<0.026) and poorer performance in processing/psychomotor speed, executive, and visuospatial domains (p's<0.046). Exposed subjects displayed higher MD, AD, and RD in all MTL ROIs (p's<0.040) and lower FA in entorhinal and parahippocampal cortices (p's<0.033), but not morphological differences. Long-term mixed-metal exposure history indirectly predicted lower processing speed performance via lower parahippocampal FA (p=0.023). Higher whole blood Mn and Cu predicted higher entorhinal diffusivity (p's<0.043) and lower Delayed Story Recall performance (p=0.007) without overall metal mixture or interaction effects. Discussion Mixed metal exposure predicted MTL structural and neuropsychological features that are similar to Alzheimer's disease at-risk populations. These data warrant follow-up as they may illuminate the path for environmental exposure to Alzheimer's disease-related health outcomes.
Collapse
Affiliation(s)
- Eun-Young Lee
- Department of Health Care and Science, Dong-A University, Busan, South-Korea
| | - Juhee Kim
- Department of Health Care and Science, Dong-A University, Busan, South-Korea
| | - Janina Manzieri Prado-Rico
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Guangwei Du
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Mechelle M. Lewis
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Lan Kong
- Department of Public Health Sciences, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Jeff D. Yanosky
- Department of Public Health Sciences, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Paul Eslinger
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Byoung-Gwon Kim
- Department of Preventive Medicine, College of Medicine, Dong-A University, Busan, South Korea
| | - Young-Seoub Hong
- Department of Preventive Medicine, College of Medicine, Dong-A University, Busan, South Korea
| | - Richard B. Mailman
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| | - Xuemei Huang
- Department of Neurology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Pharmacology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Radiology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Neurosurgery, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
- Department of Kinesiology, Pennsylvania State University-Milton S. Hershey Medical Center, Hershey PA 17033, USA
| |
Collapse
|
6
|
Schildroth S, Valeri L, Kordas K, Shi B, Friedman A, Smith D, Placidi D, Wright RO, Lucchini RG, White RF, Horton M, Claus Henn B. Assessing the mediating role of iron status on associations between an industry-relevant metal mixture and verbal learning and memory in Italian adolescents. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 906:167435. [PMID: 37774885 PMCID: PMC10918745 DOI: 10.1016/j.scitotenv.2023.167435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
BACKGROUND Metals, including lead (Pb), manganese (Mn), chromium (Cr) and copper (Cu), have been associated with neurodevelopment; iron (Fe) plays a role in the metabolism and neurotoxicity of metals, suggesting Fe may mediate metal-neurodevelopment associations. However, no study to date has examined Fe as a mediator of the association between metal mixtures and neurodevelopment. OBJECTIVE We assessed Fe status as a mediator of a mixture of Pb, Mn, Cr and Cu in relation to verbal learning and memory in a cohort of Italian adolescents. METHODS We used cross-sectional data from 383 adolescents (10-14 years) in the Public Health Impact of Metals Exposure Study. Metals were quantified in blood (Pb) or hair (Mn, Cr, Cu) using ICP-MS, and three markers of Fe status (blood hemoglobin, serum ferritin and transferrin) were quantified using luminescence assays or immunoassays. Verbal learning and memory were assessed using the California Verbal Learning Test for Children (CVLT-C). We used Bayesian Kernel Machine Regression Causal Mediation Analysis to estimate four mediation effects: the natural direct effect (NDE), natural indirect effect (NIE), controlled direct effect (CDE) and total effect (TE). Beta (β) coefficients and 95 % credible intervals (CIs) were estimated for all effects. RESULTS The metal mixture was jointly associated with a greater number of words recalled on the CVLT-C, but these associations were not mediated by Fe status. For example, when ferritin was considered as the mediator, the NIE for long delay free recall was null (β = 0.00; 95 % CI = -0.22, 0.23). Conversely, the NDE (β = 0.23; 95 % CI = 0.01, 0.44) indicated a beneficial association of the mixture with recall that operated independently of Fe status. CONCLUSION An industry-relevant metal mixture was associated with learning and memory, but there was no evidence of mediation by Fe status. Further studies in populations with Fe deficiency and greater variation in metal exposure are warranted.
Collapse
Affiliation(s)
- Samantha Schildroth
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA.
| | - Linda Valeri
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Katarzyna Kordas
- Department of Epidemiology and Environmental Health, University at Buffalo, Buffalo, NY, USA
| | - Baoyi Shi
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Alexa Friedman
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Donald Smith
- Department of Microbiology and Environmental Toxicology, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Donatella Placidi
- Department of Occupational Health, University of Brescia, Brescia, Italy
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roberto G Lucchini
- Department of Occupational Health, University of Brescia, Brescia, Italy; Department of Environmental Health Sciences, Florida International University, Miami, FL, USA
| | - Roberta F White
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA; Department of Neurology, Boston University, Boston, MA, USA
| | - Megan Horton
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Birgit Claus Henn
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| |
Collapse
|
7
|
Rezqaoui A, Ibouzine-Dine L, Elhamzaoui A, Brouzi MYE, Dimaoui A, Hessni AE, Mesfioui A. Potential Role of Oxidative Stress in the Effects of Chronic Administration of Iron on Affective and Cognitive Behavior on Male Wistar Rat. Biol Trace Elem Res 2023; 201:4812-4826. [PMID: 36683122 DOI: 10.1007/s12011-023-03560-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/05/2023] [Indexed: 01/24/2023]
Abstract
In this work, we studied the impact of chronic iron exposure, in the form of iron sulfate (FeSo4), on affective and cognitive disorders and oxidative stress in the male Wistar rat. The treatment was carried out for 8 weeks, the rats received an intraperitoneal injection of iron at different doses: 0.25, 0.5, and 1 mg/kg. Affective and cognitive disorders are assessed in open field test (OFT), elevated plus maze (EPM), forced swimming test (FST), Morris water maze (MWM), and Y-maze. The hippocampus and prefrontal cortex of each animal were taken for biochemical examination. Our results show that iron exerts anxiogenic and depressogenic effects, which were observed first at the dose of 0.5 mg/kg and continued in a dose-dependent manner up to the maximum tested dose of 1 mg/kg. According to results from the MWM and Y-maze tests, continuous exposure to iron induces cognitive disorders that are defined by the disturbance of working memory and influences spatial learning performance causing a deficit of spatial memory retention. We noted that chronic exposure to iron can be associated with the appearance of a state of oxidative stress in the hippocampus and the prefrontal cortex demonstrated by an increase in lipid peroxidation, an increase in nitric oxide, and also by disturbances in the antioxidant defense systems following a determination of the concentrations of catalase. In conclusion, we can deduce from this work that chronic iron exposure can be related to the induction of cognitive and affective disorders and oxidative stress.
Collapse
Affiliation(s)
- Ayoub Rezqaoui
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco.
| | - Laila Ibouzine-Dine
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Abdelghafour Elhamzaoui
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Mohamed Yassine El Brouzi
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Amal Dimaoui
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Aboubaker El Hessni
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Abdelhalem Mesfioui
- Laboratory of Biology and Health, Department of Biology, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| |
Collapse
|
8
|
Microglial Activation in Metal Neurotoxicity: Impact in Neurodegenerative Diseases. BIOMED RESEARCH INTERNATIONAL 2023; 2023:7389508. [PMID: 36760476 PMCID: PMC9904912 DOI: 10.1155/2023/7389508] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 02/04/2023]
Abstract
Neurodegenerative processes encompass a large variety of diseases with different pathological patterns and clinical features, such as Alzheimer's and Parkinson's diseases. Exposure to metals has been hypothesized to increase oxidative stress in brain cells leading to cell death and neurodegeneration. Neurotoxicity of metals has been demonstrated by several in vitro and in vivo experimental studies, and most probably, each metal has its specific pathway to trigger cell death. As a result, exposure to essential metals, such as manganese, iron, copper, zinc, and cobalt, and nonessential metals, including lead, aluminum, and cadmium, perturbs metal homeostasis at the cellular and organism levels leading to neurodegeneration. In this contribution, a comprehensive review of the molecular mechanisms by which metals affect microglia physiology and signaling properties is presented. Furthermore, studies that validate the disruption of microglia activation pathways as an essential mechanism of metal toxicity that can contribute to neurodegenerative disease are also presented and discussed.
Collapse
|
9
|
Schildroth S, Kordas K, Bauer JA, Wright RO, Claus Henn B. Environmental Metal Exposure, Neurodevelopment, and the Role of Iron Status: a Review. Curr Environ Health Rep 2022; 9:758-787. [PMID: 35997893 DOI: 10.1007/s40572-022-00378-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW Exposure to environmental metals, like lead (Pb), manganese (Mn), and methylmercury (Me-Hg), has consistently been implicated in neurodevelopmental dysfunction. Recent research has focused on identifying modifying factors of metal neurotoxicity in childhood, such as age, sex, and co-exposures. Iron (Fe) status is critical for normal cognitive development during childhood, and current mechanistic, animal, and human evidence suggests that Fe status may be a modifier or mediator of associations between environmental metals and neurodevelopment. The goals of this review are to describe the current state of the epidemiologic literature on the role of Fe status (i.e., hemoglobin, ferritin, blood Fe concentrations) and Fe supplementation in the relationship between metals and children's neurodevelopment, and to identify research gaps. RECENT FINDINGS We identified 30 studies in PubMed and EMBASE that assessed Fe status as a modifier, mediator, or co-exposure of associations of Pb, Me-Hg, Mn, copper (Cu), zinc (Zn), arsenic (As), or metal mixtures measured in early life (prenatal period through 8 years of age) with cognition in children. In experimental studies, co-supplementation of Fe and Zn was associated with better memory and cognition than supplementation with either metal alone. Several observational studies reported interactions between Fe status and Pb, Mn, Zn, or As in relation to developmental indices, memory, attention, and behavior, whereby adverse associations of metals with cognition were worse among Fe-deficient children compared to Fe-sufficient children. Only two studies quantified joint associations of complex metal mixtures that included Fe with neurodevelopment, though findings from these studies were not consistent. Findings support memory and attention as two possible cognitive domains that may be both vulnerable to Fe deficiency and a target of metals toxicity. Major gaps in the literature remain, including evaluating Fe status as a modifier or mediator of metal mixtures and cognition. Given that Fe deficiency is the most common nutritional deficiency worldwide, characterizing Fe status in studies of metals toxicity is important for informing public health interventions.
Collapse
Affiliation(s)
- Samantha Schildroth
- Department of Environmental Health, School of Public Health, Boston University, 715 Albany St., Boston, MA, 02118, USA.
| | - Katarzyna Kordas
- Department of Epidemiology and Environmental Health, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Julia Anglen Bauer
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Birgit Claus Henn
- Department of Environmental Health, School of Public Health, Boston University, 715 Albany St., Boston, MA, 02118, USA
| |
Collapse
|
10
|
Fernández-Albarral JA, Salobrar-García E, Matamoros JA, Fernández-Mendívil C, del Sastre E, Chen L, de Hoz R, López-Cuenca I, Sánchez-Puebla L, Ramírez JM, Salazar JJ, Lopez MG, Ramírez AI. Microglial Hemoxygenase-1 Deletion Reduces Inflammation in the Retina of Old Mice with Tauopathy. Antioxidants (Basel) 2022; 11:2151. [PMID: 36358522 PMCID: PMC9686584 DOI: 10.3390/antiox11112151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 09/26/2023] Open
Abstract
Tauopathies such as Alzheimer's disease are characterized by the accumulation of neurotoxic aggregates of tau protein. With aging and, especially, in Alzheimer's patients, the inducible enzyme heme oxygenase 1 (HO-1) progressively increases in microglia, causing iron accumulation, neuroinflammation, and neurodegeneration. The retina is an organ that can be readily accessed and can reflect changes that occur in the brain. In this context, we evaluated how the lack of microglial HO-1, using mice that do not express HO-1 in microglia (HMO-KO), impacts retinal macro and microgliosis of aged subjects (18 months old mice) subjected to tauopathy by intrahippocampal delivery of AAV-hTauP301L (TAU). Our results show that although tauopathy, measured as anti-TAUY9 and anti-AT8 positive immunostaining, was not observed in the retina of WT-TAU or HMO-KO+TAU mice, a morphometric study of retinal microglia and macroglia showed significant retinal changes in the TAU group compared to the WT group, such as: (i) increased number of activated microglia, (ii) retraction of microglial processes, (iii) increased number of CD68+ microglia, and (iv) increased retinal area occupied by GFAP (AROA) and C3 (AROC3). This retinal inflammatory profile was reduced in HMO-KO+TAU mice. Conclusion: Reduction of microglial HO-1 could be beneficial to prevent tauopathy-induced neuroinflammation.
Collapse
Affiliation(s)
- José A. Fernández-Albarral
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Elena Salobrar-García
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - José A. Matamoros
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Cristina Fernández-Mendívil
- Instituto Teófilo Hernando for Drug Discovery, Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, 28029 Madrid, Spain
- Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Eric del Sastre
- Instituto Teófilo Hernando for Drug Discovery, Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, 28029 Madrid, Spain
- Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Lejing Chen
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Rosa de Hoz
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Inés López-Cuenca
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Lidia Sánchez-Puebla
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - José M. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Medicina, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Juan J. Salazar
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Manuela G. Lopez
- Instituto Teófilo Hernando for Drug Discovery, Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, 28029 Madrid, Spain
- Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Ana I. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| |
Collapse
|
11
|
Barreiro S, Silva B, Long S, Pinto M, Remião F, Sousa E, Silva R. Fiscalin Derivatives as Potential Neuroprotective Agents. Pharmaceutics 2022; 14:pharmaceutics14071456. [PMID: 35890350 PMCID: PMC9320635 DOI: 10.3390/pharmaceutics14071456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/17/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Neurodegenerative diseases (ND) share common molecular/cellular mechanisms that contribute to their progression and pathogenesis. In this sense, we are here proposing new neuroprotection strategies by using marine-derived compounds as fiscalins. This work aims to evaluate the protective effects of fiscalin derivatives towards 1-methyl-4-phenylpyridinium (MPP+)- and iron (III)-induced cytotoxicity in differentiated SH-SY5Y cells, an in vitro disease model to study ND; and on P-glycoprotein (P-gp) transport activity, an efflux pump of drugs and neurotoxins. SH-SY5Y cells were simultaneously exposed to MPP+ or iron (III), and noncytotoxic concentrations of 18 fiscalin derivatives (0–25 μM), being the cytotoxic effect of both MPP+ and iron (III) evaluated 24 and 48 h after exposure. Fiscalins 1a and 1b showed a significant protective effect against MPP+-induced cytotoxicity and fiscalins 1b, 2b, 4 and 5 showed a protective effect against iron (III)-induced cytotoxicity. Fiscalins 4 and 5 caused a significant P-gp inhibition, while fiscalins 1c, 2a, 2b, 6 and 11 caused a modest increase in P-gp transport activity, thus suggesting a promising source of new P-gp inhibitors and activators, respectively. The obtained results highlight fiscalins with promising neuroprotective effects and with relevance for the synthesis of new derivatives for the treatment/prevention of ND.
Collapse
Affiliation(s)
- Sandra Barreiro
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (B.S.); (F.R.)
- UCIBIO—Applied Molecular Biosciences Unit, Requimte, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Correspondence: (S.B.); (R.S.)
| | - Bárbara Silva
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (B.S.); (F.R.)
- UCIBIO—Applied Molecular Biosciences Unit, Requimte, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Solida Long
- Department of Bioengineering, Royal University of Phnom Penh, Russian Confederation Blvd., Phnom Penh 12156, Cambodia;
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.P.); (E.S.)
| | - Madalena Pinto
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.P.); (E.S.)
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (B.S.); (F.R.)
- UCIBIO—Applied Molecular Biosciences Unit, Requimte, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Emília Sousa
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.P.); (E.S.)
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Renata Silva
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (B.S.); (F.R.)
- UCIBIO—Applied Molecular Biosciences Unit, Requimte, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Correspondence: (S.B.); (R.S.)
| |
Collapse
|
12
|
Wang F, Zhang M, Li Y, Li Y, Gong H, Li J, Zhang Y, Zhang C, Yan F, Sun B, He N, Wei H. Alterations in brain iron deposition with progression of late-life depression measured by magnetic resonance imaging (MRI)-based quantitative susceptibility mapping. Quant Imaging Med Surg 2022; 12:3873-3888. [PMID: 35782236 PMCID: PMC9246724 DOI: 10.21037/qims-21-1137] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/19/2022] [Indexed: 08/27/2023]
Abstract
BACKGROUND Previous studies have revealed abnormality of iron deposition in the brain of patients with depression. The progression of iron deposition associated with depression remains to be elucidated. METHODS This is a longitudinal study. We explored brain iron deposition with disease progression in 20 patients older than 55 years with depression and on antidepressants, using magnetic resonance imaging (MRI)-based quantitative susceptibility mapping (QSM). Magnetic susceptibility values of the whole brain were compared between baseline and approximately one-year follow-up scans using permutation testing. Furthermore, we examined the relationship of changes between the susceptibility values and disease improvement using Spearman's partial correlation analysis, controlling for age, gender, and the visit interval. RESULTS Compared to the initial scan, increased magnetic susceptibility values were found in the medial prefrontal cortex (mPFC), dorsal anterior cingulate cortex (dACC), occipital areas, habenula, brainstem, and cerebellum (P<0.05, corrected). The susceptibility values decreased in the dorsal part of the mPFC, middle and posterior cingulate cortex (MCC and PCC), right postcentral gyrus, right inferior parietal lobule, right precuneus, right supramarginal gyrus, left lingual gyrus, left dorsal striatum, and right thalamus (P<0.05, corrected). Notably, the increase in susceptibility values at the mPFC and dACC negatively correlated with the changes in depression scores, as calculated using the Hamilton Depression Scale (HAMD) (r=-0.613, P=0.009), and the increase in susceptibility values at the cerebellum and habenula negatively correlated with the changes in cognitive scores, which were calculated using the Mini-Mental State Examination (MMSE) (cerebellum: r=-0.500, P=0.041; habenula: r=-0.588, P=0.013). Additionally, the decreased susceptibility values at the white matter near the mPFC (anterior corona radiata) also correlated with the changes in depression scores (r=-0.541, P=0.025), and the decreased susceptibility values at the left lingual gyrus correlated with the changes in cognitive scores (r=-0.613, P=0.009). CONCLUSIONS Our study identified brain areas where iron deposition changed with the progression of depression while on antidepressants. The linear relationship of changes in the magnetic susceptibility values in the mPFC, dACC, and some subcortical areas with changes in depression symptoms and cognitive functions of patients is highlighted. Our results strengthen the understanding of the alterations of brain iron levels associated with disease progression in patients with late-life depression.
Collapse
Affiliation(s)
- Fang Wang
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufei Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hengfen Gong
- Department of Psychiatry, Shanghai Pudong New Area Mental Health Center, Tongji University School of Medicine, Shanghai, China
| | - Jun Li
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yuyao Zhang
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
| | - Chencheng Zhang
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bomin Sun
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Naying He
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongjiang Wei
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
13
|
Reversal of genetic brain iron accumulation by N,N'-bis(2-mercaptoethyl)isophthalamide, a lipophilic metal chelator, in mice. Arch Toxicol 2022; 96:1951-1962. [PMID: 35445828 DOI: 10.1007/s00204-022-03287-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/21/2022] [Indexed: 12/31/2022]
Abstract
N,N'-bis(2-mercaptoethyl)isophthalamide (NBMI) is a novel lipophilic metal chelator and antioxidant used in mercury poisoning. Recent studies have suggested that NBMI may also bind to other metals such as lead and iron. Since NBMI can enter the brain, we evaluated if NBMI removes excess iron from the iron-loaded brain and ameliorates iron-induced oxidative stress. First, NBMI exhibited preferential binding to ferrous (Fe2+) iron with a negligible binding affinity to ferric (Fe3+) iron, indicating a selective chelation of labile iron. Second, NBMI protected SH-SY5Y human neuroblastoma cells from the cytotoxic effects of high iron. NBMI also decreased cellular labile iron and lessened the production of iron-induced reactive oxygen species in these cells. Deferiprone (DFP), a commonly used oral iron chelator, failed to prevent iron-induced cytotoxicity or labile iron accumulation. Next, we validated the efficacy of NBMI in Hfe H67D mutant mice, a mouse model of brain iron accumulation (BIA). Oral gavage of NBMI for 6 weeks decreased iron accumulation in the brain as well as liver, whereas DFP showed iron chelation only in the liver, but not in the brain. Notably, depletion of brain copper and anemia were observed in BIA mice treated with DFP, but not with NBMI, suggesting a superior safety profile of NBMI over DFP for long-term use. Collectively, our study demonstrates that NBMI provides a neuroprotective effect against BIA and has therapeutic potential for neurodegenerative diseases associated with BIA.
Collapse
|
14
|
Puy L, Leboullenger C, Auger F, Bordet R, Cordonnier C, Bérézowski V. Intracerebral Hemorrhage-Induced Cognitive Impairment in Rats Is Associated With Brain Atrophy, Hypometabolism, and Network Dysconnectivity. Front Neurosci 2022; 16:882996. [PMID: 35844211 PMCID: PMC9280302 DOI: 10.3389/fnins.2022.882996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
The mechanisms underlying intracerebral hemorrhage (ICH)-related cognitive impairment (CI) remain unclear. Long-term structural and functional changes were investigated in the brains of healthy male and female Wistar rats after experimental ICH. Following double injection of autologous blood, rats underwent short-term (onset, 3 and 7 days) and long-term (3 and 6 months) radiological assessment and behavioral tests exploring spontaneous locomotion, anxiety-like behavior and working memory, spatial recognition memory and visual recognition memory. Volumetric and metabolic changes in brain areas were examined by 7Tesla-MRI and [18F] FDG-PET, respectively. Brain connectomic disorders and maladaptive processes were seeked through brain metabolic connectivity analysis and atrophy-related network analysis. From an initial hematoma mean volume of 23.35 ± 9.50 mm3, we found early spontaneous locomotor recovery and significant spontaneous blood resorption (≈ 40% of the initial lesion) from days 0 to 7. After 3 and 6 months, ICH rats exhibited CI in several domains as compared to the sham group (working memory: 58.1 ± 1.2 vs. 70.7 ± 1.2%, p < 0.001; spatial recognition memory: 48.7 ± 1.9 vs. 64 ± 1.8%, p < 0.001 and visual recognition memory: 0.14 ± 0.05 vs. 0.33 ± 0.04, p = 0.013, in female only). Rats that experienced ICH had remote and concomitant cerebral atrophy and hypometabolism of ipsilateral striatum, thalamus, limbic system and cortical areas (temporal and parietal lobes). Interestingly, both structural and metabolic deterioration was found in the limbic system connected to the affected site, but remotely from the initial insult. On the other hand, increased activity and functional connectivity occurred in the contralateral hemisphere. These connectomics results showed that both maladaptative and compensation processes coexist in the rat brain following ICH, even at young age and in a disease-free setting. These radiological findings deepen our understanding of ICH-related CI and may serve as biomarkers in the view of future therapeutic intervention.
Collapse
Affiliation(s)
- Laurent Puy
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 – LilNCog - Lille Neuroscience and Cognition, Lille, France
| | - Clémence Leboullenger
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UMS 2014 - PLBS, Lille, France
| | - Florent Auger
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UMS 2014 - PLBS, Lille, France
| | - Régis Bordet
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 – LilNCog - Lille Neuroscience and Cognition, Lille, France
| | - Charlotte Cordonnier
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 – LilNCog - Lille Neuroscience and Cognition, Lille, France
- *Correspondence: Charlotte Cordonnier,
| | - Vincent Bérézowski
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 – LilNCog - Lille Neuroscience and Cognition, Lille, France
- UArtois, Lens, France
- Vincent Bérézowski,
| |
Collapse
|
15
|
Neuroinflammation in Friedreich's Ataxia. Int J Mol Sci 2022; 23:ijms23116297. [PMID: 35682973 PMCID: PMC9181348 DOI: 10.3390/ijms23116297] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 12/24/2022] Open
Abstract
Friedreich's ataxia (FRDA) is a rare genetic disorder caused by mutations in the gene frataxin, encoding for a mitochondrial protein involved in iron handling and in the biogenesis of iron-sulphur clusters, and leading to progressive nervous system damage. Although the overt manifestations of FRDA in the nervous system are mainly observed in the neurons, alterations in non-neuronal cells may also contribute to the pathogenesis of the disease, as recently suggested for other neurodegenerative disorders. In FRDA, the involvement of glial cells can be ascribed to direct effects caused by frataxin loss, eliciting different aberrant mechanisms. Iron accumulation, mitochondria dysfunction, and reactive species overproduction, mechanisms identified as etiopathogenic in neurons in FRDA, can similarly affect glial cells, leading them to assume phenotypes that can concur to and exacerbate neuron loss. Recent findings obtained in FRDA patients and cellular and animal models of the disease have suggested that neuroinflammation can accompany and contribute to the neuropathology. In this review article, we discuss evidence about the involvement of neuroinflammatory-related mechanisms in models of FRDA and provide clues for the modulation of glial-related mechanisms as a possible strategy to improve disease features.
Collapse
|
16
|
Hassan W, Noreen H, Rehman S, Kamal MA, Teixeira da Rocha JB. Association of Oxidative Stress with Neurological Disorders. Curr Neuropharmacol 2022; 20:1046-1072. [PMID: 34781871 PMCID: PMC9886831 DOI: 10.2174/1570159x19666211111141246] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/05/2021] [Accepted: 10/06/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGORUND Oxidative stress is one of the main contributing factors involved in cerebral biochemical impairment. The higher susceptibility of the central nervous system to reactive oxygen species mediated damage could be attributed to several factors. For example, neurons use a greater quantity of oxygen, many parts of the brain have higher concentraton of iron, and neuronal mitochondria produce huge content of hydrogen peroxide. In addition, neuronal membranes have polyunsaturated fatty acids, which are predominantly vulnerable to oxidative stress (OS). OS is the imbalance between reactive oxygen species generation and cellular antioxidant potential. This may lead to various pathological conditions and diseases, especially neurodegenerative diseases such as, Parkinson's, Alzheimer's, and Huntington's diseases. OBJECTIVES In this study, we explored the involvement of OS in neurodegenerative diseases. METHODS We used different search terms like "oxidative stress and neurological disorders" "free radicals and neurodegenerative disorders" "oxidative stress, free radicals, and neurological disorders" and "association of oxidative stress with the name of disorders taken from the list of neurological disorders. We tried to summarize the source, biological effects, and physiologic functions of ROS. RESULTS Finally, it was noted that more than 190 neurological disorders are associated with oxidative stress. CONCLUSION More elaborated studies in the future will certainly help in understanding the exact mechanism involved in neurological diseases and provide insight into revelation of therapeutic targets.
Collapse
Affiliation(s)
- Waseem Hassan
- Institute of Chemical Sciences, University of Peshawar, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Hamsa Noreen
- Institute of Chemical Sciences, University of Peshawar, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Shakila Rehman
- Institute of Chemical Sciences, University of Peshawar, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia
- Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770, Australia
| | - Joao Batista Teixeira da Rocha
- Departamento de Bioquímica e Biologia Molecular, Programa de Pós-Graduação em Bioquímica, Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS 97105-900, Brazil
| |
Collapse
|
17
|
Bardestani A, Ebrahimpour S, Esmaeili A, Esmaeili A. Quercetin attenuates neurotoxicity induced by iron oxide nanoparticles. J Nanobiotechnology 2021; 19:327. [PMID: 34663344 PMCID: PMC8522232 DOI: 10.1186/s12951-021-01059-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/24/2021] [Indexed: 01/19/2023] Open
Abstract
Iron oxide nanoparticles (IONPs) have been proposed as targeted carriers to deliver therapeutic molecules in the central nervous system (CNS). However, IONPs may damage neural tissue via free iron accumulation, protein aggregation, and oxidative stress. Neuroprotective effects of quercetin (QC) have been proven due to its antioxidant and anti-inflammatory properties. However, poor solubility and low bioavailability of QC have also led researchers to make various QC-involved nanoparticles to overcome these limitations. We wondered how high doses or prolonged treatment with quercetin conjugated superparamagnetic iron oxide nanoparticles (QCSPIONs) could improve cognitive dysfunction and promote neurogenesis without any toxicity. It can be explained that the QC inhibits protein aggregation and acts against iron overload via iron-chelating activity, iron homeostasis genes regulation, radical scavenging, and attenuation of Fenton/Haber-Weiss reaction. In this review, first, we present brain iron homeostasis, molecular mechanisms of iron overload that induced neurotoxicity, and the role of iron in dementia-associated diseases. Then by providing evidence of IONPs neurotoxicity, we discuss how QC neutralizes IONPs neurotoxicity, and finally, we make a brief comparison between QC and conventional iron chelators. In this review, we highlight that QC as supplementation and especially in conjugated form reduces iron oxide nanoparticles neurotoxicity in clinical application.
Collapse
Affiliation(s)
- Akram Bardestani
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, P.O. Box: 8174673441, Isfahan, Iran
| | - Shiva Ebrahimpour
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, P.O. Box: 8174673441, Isfahan, Iran
| | - Ali Esmaeili
- School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abolghasem Esmaeili
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, P.O. Box: 8174673441, Isfahan, Iran.
| |
Collapse
|
18
|
Se Thoe E, Fauzi A, Tang YQ, Chamyuang S, Chia AYY. A review on advances of treatment modalities for Alzheimer's disease. Life Sci 2021; 276:119129. [PMID: 33515559 DOI: 10.1016/j.lfs.2021.119129] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/10/2021] [Accepted: 01/19/2021] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease which is mainly characterized by progressive impairment in cognition, emotion, language and memory in older population. Considering the impact of AD, formulations of pharmaceutical drugs and cholinesterase inhibitors have been widely propagated, receiving endorsement by FDA as a form of AD treatment. However, these medications were gradually discovered to be ineffective in removing the root of AD pathogenesis but merely targeting the symptoms so as to improve a patient's cognitive outcome. Hence, a search for better disease-modifying alternatives is put into motion. Having a clear understanding of the neuroprotective mechanisms and diverse properties undertaken by specific genes, antibodies and nanoparticles is central towards designing novel therapeutic agents. In this review, we provide a brief introduction on the background of Alzheimer's disease, the biology of blood-brain barrier, along with the potentials and drawbacks associated with current therapeutic treatment avenues pertaining to gene therapy, immunotherapy and nanotherapy for better diagnosis and management of Alzheimer's disease.
Collapse
Affiliation(s)
- Ewen Se Thoe
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 47500 Selangor, Malaysia
| | - Ayesha Fauzi
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 47500 Selangor, Malaysia
| | - Yin Quan Tang
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 47500 Selangor, Malaysia
| | - Sunita Chamyuang
- School of Science, Mae Fah Luang University, Chaing Rai 57100, Thailand; Microbial Products and Innovation Research Group, Mae Fah Luang University, Chaing Rai 57100, Thailand
| | - Adeline Yoke Yin Chia
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 47500 Selangor, Malaysia.
| |
Collapse
|
19
|
Smethurst DGJ, Kovalev N, McKenzie ER, Pestov DG, Shcherbik N. Iron-mediated degradation of ribosomes under oxidative stress is attenuated by manganese. J Biol Chem 2020; 295:17200-17214. [PMID: 33040024 PMCID: PMC7863898 DOI: 10.1074/jbc.ra120.015025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 10/05/2020] [Indexed: 02/05/2023] Open
Abstract
Protein biosynthesis is fundamental to cellular life and requires the efficient functioning of the translational machinery. At the center of this machinery is the ribosome, a ribonucleoprotein complex that depends heavily on Mg2+ for structure. Recent work has indicated that other metal cations can substitute for Mg2+, raising questions about the role different metals may play in the maintenance of the ribosome under oxidative stress conditions. Here, we assess ribosomal integrity following oxidative stress both in vitro and in cells to elucidate details of the interactions between Fe2+ and the ribosome and identify Mn2+ as a factor capable of attenuating oxidant-induced Fe2+-mediated degradation of rRNA. We report that Fe2+ promotes degradation of all rRNA species of the yeast ribosome and that it is bound directly to RNA molecules. Furthermore, we demonstrate that Mn2+ competes with Fe2+ for rRNA-binding sites and that protection of ribosomes from Fe2+-mediated rRNA hydrolysis correlates with the restoration of cell viability. Our data, therefore, suggest a relationship between these two transition metals in controlling ribosome stability under oxidative stress.
Collapse
Affiliation(s)
- Daniel G J Smethurst
- Department of Cell Biology and Neuroscience, Rowan University, School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Nikolay Kovalev
- Department of Cell Biology and Neuroscience, Rowan University, School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Erica R McKenzie
- Civil and Environmental Engineering Department, Temple University, Philadelphia, Pennsylvania, USA
| | - Dimitri G Pestov
- Department of Cell Biology and Neuroscience, Rowan University, School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Natalia Shcherbik
- Department of Cell Biology and Neuroscience, Rowan University, School of Osteopathic Medicine, Stratford, New Jersey, USA.
| |
Collapse
|
20
|
Abedi M, Rahgozar S, Esmaeili A. Iron protects childhood acute lymphoblastic leukemia cells from methotrexate cytotoxicity. Cancer Med 2020; 9:3537-3550. [PMID: 32176452 PMCID: PMC7221302 DOI: 10.1002/cam4.2982] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/19/2020] [Accepted: 02/22/2020] [Indexed: 12/14/2022] Open
Abstract
Drug resistance is a fundamental clinical concern in pediatric acute lymphoblastic leukemia (pALL), and methotrexate (MTX) is an essential chemotherapy drug administered for the treatment. In the current study, the effect of iron in response to methotrexate and its underlying mechanisms were investigated in pALL cells. CCRF-CEM and Nalm6 cell lines were selected as T and B-ALL subtypes. Cells were pretreated with ferric ammonium citrate, exposed to the IC50 concentration of MTX and cell viability was assessed using MTT, colony formation, and flow cytometry assays. Iron-loaded cells were strongly resistant to MTX cytotoxicity. The inhibitory effect of N-acetyl cysteine to reverse the acquired MTX resistance was greater than that of the iron chelator, deferasirox, highlighting the importance of iron-mediated ROS in MTX resistance. Subsequently, the upregulation of BCL2, SOD2, NRF2, and MRP1 was confirmed using quantitative RT-PCR. Moreover, a positive correlation was demonstrated between the MRP1 expression levels and bone marrow iron storage in pALL patients. Further supporting our findings were the hematoxylin and eosin-stained histological sections showing that iron-treated nude mice xenografts demonstrated significantly more liver damage than those unexposed to iron. Overall, iron is introduced as a player with a novel role contributing to methotrexate resistance in pALL. Our findings suggest that the patients' bone marrow iron stores are necessary to be assessed during the chemotherapy, and transfusions should be carefully administrated.
Collapse
Affiliation(s)
- Marjan Abedi
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Soheila Rahgozar
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Abolghasem Esmaeili
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
21
|
Escobar I, Xu J, Jackson CW, Perez-Pinzon MA. Altered Neural Networks in the Papez Circuit: Implications for Cognitive Dysfunction after Cerebral Ischemia. J Alzheimers Dis 2020; 67:425-446. [PMID: 30584147 PMCID: PMC6398564 DOI: 10.3233/jad-180875] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cerebral ischemia remains a leading cause of mortality worldwide. Although the incidence of death has decreased over the years, surviving patients may suffer from long-term cognitive impairments and have an increased risk for dementia. Unfortunately, research aimed toward developing therapies that can improve cognitive outcomes following cerebral ischemia has proved difficult given the fact that little is known about the underlying processes involved. Nevertheless, mechanisms that disrupt neural network activity may provide valuable insight, since disturbances in both local and global networks in the brain have been associated with deficits in cognition. In this review, we suggest that abnormal neural dynamics within different brain networks may arise from disruptions in synaptic plasticity processes and circuitry after ischemia. This discussion primarily concerns disruptions in local network activity within the hippocampus and other extra-hippocampal components of the Papez circuit, given their role in memory processing. However, impaired synaptic plasticity processes and disruptions in structural and functional connections within the Papez circuit have important implications for alterations within the global network, as well. Although much work is required to establish this relationship, evidence thus far suggests there is a link. If pursued further, findings may lead toward a better understanding of how deficits in cognition arise, not only in cerebral ischemia, but in other neurological diseases as well.
Collapse
Affiliation(s)
- Iris Escobar
- Department of Neurology, Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL, USA.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jing Xu
- Department of Neurology, Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL, USA.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Charles W Jackson
- Department of Neurology, Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL, USA.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Miguel A Perez-Pinzon
- Department of Neurology, Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL, USA.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
22
|
Popugaeva E, Bezprozvanny I, Chernyuk D. Reversal of Calcium Dysregulation as Potential Approach for Treating Alzheimer's Disease. Curr Alzheimer Res 2020; 17:344-354. [PMID: 32469698 PMCID: PMC8210816 DOI: 10.2174/1567205017666200528162046] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 02/25/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Despite decades of research and effort, there is still no effective disease-modifying treatment for Alzheimer's Disease (AD). Most of the recent AD clinical trials were targeting amyloid pathway, but all these trials failed. Although amyloid pathology is a hallmark and defining feature of AD, targeting the amyloid pathway has been very challenging due to low efficacy and serious side effects. Alternative approaches or mechanisms for our understanding of the major cause of memory loss in AD need to be considered as potential therapeutic targets. Increasing studies suggest that Ca2+ dysregulation in AD plays an important role in AD pathology and is associated with other AD abnormalities, such as excessive inflammation, increased ROS, impaired autophagy, neurodegeneration, synapse, and cognitive dysfunction. Ca2+ dysregulation in cytosolic space, Endoplasmic Reticulum (ER) and mitochondria have been reported in the context of various AD models. Drugs or strategies, to correct the Ca2+ dysregulation in AD, have been demonstrated to be promising as an approach for the treatment of AD in preclinical models. This review will discuss the mechanisms of Ca2+ dysregulation in AD and associated pathology and discuss potential approaches or strategies to develop novel drugs for the treatment of AD by targeting Ca2+ dysregulation.
Collapse
Affiliation(s)
- Elena Popugaeva
- Department of Medical Physics, Laboratory of Molecular Neurodegeneration, Peter the Great St Petersburg Polytechnic University, St Petersburg, Russia
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center, Dallas, USA
| | - Daria Chernyuk
- Department of Medical Physics, Laboratory of Molecular Neurodegeneration, Peter the Great St Petersburg Polytechnic University, St Petersburg, Russia
| |
Collapse
|
23
|
Ye Q, Trivedi M, Zhang Y, Böhlke M, Alsulimani H, Chang J, Maher T, Deth R, Kim J. Brain iron loading impairs DNA methylation and alters GABAergic function in mice. FASEB J 2019; 33:2460-2471. [PMID: 30277817 PMCID: PMC6338660 DOI: 10.1096/fj.201801116rr] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 09/04/2018] [Indexed: 12/12/2022]
Abstract
Iron deficiency is closely associated with altered GABA metabolism and affective behavior. While mutation in the hemochromatosis ( HFE) gene disrupts iron homeostasis and promotes oxidative stress that increases the risk of neurodegeneration, it is largely unknown whether HFE mutation modifies GABAergic homeostasis and emotional behavior. The goal of our study was to investigate the impact of HFE on GABAergic neurochemistry and redox-epigenetic regulation in the brain using H67D HFE-mutant mice that recapitulates the H63D-HFE mutation in humans. H67D mice displayed elevated redox-active iron levels in the brain by 32% compared to age-matched wild-type mice. Moreover, the H67D brain had increased isoprostane and decreased glutathione, indicating elevated oxidative stress. Additionally, the H67D brain had decreased global methylation and attenuated DNA methyltransferase (DNMT) activity. Direct addition of iron to purified DNMT in vitro decreased enzyme activity in a concentration-dependent manner. Last, H67D mice exhibited decreased anxiety-like behavior, which was associated with increased expression of the GABAA receptor α2 subunits by 93%, and these changes were also observed in H67D mice fed a low-iron diet. Taken together, our results suggest a putative role of HFE in regulating labile iron status in the brain, and mutation in H67D perturbs redox-methylation status, contributing to GABAergic dysfunction.-Ye, Q., Trivedi, M., Zhang, Y., Böhlke, M., Alsulimani, H., Chang, J., Maher, T., Deth, R., Kim, J. Brain iron loading impairs DNA methylation and alters GABAergic function in mice.
Collapse
Affiliation(s)
- Qi Ye
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Malav Trivedi
- Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, Florida, USA; and
| | - Yiting Zhang
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Mark Böhlke
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Science (MCPHS) University, Boston, Massachusetts, USA
| | - Helal Alsulimani
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| | - JuOae Chang
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Timothy Maher
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Science (MCPHS) University, Boston, Massachusetts, USA
| | - Richard Deth
- Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, Florida, USA; and
| | - Jonghan Kim
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Alvarez-Cordoba M, Villanueva-Paz M, Villalón-García I, Povea-Cabello S, Suárez-Rivero JM, Talaverón-Rey M, Abril-Jaramillo J, Vintimilla-Tosi AB, Sánchez-Alcázar JA. Precision medicine in pantothenate kinase-associated neurodegeneration. Neural Regen Res 2019; 14:1177-1185. [PMID: 30804242 PMCID: PMC6425824 DOI: 10.4103/1673-5374.251203] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Neurodegeneration with brain iron accumulation is a broad term that describes a heterogeneous group of progressive and invalidating neurologic disorders in which iron deposits in certain brain areas, mainly the basal ganglia. The predominant clinical symptoms include spasticity, progressive dystonia, Parkinson’s disease-like symptoms, neuropsychiatric alterations, and retinal degeneration. Among the neurodegeneration with brain iron accumulation disorders, the most frequent subtype is pantothenate kinase-associated neurodegeneration (PKAN) caused by defects in the gene encoding the enzyme pantothenate kinase 2 (PANK2) which catalyzed the first reaction of the coenzyme A biosynthesis pathway. Currently there is no effective treatment to prevent the inexorable course of these disorders. The aim of this review is to open up a discussion on the utility of using cellular models derived from patients as a valuable tool for the development of precision medicine in PKAN. Recently, we have described that dermal fibroblasts obtained from PKAN patients can manifest the main pathological changes of the disease such as intracellular iron accumulation accompanied by large amounts of lipofuscin granules, mitochondrial dysfunction and a pronounced increase of markers of oxidative stress. In addition, PKAN fibroblasts showed a morphological senescence-like phenotype. Interestingly, pantothenate supplementation, the substrate of the PANK2 enzyme, corrected all pathophysiological alterations in responder PKAN fibroblasts with low/residual PANK2 enzyme expression. However, pantothenate treatment had no favourable effect on PKAN fibroblasts harbouring mutations associated with the expression of a truncated/incomplete protein. The correction of pathological alterations by pantothenate in individual mutations was also verified in induced neurons obtained by direct reprograming of PKAN fibroblasts. Our observations indicate that pantothenate supplementation can increase/stabilize the expression levels of PANK2 in specific mutations. Fibroblasts and induced neurons derived from patients can provide a useful tool for recognizing PKAN patients who can respond to pantothenate treatment. The presence of low but significant PANK2 expression which can be increased in particular mutations gives valuable information which can support the treatment with high dose of pantothenate. The evaluation of personalized treatments in vitro of fibroblasts and neuronal cells derived from PKAN patients with a wide range of pharmacological options currently available, and monitoring its effect on the pathophysiological changes, can help for a better therapeutic strategy. In addition, these cell models will be also useful for testing the efficacy of new therapeutic options developed in the future.
Collapse
Affiliation(s)
- Mónica Alvarez-Cordoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Marina Villanueva-Paz
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Irene Villalón-García
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Suleva Povea-Cabello
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Juan M Suárez-Rivero
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Marta Talaverón-Rey
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | | | | | - José A Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| |
Collapse
|
25
|
Sharma A, Pachauri V, Flora SJS. Advances in Multi-Functional Ligands and the Need for Metal-Related Pharmacology for the Management of Alzheimer Disease. Front Pharmacol 2018; 9:1247. [PMID: 30498443 PMCID: PMC6249274 DOI: 10.3389/fphar.2018.01247] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/12/2018] [Indexed: 01/07/2023] Open
Abstract
Alzheimer’s disease (AD) is the age linked neurodegenerative disorder with no disease modifying therapy currently available. The available therapy only offers short term symptomatic relief. Several hypotheses have been suggested for the pathogenesis of the disease while the molecules developed as possible therapeutic agent in the last decade, largely failed in the clinical trials. Several factors like tau protein hyperphosphorylation, amyloid-β (Aβ) peptide aggregation, decline in acetyl cholinesterase and oxidative stress might be contributing toward the pathogenesis of AD. Additionally, biometals dyshomeostasis (Iron, Copper, and Zinc) in the brain are also reported to be involved in the pathogenesis of AD. Thus, targeting these metal ions may be an effective strategy for the development of a drug to treat AD. Chelation therapy is currently employed for the metal intoxication but we lack a safe and effective chelating agents with additional biological properties for their possible use as multi target directed ligands for a complex disease like AD. Chelating agents possess the ability to disaggregate Aβ aggregation, dissolve amyloid plaques, and delay the cognitive impairment. Thus there is an urgent need to develop disease modifying therapeutic molecules with multiple beneficial features like targeting more than one factor responsible of the disease. These molecules, as disease modifying therapeutic agents for AD, should possess the potential to inhibit Aβ-metal interactions, the formation of toxic Aβ aggregates; and the capacity to reinstate metal homeostasis.
Collapse
Affiliation(s)
- Abha Sharma
- Department of Pharmacology and Toxicology and Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Raebareli, India
| | - Vidhu Pachauri
- Department of Pharmacology and Toxicology and Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Raebareli, India
| | - S J S Flora
- Department of Pharmacology and Toxicology and Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Raebareli, India
| |
Collapse
|
26
|
Pantothenate Rescues Iron Accumulation in Pantothenate Kinase-Associated Neurodegeneration Depending on the Type of Mutation. Mol Neurobiol 2018; 56:3638-3656. [PMID: 30173408 DOI: 10.1007/s12035-018-1333-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 08/24/2018] [Indexed: 12/18/2022]
Abstract
Neurodegeneration with brain iron accumulation (NBIA) is a group of inherited neurologic disorders in which iron accumulates in the basal ganglia resulting in progressive dystonia, spasticity, parkinsonism, neuropsychiatric abnormalities, and optic atrophy or retinal degeneration. The most prevalent form of NBIA is pantothenate kinase-associated neurodegeneration (PKAN) associated with mutations in the gene of pantothenate kinase 2 (PANK2), which is essential for coenzyme A (CoA) synthesis. There is no cure for NBIA nor is there a standard course of treatment. In the current work, we describe that fibroblasts derived from patients harbouring PANK2 mutations can reproduce many of the cellular pathological alterations found in the disease, such as intracellular iron and lipofuscin accumulation, increased oxidative stress, and mitochondrial dysfunction. Furthermore, mutant fibroblasts showed a characteristic senescent morphology. Treatment with pantothenate, the PANK2 enzyme substrate, was able to correct all pathological alterations in responder mutant fibroblasts with residual PANK2 enzyme expression. However, pantothenate had no effect on mutant fibroblasts with truncated/incomplete protein expression. The positive effect of pantothenate in particular mutations was also confirmed in induced neurons obtained by direct reprograming of mutant fibroblasts. Our results suggest that pantothenate treatment can stabilize the expression levels of PANK2 in selected mutations. These results encourage us to propose our screening model as a quick and easy way to detect pantothenate-responder patients with PANK2 mutations. The existence of residual enzyme expression in some affected individuals raises the possibility of treatment using high dose of pantothenate.
Collapse
|
27
|
Ferrous and ferric differentially deteriorate proliferation and differentiation of osteoblast-like UMR-106 cells. Biometals 2018; 31:873-889. [DOI: 10.1007/s10534-018-0130-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/10/2018] [Indexed: 12/29/2022]
|
28
|
Piloni NE, Caro AA, Puntarulo S. Iron overload prevents oxidative damage to rat brain after chlorpromazine administration. Biometals 2018; 31:561-570. [PMID: 29766365 DOI: 10.1007/s10534-018-0104-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 04/12/2018] [Indexed: 01/24/2023]
Abstract
The hypothesis tested is that Fe administration leads to a response in rat brain modulating the effects of later oxidative challenges such as chlorpromazine (CPZ) administration. Either a single dose (acute Fe overload) or 6 doses every second day (sub-chronic Fe overload) of 500 or 50 mg Fe-dextran/kg, respectively, were injected intraperitoneally (ip) to rats. A single dose of 10 mg CPZ/kg was injected ip 8 h after Fe treatment. DNA integrity was evaluated by quantitative PCR, lipid radical (LR·) generation rate by electron paramagnetic resonance (EPR), and catalase (CAT) activity by UV spectrophotometry in isolated brains. The maximum increase in total Fe brain was detected after 6 or 2 h in the acute and sub-chronic Fe overload model, respectively. Mitochondrial and nuclear DNA integrity decreased after acute Fe overload at the time of maximal Fe content; the decrease in DNA integrity was lower after sub-chronic than after acute Fe overload. CPZ administration increased LR· generation rate in control rat brain after 1 and 2 h; however, CPZ administration after acute or sub-chronic Fe overload did not affect LR· generation rate. CPZ treatment did not affect CAT activity after 1-4 h neither in control rats nor in acute Fe-overloaded rats. However, CPZ administration to rats treated sub-chronically with Fe showed increased brain CAT activity after 2 or 4 h, as compared to control values. Fe supplementation prevented brain damage in both acute and sub-chronic models of Fe overload by selectively activating antioxidant pathways.
Collapse
Affiliation(s)
- Natacha E Piloni
- Facultad de Farmacia y Bioquímica, Fisicoquímica-IBIMOL, Universidad de Buenos Aires, Junín 956, CAAD1113, Buenos Aires, Argentina.,Instituto de Bioquímica y Medicina Molecular (IBIMOL), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Andres A Caro
- Chemistry Department, Hendrix College, Conway, AR, 72032, USA
| | - Susana Puntarulo
- Facultad de Farmacia y Bioquímica, Fisicoquímica-IBIMOL, Universidad de Buenos Aires, Junín 956, CAAD1113, Buenos Aires, Argentina. .,Instituto de Bioquímica y Medicina Molecular (IBIMOL), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
29
|
Helley MP, Pinnell J, Sportelli C, Tieu K. Mitochondria: A Common Target for Genetic Mutations and Environmental Toxicants in Parkinson's Disease. Front Genet 2017; 8:177. [PMID: 29204154 PMCID: PMC5698285 DOI: 10.3389/fgene.2017.00177] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/01/2017] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is a devastating neurological movement disorder. Since its first discovery 200 years ago, genetic and environmental factors have been identified to play a role in PD development and progression. Although genetic studies have been the predominant driving force in PD research over the last few decades, currently only a small fraction of PD cases can be directly linked to monogenic mutations. The remaining cases have been attributed to other risk associated genes, environmental exposures and gene-environment interactions, making PD a multifactorial disorder with a complex etiology. However, enormous efforts from global research have yielded significant insights into pathogenic mechanisms and potential therapeutic targets for PD. This review will highlight mitochondrial dysfunction as a common pathway involved in both genetic mutations and environmental toxicants linked to PD.
Collapse
Affiliation(s)
- Martin P. Helley
- Department of Environmental Health Sciences, Florida International University, Miami, FL, United States
| | - Jennifer Pinnell
- Department of Environmental Health Sciences, Florida International University, Miami, FL, United States
- Peninsula Schools of Medicine and Dentistry, Plymouth University, Plymouth, United Kingdom
| | - Carolina Sportelli
- Department of Environmental Health Sciences, Florida International University, Miami, FL, United States
- Peninsula Schools of Medicine and Dentistry, Plymouth University, Plymouth, United Kingdom
| | - Kim Tieu
- Department of Environmental Health Sciences, Florida International University, Miami, FL, United States
| |
Collapse
|
30
|
Ivanova J, Petrova E, Kamenova K, Gluhcheva Y. Comparative effects of meso-2,3- dimercaptosuccinic acid, monensin, and salinomycin on cadmium-induced brain dysfunction in cadmium-intoxicated mice. Interdiscip Toxicol 2017; 10:107-113. [PMID: 30174534 PMCID: PMC6107650 DOI: 10.1515/intox-2017-0017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/02/2017] [Indexed: 12/15/2022] Open
Abstract
Cadmium (Cd) is a risk factor for neurodegenerative diseases. The purpose of this study was to compare the effects of meso-2,3-dimercaptosuccinic acid (DMSA) and the polyether ionophorous antibiotics monensin and salinomycin on Cd-induced neurodegenerative alterations in mice. The results show that subacute intoxication of mice with Cd (II) acetate (20 mg/kg body weight (BW) for 14 days) caused a significant accumulation of cadmium (Cd) in the brain. Treatment of Cd-exposed mice with DMSA (20 mg/kg BW for 14 days) significantly increased the Cd concentration in the brains compared to those of the Cd-treated group. However, administration of monensin (20 mg/kg BW for 14 days) or salinomycin (20 mg/kg BW for 14 days) significantly reduced the Cd concentration in the brains of Cd-treated mice compared to the toxic control group. Histopathological analysis of brain tissues from the Cd-treated mice revealed that Cd induced neuronal necrosis, characterized by many shrunken, darkly stained pyknotic neurons with prominent perineuronal spaces. Whereas monensin and salinomycin significantly reduced the adverse effects of Cd on brain morphology of Cd-treated mice, DMSA did not. Monensin slightly increased the copper and iron endogenous levels in the brains of Cd-exposed mice compared to those of the untreated mice. Salinomycin did not affect the concentrations of biometal ions in the brain of Cd-exposed mice compared to untreated controls. The results demonstrated salinomycin to be a better potential chelating agent for treatment of Cd-induced brain injury compared to DMSA and monensin.
Collapse
Affiliation(s)
- Juliana Ivanova
- Faculty of Medicine, Sofia University ‘St. Kliment Ohridski’, Kozjak Str., 1, 1407-Sofia, Bulgaria
| | - Emilia Petrova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Acad. Georgi Bonchev Str., Bl. 25, 1113-Sofia, Bulgaria
| | - Kalina Kamenova
- Faculty of Chemistry and Pharmacy, Sofia University ‘St. Kliment Ohridski’, J. Bourchier Ave., 1, 1164 - Sofia, Bulgaria
| | - Yordanka Gluhcheva
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Acad. Georgi Bonchev Str., Bl. 25, 1113-Sofia, Bulgaria
| |
Collapse
|
31
|
Yarjanli Z, Ghaedi K, Esmaeili A, Rahgozar S, Zarrabi A. Iron oxide nanoparticles may damage to the neural tissue through iron accumulation, oxidative stress, and protein aggregation. BMC Neurosci 2017; 18:51. [PMID: 28651647 PMCID: PMC5485499 DOI: 10.1186/s12868-017-0369-9] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 06/22/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In the recent decade, iron oxide nanoparticles (IONPs) have been proposed for several applications in the central nervous system (CNS), including targeting amyloid beta (Aβ) in the arteries, inhibiting the microglial cells, delivering drugs, and increasing contrast in magnetic resonance imaging. Conversely, a notable number of studies have reported the role of iron in neurodegenerative diseases. Therefore, this study has reviewed the recent studies to determine whether IONPs iron can threaten the cellular viability same as iron. RESULTS Iron contributes in Fenton's reaction and produces reactive oxygen species (ROS). ROS cause to damage the macromolecules and organelles of the cell via oxidative stress. Iron accumulation and oxidative stress are able to aggregate some proteins, including Aβ and α-synuclein, which play a critical role in Alzheimer's and Parkinson's diseases, respectively. Iron accumulation, oxidative stress, and protein aggregation make a positive feedback loop, which can be toxic for the cell. The release of iron ions from IONPs may result in iron accumulation in the targeted tissue, and thus, activate the positive feedback loop. However, the levels of IONPs induced toxicity depend on the size, concentration, surface charge, and the type of coating and functional groups of IONPs. CONCLUSION IONPs depending on their properties can lead to iron accumulation, oxidative stress and protein aggregation in the neural cells. Therefore, in order to apply IONPs in the CNS, the consideration of IONPs properties is crucial.
Collapse
Affiliation(s)
- Zahra Yarjanli
- Department of Biology, Faculty of Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| | - Kamran Ghaedi
- Department of Biology, Faculty of Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| | - Abolghasem Esmaeili
- Department of Biology, Faculty of Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| | - Soheila Rahgozar
- Department of Biology, Faculty of Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| | - Ali Zarrabi
- Department of Biotechnology, Faculty of Advanced Sciences and Technologies, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| |
Collapse
|
32
|
Skalny AV, Simashkova NV, Klyushnik TP, Grabeklis AR, Radysh IV, Skalnaya MG, Tinkov AA. Analysis of Hair Trace Elements in Children with Autism Spectrum Disorders and Communication Disorders. Biol Trace Elem Res 2017; 177:215-223. [PMID: 27785740 DOI: 10.1007/s12011-016-0878-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/14/2016] [Indexed: 12/24/2022]
Abstract
The primary objective of the present study is analysis of hair trace elements content in children with communication disorder (CD) and autism spectrum disorder (ASD). A total of 99 children from control, CD, and ASD groups (n = 33) were examined. All children were additionally divided into two subgroups according to age. Hair levels of trace elements were assessed using inductively coupled plasma mass spectrometry. The difference was considered significant at p < 0.01. The obtained data demonstrate that children with CD are characterized by significantly increased hair lithium (Li) (96 %; p = 0.008), selenium (Se) (66 %; p < 0.001), arsenic (As) (96 %; p = 0.005), beryllium (Be) (150 %; p < 0.001), and cadmium (Cd) (72 %; p = 0.007) content, being higher than the respective control values. In the ASD group, hair copper (Cu), iodine (I), and Be levels tended to be lower than the control values. In turn, the scalp hair content of Se significantly exceeded the control values (33 %; p = 0.004), whereas the level of iron (Fe) and aluminum (Al) tended to increase. After gradation for age, the most prominent differences in children with CD were detected in the elder group (5-8 years), whereas in the case of ASD-in the younger group (3-4 years old). Taking into account the role of hair as excretory mechanism for certain elements including the toxic ones, it can be proposed that children suffering from ASD are characterized by more profound alteration of metal handling and excretion in comparison to CD.
Collapse
Affiliation(s)
- Anatoly V Skalny
- All-Russian Research Institute of Medicinal and Aromatic Plants, Moscow, Russia.
- Orenburg State University, Orenburg, Russia.
- Yaroslavl State University, Yaroslavl, Russia.
- RUDN University, Moscow, Russia.
| | - Natalia V Simashkova
- Scientific Center for Mental Health, Russian Academy of Medical Sciences, Moscow, Russia
| | - Tatiana P Klyushnik
- Scientific Center for Mental Health, Russian Academy of Medical Sciences, Moscow, Russia
| | | | | | | | - Alexey A Tinkov
- Orenburg State University, Orenburg, Russia
- Yaroslavl State University, Yaroslavl, Russia
- RUDN University, Moscow, Russia
- Orenburg State Medical University, Orenburg, Russia
| |
Collapse
|
33
|
Shultz RB, Zhong Y. Minocycline targets multiple secondary injury mechanisms in traumatic spinal cord injury. Neural Regen Res 2017; 12:702-713. [PMID: 28616020 PMCID: PMC5461601 DOI: 10.4103/1673-5374.206633] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Minocycline hydrochloride (MH), a semi-synthetic tetracycline derivative, is a clinically available antibiotic and anti-inflammatory drug that also exhibits potent neuroprotective activities. It has been shown to target multiple secondary injury mechanisms in spinal cord injury, via its anti-inflammatory, anti-oxidant, and anti-apoptotic properties. The secondary injury mechanisms that MH can potentially target include inflammation, free radicals and oxidative stress, glutamate excitotoxicity, calcium influx, mitochondrial dysfunction, ischemia, hemorrhage, and edema. This review discusses the potential mechanisms of the multifaceted actions of MH. Its anti-inflammatory and neuroprotective effects are partially achieved through conserved mechanisms such as modulation of p38 mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/Akt signaling pathways as well as inhibition of matrix metalloproteinases (MMPs). Additionally, MH can directly inhibit calcium influx through the N-methyl-D-aspartate (NMDA) receptors, mitochondrial calcium uptake, poly(ADP-ribose) polymerase-1 (PARP-1) enzymatic activity, and iron toxicity. It can also directly scavenge free radicals. Because it can target many secondary injury mechanisms, MH treatment holds great promise for reducing tissue damage and promoting functional recovery following spinal cord injury.
Collapse
Affiliation(s)
- Robert B Shultz
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Yinghui Zhong
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
34
|
Mercadante CJ, Herrera C, Pettiglio MA, Foster ML, Johnson LC, Dorman DC, Bartnikas TB. The effect of high dose oral manganese exposure on copper, iron and zinc levels in rats. Biometals 2016; 29:417-22. [PMID: 26988220 PMCID: PMC5560020 DOI: 10.1007/s10534-016-9924-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/03/2016] [Indexed: 01/08/2023]
Abstract
Manganese is an essential dietary nutrient and trace element with important roles in mammalian development, metabolism, and antioxidant defense. In healthy individuals, gastrointestinal absorption and hepatobiliary excretion are tightly regulated to maintain systemic manganese concentrations at physiologic levels. Interactions of manganese with other essential metals following high dose ingestion are incompletely understood. We previously reported that gavage manganese exposure in rats resulted in higher tissue manganese concentrations when compared with equivalent dietary or drinking water manganese exposures. In this study, we performed follow-up evaluations to determine whether oral manganese exposure perturbs iron, copper, or zinc tissue concentrations. Rats were exposed to a control diet with 10 ppm manganese or dietary, drinking water, or gavage exposure to approximately 11.1 mg manganese/kg body weight/day for 7 or 61 exposure days. While manganese exposure affected levels of all metals, particularly in the frontal cortex and liver, copper levels were most prominently affected. This result suggests an under-appreciated effect of manganese exposure on copper homeostasis which may contribute to our understanding of the pathophysiology of manganese toxicity.
Collapse
Affiliation(s)
- Courtney J Mercadante
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Carolina Herrera
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Michael A Pettiglio
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Melanie L Foster
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, 27607, USA
| | - Laura C Johnson
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, 27607, USA
| | - David C Dorman
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, 27607, USA
| | - Thomas B Bartnikas
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
35
|
Abstract
Metals play important roles in the human body, maintaining cell structure and regulating gene expression, neurotransmission, and antioxidant response, to name a few. However, excessive metal accumulation in the nervous system may be toxic, inducing oxidative stress, disrupting mitochondrial function, and impairing the activity of numerous enzymes. Damage caused by metal accumulation may result in permanent injuries, including severe neurological disorders. Epidemiological and clinical studies have shown a strong correlation between aberrant metal exposure and a number of neurological diseases, including Alzheimer’s disease, amyotrophic lateral sclerosis, autism spectrum disorders, Guillain–Barré disease, Gulf War syndrome, Huntington’s disease, multiple sclerosis, Parkinson’s disease, and Wilson’s disease. Here, we briefly survey the literature relating to the role of metals in neurodegeneration.
Collapse
Affiliation(s)
- Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York, USA
| | - Mahfuzur Rahman Miah
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
36
|
Gueguen N, Desquiret-Dumas V, Leman G, Chupin S, Baron S, Nivet-Antoine V, Vessières E, Ayer A, Henrion D, Lenaers G, Reynier P, Procaccio V. Resveratrol Directly Binds to Mitochondrial Complex I and Increases Oxidative Stress in Brain Mitochondria of Aged Mice. PLoS One 2015; 10:e0144290. [PMID: 26684010 PMCID: PMC4694087 DOI: 10.1371/journal.pone.0144290] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/16/2015] [Indexed: 12/23/2022] Open
Abstract
Resveratrol is often described as a promising therapeutic molecule for numerous diseases, especially in metabolic and neurodegenerative disorders. While the mechanism of action is still debated, an increasing literature reports that resveratrol regulates the mitochondrial respiratory chain function. In a recent study we have identified mitochondrial complex I as a direct target of this molecule. Nevertheless, the mechanisms and consequences of such an interaction still require further investigation. In this study, we identified in silico by docking study a binding site for resveratrol at the nucleotide pocket of complex I. In vitro, using solubilized complex I, we demonstrated a competition between NAD+ and resveratrol. At low doses (<5μM), resveratrol stimulated complex I activity, whereas at high dose (50 μM) it rather decreased it. In vivo, in brain mitochondria from resveratrol treated young mice, we showed that complex I activity was increased, whereas the respiration rate was not improved. Moreover, in old mice with low antioxidant defenses, we demonstrated that complex I activation by resveratrol led to oxidative stress. These results bring new insights into the mechanism of action of resveratrol on mitochondria and highlight the importance of the balance between pro- and antioxidant effects of resveratrol depending on its dose and age. These parameters should be taken into account when clinical trials using resveratrol or analogues have to be designed.
Collapse
Affiliation(s)
- Naïg Gueguen
- Université d’Angers, Angers, F-49000, France
- Département de Biochimie et Génétique, CHU d’Angers, Angers, F-49000, France
- UMR CNRS 6214-INSERM U1083, Angers, F-49000, France
- * E-mail:
| | - Valérie Desquiret-Dumas
- Université d’Angers, Angers, F-49000, France
- Département de Biochimie et Génétique, CHU d’Angers, Angers, F-49000, France
- UMR CNRS 6214-INSERM U1083, Angers, F-49000, France
| | - Géraldine Leman
- Université d’Angers, Angers, F-49000, France
- UMR CNRS 6214-INSERM U1083, Angers, F-49000, France
| | - Stéphanie Chupin
- Université d’Angers, Angers, F-49000, France
- Département de Biochimie et Génétique, CHU d’Angers, Angers, F-49000, France
- UMR CNRS 6214-INSERM U1083, Angers, F-49000, France
| | - Stéphanie Baron
- EA 4466, Université Paris Descartes, Faculté de Pharmacie, Paris, F-75270, France
| | | | - Emilie Vessières
- Université d’Angers, Angers, F-49000, France
- UMR CNRS 6214-INSERM U1083, Angers, F-49000, France
| | - Audrey Ayer
- Université d’Angers, Angers, F-49000, France
- UMR CNRS 6214-INSERM U1083, Angers, F-49000, France
| | - Daniel Henrion
- Université d’Angers, Angers, F-49000, France
- UMR CNRS 6214-INSERM U1083, Angers, F-49000, France
| | - Guy Lenaers
- Université d’Angers, Angers, F-49000, France
- UMR CNRS 6214-INSERM U1083, Angers, F-49000, France
| | - Pascal Reynier
- Université d’Angers, Angers, F-49000, France
- Département de Biochimie et Génétique, CHU d’Angers, Angers, F-49000, France
- UMR CNRS 6214-INSERM U1083, Angers, F-49000, France
| | - Vincent Procaccio
- Université d’Angers, Angers, F-49000, France
- Département de Biochimie et Génétique, CHU d’Angers, Angers, F-49000, France
- UMR CNRS 6214-INSERM U1083, Angers, F-49000, France
| |
Collapse
|
37
|
Menon AV, Chang J, Kim J. Mechanisms of divalent metal toxicity in affective disorders. Toxicology 2015; 339:58-72. [PMID: 26551072 DOI: 10.1016/j.tox.2015.11.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/19/2015] [Accepted: 11/03/2015] [Indexed: 01/01/2023]
Abstract
Metals are required for proper brain development and play an important role in a number of neurobiological functions. The divalent metal transporter 1 (DMT1) is a major metal transporter involved in the absorption and metabolism of several essential metals like iron and manganese. However, non-essential divalent metals are also transported through this transporter. Therefore, altered expression of DMT1 can modify the absorption of toxic metals and metal-induced toxicity. An accumulating body of evidence has suggested that increased metal stores in the brain are associated with elevated oxidative stress promoted by the ability of metals to catalyze redox reactions, resulting in abnormal neurobehavioral function and the progression of neurodegenerative diseases. Metal overload has also been implicated in impaired emotional behavior, although the underlying mechanisms are not well understood with limited information. The current review focuses on psychiatric dysfunction associated with imbalanced metabolism of metals that are transported by DMT1. The investigations with respect to the toxic effects of metal overload on behavior and their underlying mechanisms of toxicity could provide several new therapeutic targets to treat metal-associated affective disorders.
Collapse
Affiliation(s)
| | - JuOae Chang
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Jonghan Kim
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
38
|
Grishina EV, Khaustova YV, Vasilieva AA, Mayevsky EI. Age-related peculiarities of succinate effect on induced lipid peroxidation in rat liver mitochondria. Biophysics (Nagoya-shi) 2015. [DOI: 10.1134/s0006350915040119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
39
|
Chtourou Y, Slima AB, Gdoura R, Fetoui H. Naringenin Mitigates Iron-Induced Anxiety-Like Behavioral Impairment, Mitochondrial Dysfunctions, Ectonucleotidases and Acetylcholinesterase Alteration Activities in Rat Hippocampus. Neurochem Res 2015; 40:1563-75. [PMID: 26050208 DOI: 10.1007/s11064-015-1627-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 05/21/2015] [Accepted: 05/25/2015] [Indexed: 01/12/2023]
Abstract
Studies demonstrated that the iron chelating antioxidant restores brain dysfunction induced by iron toxicity in animals. Earlier, we found that iron overload-induced cerebral cortex apoptosis correlated with oxidative stress could be protected by naringenin (NGEN). In this respect, the present study is focused on the mechanisms associated with the protective efficacy of NGEN, natural flavonoid compound abundant in the peels of citrus fruit, on iron induced impairment of the anxiogenic-like behaviour, purinergic and cholinergic dysfunctions with oxidative stress related disorders on mitochondrial function in the rat hippocampus. Results showed that administration of NGEN (50 mg/kg/day) by gavage significantly ameliorated anxiogenic-like behaviour impairment induced by the exposure to 50 mg of Fe-dextran/kg/day intraperitoneally for 28 days in rats, decreased iron-induced reactive oxygen species formation and restored the iron-induced decrease of the acetylcholinesterase expression level, mitochondrial membrane potential and mitochondrial complexes activities in the hippocampus of rats. Moreover, NGEN was able to restore the alteration on the activity and expression of ectonucleotidases such as adenosine triphosphate diphosphohydrolase and 5'-nucleotidase, enzymes which hydrolyze and therefore control extracellular ATP and adenosine concentrations in the synaptic cleft. These results may contribute to a better understanding of the neuroprotective role of NGEN, emphasizing the influence of including this flavonoid in the diet for human health, possibly preventing brain injury associated with iron overload.
Collapse
Affiliation(s)
- Yassine Chtourou
- Toxicology-Microbiology and Environmental Health Unit (UR11ES70), Life Sciences Department, Faculty of Sciences, University of Sfax, Street Soukra Km 3.5, BP 1171, 3000, Sfax, Tunisia,
| | | | | | | |
Collapse
|
40
|
Dubinina EE, Schedrina LV, Neznanov NG, Zalutskaya NM, Zakharchenko DV. [Oxidative stress and its effect on cells functional activity of alzheimer's disease]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2015; 61:57-69. [PMID: 25762599 DOI: 10.18097/pbmc20156101057] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The paper summarizes literature data on the importance of oxidative stress as one of the pathogenetic mechanisms in Alzheimer's disease. The paper describes the main specific and nonspecific ways of reactive oxygen species generation in the course of the disease development. The effect of reactive oxygen species generated by the functional activity of cells, i.e. apoptosis and mitotic cycle, is shown. The role of the regulatory system of nodal cells is performed by phosphorylation/dephosphorylation process which is associated with intense phosphorylation of tau protein and mitosis-specific proteins. In Alzheimer's disease, the regulating function of peptidyl-prolyl isomerases in particular of Pin1 associated with maintaining a balanced state of phosphorylation/dephosphorylation processes is disturbed. Taking into consideration the multifactorial impairment of the cell cycle control, this process should be considered from the standpoint of the general state of metabolic processes, and oxidative stress has one of the key positions in aging.
Collapse
|
41
|
Chen X, Guo C, Kong J. Oxidative stress in neurodegenerative diseases. Neural Regen Res 2015; 7:376-85. [PMID: 25774178 PMCID: PMC4350122 DOI: 10.3969/j.issn.1673-5374.2012.05.009] [Citation(s) in RCA: 220] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Accepted: 11/22/2011] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species are constantly produced in aerobic organisms as by-products of normal oxygen metabolism and include free radicals such as superoxide anion (O2−) and hydroxyl radical (OH−), and non-radical hydrogen peroxide (H2O2). The mitochondrial respiratory chain and enzymatic reactions by various enzymes are endogenous sources of reactive oxygen species. Exogenous reactive oxygen species -inducing stressors include ionizing radiation, ultraviolet light, and divergent oxidizing chemicals. At low concentrations, reactive oxygen species serve as an important second messenger in cell signaling; however, at higher concentrations and long-term exposure, reactive oxygen species can damage cellular macromolecules such as DNA, proteins, and lipids, which leads to necrotic and apoptotic cell death. Oxidative stress is a condition of imbalance between reactive oxygen species formation and cellular antioxidant capacity due to enhanced ROS generation and/or dysfunction of the antioxidant system. Biochemical alterations in these macromolecular components can lead to various pathological conditions and human diseases, especially neurodegenerative diseases. Neurodegenerative diseases are morphologically featured by progressive cell loss in specific vulnerable neuronal cells, often associated with cytoskeletal protein aggregates forming inclusions in neurons and/or glial cells. Deposition of abnormal aggregated proteins and disruption of metal ions homeostasis are highly associated with oxidative stress. The main aim of this review is to present as much detailed information as possible that is available on various neurodegenerative disorders and their connection with oxidative stress. A variety of therapeutic strategies designed to address these pathological processes are also described. For the future therapeutic direction, one specific pathway that involves the transcription factor nuclear factor erythroid 2-related factor 2 is receiving considerable attention.
Collapse
Affiliation(s)
- Xueping Chen
- Department of Human Anatomy and Cell Science, University of Manitoba, Manitoba, Canada
| | - Chunyan Guo
- Department of Pharmacy, Hebei North University, Zhangjiakou 075000, Hebei Province, China
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, University of Manitoba, Manitoba, Canada
| |
Collapse
|
42
|
Abstract
No disease modifying therapy exists for Alzheimer's disease (AD). The growing burden of this disease to our society necessitates continued investment in drug development. Over the last decade, multiple phase 3 clinical trials testing drugs that were designed to target established disease mechanisms of AD have all failed to benefit patients. There is, therefore, a need for new treatment strategies. Changes to the transition metals, zinc, copper, and iron, in AD impact on the molecular mechanisms of disease, and targeting these metals might be an alternative approach to treat the disease. Here we review how metals feature in molecular mechanisms of AD, and we describe preclinical and clinical data that demonstrate the potential for metal-based drug therapy.
Collapse
Affiliation(s)
- Scott Ayton
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, 3052 VIC Australia
| | - Peng Lei
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, 3052 VIC Australia
| | - Ashley I. Bush
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, 3052 VIC Australia
| |
Collapse
|
43
|
Janaway BM, Simpson JE, Hoggard N, Highley JR, Forster G, Drew D, Gebril OH, Matthews FE, Brayne C, Wharton SB, Ince PG. Brain haemosiderin in older people: pathological evidence for an ischaemic origin of magnetic resonance imaging (MRI) microbleeds. Neuropathol Appl Neurobiol 2014; 40:258-69. [PMID: 23678850 PMCID: PMC4282337 DOI: 10.1111/nan.12062] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 05/10/2013] [Indexed: 01/03/2023]
Abstract
Introduction Magnetic resonance imaging (MRI) cerebral microbleeds (CMB) arise from ferromagnetic haemosiderin iron assumed to derive from extravasation of erythrocytes. Light microscopy of ageing brain frequently reveals foci of haemosiderin from single crystalloids to larger, predominantly perivascular, aggregates. The pathological and radiological relationship between these findings is not resolved. Methods Haemosiderin deposition and vascular pathology in the putamen were quantified in 200 brains donated to the population-representative Medical Research Council Cognitive Function and Ageing Study. Molecular markers of gliosis and tissue integrity were assessed by immunohistochemistry in brains with highest (n = 20) and lowest (n = 20) levels of putamen haemosiderin. The association between haemosiderin counts and degenerative and vascular brain pathology, clinical data, and the haemochromatosis (HFE) gene H63D genotype were analysed. The frequency of MRI CMB in 10 cases with highest and lowest burden of putamen haemosiderin, was compared using post mortem 3T MRI. Results Greater putamen haemosiderin was significantly associated with putaminal indices of small vessel ischaemia (microinfarcts, P < 0.05; arteriolosclerosis, P < 0.05; perivascular attenuation, P < 0.001) and with lacunes in any brain region (P < 0.023) but not large vessel disease, or whole brain measures of neurodegenerative pathology. Higher levels of putamen haemosiderin correlated with more CMB (P < 0.003). Conclusions The MRI-CMB concept should take account of brain iron homeostasis, and small vessel ischaemic change in later life, rather than only as a marker for minor episodes of cerebrovascular extravasation. These data are of clinical relevance, suggesting that basal ganglia MRI microbleeds may be a surrogate for ischaemic small vessel disease rather than exclusively a haemorrhagic diathesis.
Collapse
Affiliation(s)
- B M Janaway
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Wei S, Shi W, Li M, Gao Q. Calorie restriction down-regulates expression of the iron regulatory hormone hepcidin in normal and D-galactose-induced aging mouse brain. Rejuvenation Res 2014; 17:19-26. [PMID: 24044515 DOI: 10.1089/rej.2013.1450] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It has been shown that iron progressively accumulates in the brain with age. Calorie restriction (CR) may allay many of the adverse effects of aging on the brain, yet the underlying mechanisms, in particular in relation to brain iron metabolism, remain unclear. This study aimed to investigate the role of CR in the regulation of cerebral cellular iron homeostasis. C57BL/6 mice were randomly divided into four groups of eight. The control group was fed a conventional diet ad libitum; the CR group received 70% of the calories of the control mouse intake per day; the D-galactose (D-gal) group received subcutaneous injection of D-gal at a dose of 100 mg/kg once daily to produce mouse model of aging; the D-gal plus CR group received both of the two interventions for 14 weeks. The Morris water maze (MWM) was employed to test the cognitive performance of all animals, and the expression of iron regulatory genes, ferroportin and hepcidin, in the cortex and hippocampus were detected by quantitative real-time PCR. Compared to the controls, the D-gal group mice showed significant spatial reference memory deficits in the MWM test, whereas the D-gal-CR group mice exhibited almost normal cognitive function, indicating that CR protects against D-gal-induced learning and memory impairment. Hepcidin mRNA expression was increased in the D-gal group, decreased in the CR group, and was basically unchanged in the D-gal-CR group. There was no statistical difference in the transmembrane iron exporter ferroportin expression between control and any of the experimental groups. The results suggest that the anti-aging effects of CR might partially lie in its capacity to reduce or avoid age-related iron accumulation in the brain through down-regulating expression of brain hepcidin--the key negative regulator for intracellular iron efflux--and that facilitating the balance of brain iron metabolism may be a promising anti-aging measure.
Collapse
Affiliation(s)
- Shougang Wei
- 1 Municipal Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University , Beijing, China
| | | | | | | |
Collapse
|
45
|
Oxidative Stress Gated by Fenton and Haber Weiss Reactions and Its Association With Alzheimer’s Disease. ARCHIVES OF NEUROSCIENCE 2014. [DOI: 10.5812/archneurosci.20078] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
46
|
Di Paola M, Phillips OR, Sanchez-Castaneda C, Di Pardo A, Maglione V, Caltagirone C, Sabatini U, Squitieri F. MRI measures of corpus callosum iron and myelin in early Huntington's disease. Hum Brain Mapp 2014; 35:3143-51. [PMID: 24895252 PMCID: PMC6869772 DOI: 10.1002/hbm.22391] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 07/19/2013] [Accepted: 07/29/2013] [Indexed: 11/09/2022] Open
Abstract
Increased iron in subcortical gray matter (GM) structures of patients with Huntington's disease (HD) has been suggested as a causal factor in neuronal degeneration. But how iron content is related to white matter (WM) changes in HD is still unknown. For example, it is not clear whether WM changes share the same physiopathology (i.e. iron accumulation) with GM or whether there is a different mechanism. The present study used MRI to examine iron content in premanifest gene carriers (PreHD, n = 25) and in early HD patients (n = 25) compared with healthy controls (n = 50). 3T MRI acquisitions included high resolution 3D T1, EPI sequences for diffusion tensor imaging (DTI) as an indirect measure of tissue integrity, and T2*-weighted gradient echo-planar imaging for MR-based relaxometry (R2*), which provides an indirect measure of ferritin/iron deposition in the brain. Myelin breakdown starts in the PreHD stage, but there is no difference in iron content values. Iron content reduction manifests later, in the early HD stage, in which we found a lower R2* parameter value in the isthmus. The WM iron reduction in HD is temporally well-defined (no iron differences in PreHD subjects and iron differences only in early HD patients). Iron level in callosal WM may be regarded as a marker of disease state, as iron does not differentiate PreHD subjects from controls but distinguishes between PreHD and HD.
Collapse
Affiliation(s)
- M Di Paola
- Department of Clinical and Behavioural Neurology, IRCCS Santa Lucia Foundation, Rome, Italy; Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Sharifi S, Nederveen AJ, Booij J, van Rootselaar AF. Neuroimaging essentials in essential tremor: a systematic review. NEUROIMAGE-CLINICAL 2014; 5:217-31. [PMID: 25068111 PMCID: PMC4110352 DOI: 10.1016/j.nicl.2014.05.003] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 05/03/2014] [Accepted: 05/05/2014] [Indexed: 01/04/2023]
Abstract
Background Essential tremor is regarded to be a disease of the central nervous system. Neuroimaging is a rapidly growing field with potential benefits to both diagnostics and research. The exact role of imaging techniques with respect to essential tremor in research and clinical practice is not clear. A systematic review of the different imaging techniques in essential tremor is lacking in the literature. Methods We performed a systematic literature search combining the terms essential tremor and familial tremor with the following keywords: imaging, MRI, VBM, DWI, fMRI, PET and SPECT, both in abbreviated form as well as in full form. We summarize and discuss the quality and the external validity of each study and place the results in the context of existing knowledge regarding the pathophysiology of essential tremor. Results A total of 48 neuroimaging studies met our search criteria, roughly divided into 19 structural and 29 functional and metabolic studies. The quality of the studies varied, especially concerning inclusion criteria. Functional imaging studies indicated cerebellar hyperactivity during rest and during tremor. The studies also pointed to the involvement of the thalamus, the inferior olive and the red nucleus. Structural studies showed less consistent results. Discussion and conclusion Neuroimaging techniques in essential tremor give insight into the pathophysiology of essential tremor indicating the involvement of the cerebellum as the most consistent finding. GABAergic dysfunction might be a major premise in the pathophysiological hypotheses. Inconsistencies between studies can be partly explained by the inclusion of heterogeneous patient groups. Improvement of scientific research requires more stringent inclusion criteria and application of advanced analysis techniques. Also, the use of multimodal neuroimaging techniques is a promising development in movement disorders research. Currently, the role of imaging techniques in essential tremor in daily clinical practice is limited. We conducted a systematic review of neuroimaging studies in essential tremor. Cerebellar involvement is the most consistent finding. GABAergic dysfunction is worthwhile investigating more intensively. We encourage multimodal neuroimaging focussing on brain networks.
Collapse
Affiliation(s)
- Sarvi Sharifi
- Department of Neurology, Academic Medical Center, Amsterdam, The Netherlands ; Brain Imaging Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Aart J Nederveen
- Brain Imaging Center, Academic Medical Center, Amsterdam, The Netherlands ; Department of Radiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Jan Booij
- Brain Imaging Center, Academic Medical Center, Amsterdam, The Netherlands ; Department of Nuclear Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Anne-Fleur van Rootselaar
- Department of Neurology, Academic Medical Center, Amsterdam, The Netherlands ; Brain Imaging Center, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
48
|
Mariani S, Ventriglia M, Simonelli I, Spalletta G, Bucossi S, Siotto M, Assogna F, Melgari JM, Vernieri F, Squitti R. Effects of hemochromatosis and transferrin gene mutations on peripheral iron dyshomeostasis in mild cognitive impairment and Alzheimer's and Parkinson's diseases. Front Aging Neurosci 2013; 5:37. [PMID: 23935582 PMCID: PMC3733023 DOI: 10.3389/fnagi.2013.00037] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/01/2013] [Indexed: 12/31/2022] Open
Abstract
Deregulation of iron metabolism has been observed in patients with neurodegenerative diseases. We have carried out a molecular analysis investigating the interaction between iron specific gene variants [transferrin (TF, P589S), hemochromatosis (HFE) C282Y and (H63D)], iron biochemical variables [iron, Tf, ceruloplasmin (Cp), Cp:Tf ratio and % of Tf saturation (% Tf-sat)] and apolipoprotein E (APOE) gene variants in 139 Alzheimer's disease (AD), 27 Mild Cognitive Impairment (MCI), 78 Parkinson's disease (PD) patients and 139 healthy controls to investigate mechanisms of iron regulation or toxicity. No difference in genetic variant distributions between patients and controls was found in our Italian sample, but the stratification for the APOEε4 allele revealed that among the APOEε4 carriers was higher the frequency of those carriers of at least a mutated TF P589S allele. Decreased Tf in both AD and MCI and increased Cp:Tf ratio in AD vs. controls were detected. A multinomial logistic regression model revealed that increased iron and Cp:Tf ratio and being man instead of woman increased the risk of having PD, that increased values of Cp:Tf ratio corresponded to a 4-fold increase of the relative risk of having MCI, while higher Cp levels were protective for PD and MCI. Our study has some limitations: the small size of the samples, one ethnic group considered, the rarity of some alleles which prevent the statistical power of some genetic analysis. Even though they need confirmation in larger cohorts, our data suggest the hypothesis that deregulation of iron metabolism, in addition to other factors, has some effect on the PD disease risk.
Collapse
Affiliation(s)
- S Mariani
- Neurology, University "Campus Biomedico" Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Uranga RM, Katz S, Salvador GA. Enhanced phosphatidylinositol 3-kinase (PI3K)/Akt signaling has pleiotropic targets in hippocampal neurons exposed to iron-induced oxidative stress. J Biol Chem 2013; 288:19773-84. [PMID: 23687303 DOI: 10.1074/jbc.m113.457622] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The PI3K/Akt pathway is a key component in synaptic plasticity and neuronal survival. The aim of this work was to investigate the participation of the PI3K/Akt pathway and its outcome on different molecular targets such as glycogen synthase kinase 3β (GSK3β) and Forkhead box-O (FoxO) transcription factors during mild oxidative stress triggered by iron overload. The exposure of mouse hippocampal neurons (HT22) to different concentrations of Fe(2+) (25-200 μm) for 24 h led us to define a mild oxidative injury status (50 μm Fe(2+)) in which cell morphology showed changes typical of neuronal damage with increased lipid peroxidation and cellular oxidant levels but no alteration of cellular viability. There was a simultaneous increase in both Akt and GSK3β phosphorylation. Levels of phospho-FoxO3a (inactive form) increased in the cytosolic fraction of cells treated with iron in a PI3K-dependent manner. Moreover, PI3K and Akt translocated to the nucleus in response to oxidative stress. Iron-overloaded cells harboring a constitutively active form of Akt showed decreased oxidants levels. Indeed, GSH synthesis under oxidative stress conditions was regulated by activated Akt. Our results show that activation of the PI3K/Akt pathway during iron-induced neurotoxicity regulates multiple targets such as GSK3β, FoxO transcriptional activity, and glutathione metabolism, thus modulating the neuronal response to oxidative stress.
Collapse
Affiliation(s)
- Romina María Uranga
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur, 8000 Bahía Blanca, Argentina
| | | | | |
Collapse
|
50
|
Novellino F, Cherubini A, Chiriaco C, Morelli M, Salsone M, Arabia G, Quattrone A. Brain iron deposition in essential tremor: a quantitative 3-Tesla magnetic resonance imaging study. Mov Disord 2012; 28:196-200. [PMID: 23238868 DOI: 10.1002/mds.25263] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 09/17/2012] [Accepted: 09/27/2012] [Indexed: 01/18/2023] Open
Abstract
Studies have demonstrated brain iron deposition in neurodegenerative disease and in normal aging. Data on this topic are lacking in essential tremor (ET). The aim of our study was to investigate brain iron content in patients with ET, using quantitative magnetic resonance imaging (MRI) T2*-relaxometry. We enrolled 24 patients with ET and 25 age-matched healthy controls. Subjects were examined using a 3T MRI scanner. The protocol included conventional MRI sequences and quantitative T2*-relaxometry. Whole-brain voxel-based analyses showed significant differences in T2* values in bilateral globus pallidus, substantia nigra, and in right dentate nucleus (P < .001 uncorrected). In the bilateral pallidum, differences survived family-wise-error (FWE) correction for multiple comparisons (P < .05). The present study provides the first evidence of increased brain iron accumulation in ET patients. Our results are suggestive of a possible involvement of motor systems outside of the cerebellum/cerebellar pathway and, more specifically, of the globus pallidus.
Collapse
Affiliation(s)
- Fabiana Novellino
- Neuroimaging Research Unit, National Research Council, Catanzaro, Italy
| | | | | | | | | | | | | |
Collapse
|