1
|
Merenstein JL, Zhao J, Madden DJ. Depthwise cortical iron relates to functional connectivity and fluid cognition in healthy aging. Neurobiol Aging 2025; 148:27-40. [PMID: 39893877 DOI: 10.1016/j.neurobiolaging.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/28/2024] [Accepted: 01/08/2025] [Indexed: 02/04/2025]
Abstract
Age-related differences in fluid cognition have been associated with both the merging of functional brain networks, defined from resting-state functional magnetic resonance imaging (rsfMRI), and with elevated cortical iron, assessed by quantitative susceptibility mapping (QSM). Limited information is available, however, regarding the depthwise profile of cortical iron and its potential relation to functional connectivity. Here, using an adult lifespan sample (n = 138; 18-80 years), we assessed relations among graph theoretical measures of functional connectivity, column-based depthwise measures of cortical iron, and fluid cognition (i.e., tests of memory, perceptual-motor speed, executive function). Increased age was related both to less segregated functional networks and to increased cortical iron, especially for superficial depths. Functional network segregation mediated age-related differences in memory, whereas depthwise iron mediated age-related differences in general fluid cognition. Lastly, higher mean parietal iron predicted lower network segregation for adults younger than 45 years of age. These findings suggest that functional connectivity and depthwise cortical iron have distinct, complementary roles in the relation between age and fluid cognition in healthy adults.
Collapse
Affiliation(s)
- Jenna L Merenstein
- Brain Imaging and Analysis Center, Duke University Medical Center, Durham, NC 27710, USA.
| | - Jiayi Zhao
- Brain Imaging and Analysis Center, Duke University Medical Center, Durham, NC 27710, USA
| | - David J Madden
- Brain Imaging and Analysis Center, Duke University Medical Center, Durham, NC 27710, USA; Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA; Center for Cognitive Neuroscience, Duke University, Durham, NC 27708, USA
| |
Collapse
|
2
|
Cohen Z, Lau L, Ahmed M, Jack CR, Liu C. Quantitative susceptibility mapping in the brain reflects spatial expression of genes involved in iron homeostasis and myelination. Hum Brain Mapp 2024; 45:e26688. [PMID: 38896001 PMCID: PMC11187871 DOI: 10.1002/hbm.26688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 06/21/2024] Open
Abstract
Quantitative susceptibility mapping (QSM) is an MRI modality used to non-invasively measure iron content in the brain. Iron exhibits a specific anatomically varying pattern of accumulation in the brain across individuals. The highest regions of accumulation are the deep grey nuclei, where iron is stored in paramagnetic molecule ferritin. This form of iron is considered to be what largely contributes to the signal measured by QSM in the deep grey nuclei. It is also known that QSM is affected by diamagnetic myelin contents. Here, we investigate spatial gene expression of iron and myelin related genes, as measured by the Allen Human Brain Atlas, in relation to QSM images of age-matched subjects. We performed multiple linear regressions between gene expression and the average QSM signal within 34 distinct deep grey nuclei regions. Our results show a positive correlation (p < .05, corrected) between expression of ferritin and the QSM signal in deep grey nuclei regions. We repeated the analysis for other genes that encode proteins thought to be involved in the transport and storage of iron in the brain, as well as myelination. In addition to ferritin, our findings demonstrate a positive correlation (p < .05, corrected) between the expression of ferroportin, transferrin, divalent metal transporter 1, several gene markers of myelinating oligodendrocytes, and the QSM signal in deep grey nuclei regions. Our results suggest that the QSM signal reflects both the storage and active transport of iron in the deep grey nuclei regions of the brain.
Collapse
Affiliation(s)
- Zoe Cohen
- Department of Electrical Engineering and Computer SciencesUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Laurance Lau
- Department of Electrical Engineering and Computer SciencesUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Maruf Ahmed
- Department of Electrical Engineering and Computer SciencesUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Clifford R. Jack
- Mayo Foundation for Medical Education and ResearchRochesterMinnesotaUSA
| | - Chunlei Liu
- Department of Electrical Engineering and Computer SciencesUniversity of California, BerkeleyBerkeleyCaliforniaUSA
- Helen Wills Neuroscience InstituteUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| |
Collapse
|
3
|
Irrsack E, Aydin S, Bleckmann K, Schuller J, Dringen R, Koch M. Local Administrations of Iron Oxide Nanoparticles in the Prefrontal Cortex and Caudate Putamen of Rats Do Not Compromise Working Memory and Motor Activity. Neurotox Res 2023; 42:6. [PMID: 38133743 PMCID: PMC10746586 DOI: 10.1007/s12640-023-00684-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/10/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
Iron oxide nanoparticles (IONPs) have come into focus for their use in medical applications although possible health risks for humans, especially in terms of brain functions, have not yet been fully clarified. The present study investigates the effects of IONPs on neurobehavioural functions in rats. For this purpose, we infused dimercaptosuccinic acid-coated IONPs into the medial prefrontal cortex (mPFC) and caudate putamen (CPu). Saline (VEH) and ferric ammonium citrate (FAC) were administered as controls. One- and 4-week post-surgery mPFC-infused animals were tested for their working memory performance in the delayed alternation T-maze task and in the open field (OF) for motor activity, and CPu-infused rats were tested for their motor activity in the OF. After completion of the experiments, the brains were examined histologically and immunohistochemically. We did not observe any behavioural or structural abnormalities in the rats after administration of IONPs in the mPFC and the CPu. In contrast, administration of FAC into the CPu resulted in decreased motor activity and increased the number of microglia in the mPFC. Perls' Prussian blue staining revealed that FAC- and IONP-treated rats had more iron-containing ramified cells than VEH-treated rats, indicating iron uptake by microglia. Our results demonstrate that local infusions of IONPs into selected brain regions have no adverse impact on locomotor behaviour and working memory.
Collapse
Affiliation(s)
- Ellen Irrsack
- Department of Neuropharmacology, Centre for Cognitive Sciences, University of Bremen, PO Box 330440, Bremen, 28334, Germany.
| | - Sidar Aydin
- Department of Neuropharmacology, Centre for Cognitive Sciences, University of Bremen, PO Box 330440, Bremen, 28334, Germany
| | - Katja Bleckmann
- Department of Neuropharmacology, Centre for Cognitive Sciences, University of Bremen, PO Box 330440, Bremen, 28334, Germany
| | - Julia Schuller
- Department of Neuropharmacology, Centre for Cognitive Sciences, University of Bremen, PO Box 330440, Bremen, 28334, Germany
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen (CBIB), and Centre for Environmental Research and Sustainable, Technology, University of Bremen, PO Box 330440, Bremen, 28334, Germany
| | - Michael Koch
- Department of Neuropharmacology, Centre for Cognitive Sciences, University of Bremen, PO Box 330440, Bremen, 28334, Germany
| |
Collapse
|
4
|
An JR, Wang QF, Sun GY, Su JN, Liu JT, Zhang C, Wang L, Teng D, Yang YF, Shi Y. The Role of Iron Overload in Diabetic Cognitive Impairment: A Review. Diabetes Metab Syndr Obes 2023; 16:3235-3247. [PMID: 37872972 PMCID: PMC10590583 DOI: 10.2147/dmso.s432858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/29/2023] [Indexed: 10/25/2023] Open
Abstract
It is well documented that diabetes mellitus (DM) is strongly associated with cognitive decline and structural damage to the brain. Cognitive deficits appear early in DM and continue to worsen as the disease progresses, possibly due to different underlying mechanisms. Normal iron metabolism is necessary to maintain normal physiological functions of the brain, but iron deposition is one of the causes of some neurodegenerative diseases. Increasing evidence shows that iron overload not only increases the risk of DM, but also contributes to the development of cognitive impairment. The current review highlights the role of iron overload in diabetic cognitive impairment (DCI), including the specific location and regulation mechanism of iron deposition in the diabetic brain, the factors that trigger iron deposition, and the consequences of iron deposition. Finally, we also discuss possible therapies to improve DCI and brain iron deposition.
Collapse
Affiliation(s)
- Ji-Ren An
- Liaoning Key Laboratory of Chinese Medicine Combining Disease and Syndrome of Diabetes, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
- College of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, People’s Republic of China
| | - Qing-Feng Wang
- Liaoning Key Laboratory of Chinese Medicine Combining Disease and Syndrome of Diabetes, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Gui-Yan Sun
- Liaoning Key Laboratory of Chinese Medicine Combining Disease and Syndrome of Diabetes, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Jia-Nan Su
- Liaoning Key Laboratory of Chinese Medicine Combining Disease and Syndrome of Diabetes, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Jun-Tong Liu
- Liaoning Key Laboratory of Chinese Medicine Combining Disease and Syndrome of Diabetes, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Chi Zhang
- Liaoning Key Laboratory of Chinese Medicine Combining Disease and Syndrome of Diabetes, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Li Wang
- Liaoning Key Laboratory of Chinese Medicine Combining Disease and Syndrome of Diabetes, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Dan Teng
- He University, Shenyang, 110163, People’s Republic of China
| | - Yu-Feng Yang
- Liaoning Key Laboratory of Chinese Medicine Combining Disease and Syndrome of Diabetes, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Yan Shi
- Liaoning Key Laboratory of Chinese Medicine Combining Disease and Syndrome of Diabetes, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| |
Collapse
|
5
|
Distéfano-Gagné F, Bitarafan S, Lacroix S, Gosselin D. Roles and regulation of microglia activity in multiple sclerosis: insights from animal models. Nat Rev Neurosci 2023:10.1038/s41583-023-00709-6. [PMID: 37268822 DOI: 10.1038/s41583-023-00709-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/04/2023]
Abstract
As resident macrophages of the CNS, microglia are critical immune effectors of inflammatory lesions and associated neural dysfunctions. In multiple sclerosis (MS) and its animal models, chronic microglial inflammatory activity damages myelin and disrupts axonal and synaptic activity. In contrast to these detrimental effects, the potent phagocytic and tissue-remodelling capabilities of microglia support critical endogenous repair mechanisms. Although these opposing capabilities have long been appreciated, a precise understanding of their underlying molecular effectors is only beginning to emerge. Here, we review recent advances in our understanding of the roles of microglia in animal models of MS and demyelinating lesions and the mechanisms that underlie their damaging and repairing activities. We also discuss how the structured organization and regulation of the genome enables complex transcriptional heterogeneity within the microglial cell population at demyelinating lesions.
Collapse
Affiliation(s)
- Félix Distéfano-Gagné
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - Sara Bitarafan
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - Steve Lacroix
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - David Gosselin
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada.
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada.
| |
Collapse
|
6
|
Thompson C, Evans B, Zhao D, Purcell E. Spatiotemporal Expression of RNA-Seq Identified Proteins at the Electrode Interface. Acta Biomater 2023; 164:209-222. [PMID: 37116634 DOI: 10.1016/j.actbio.2023.04.028] [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: 12/20/2022] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/30/2023]
Abstract
Implantation of electrodes in the brain can be used to record from or stimulate neural tissues to treat neurological disease and injury. However, the tissue response to implanted devices can limit their functional longevity. Recent RNA-seq datasets identify hundreds of genes associated with gliosis, neuronal function, myelination, and cellular metabolism that are spatiotemporally expressed in neural tissues following the insertion of microelectrodes. To validate mRNA as a predictor of protein expression, this study evaluates a sub-set of RNA-seq identified proteins (RSIP) at 24-hours, 1-week, and 6-weeks post-implantation using quantitative immunofluorescence methods. This study found that expression of RSIPs associated with glial activation (Glial fibrillary acidic protein (GFAP), Polypyrmidine tract binding protein-1 (Ptbp1)), neuronal structure (Neurofilament heavy chain (Nefh), Proteolipid protein-1 (Plp1), Myelin Basic Protein (MBP)), and iron metabolism (Transferrin (TF), Ferritin heavy chain-1 (Fth1)) reinforce transcriptional data. This study also provides additional context to the cellular distribution of RSIPs using a MATLAB-based approach to quantify immunofluorescence intensity within specific cell types. Ptbp1, TF, and Fth1 were found to be spatiotemporally distributed within neurons, astrocytes, microglia, and oligodendrocytes at the device interface relative to distal and contralateral tissues. The altered distribution of RSIPs relative to distal tissue is largely localized within 100µm of the device injury, which approaches the functional recording range of implanted electrodes. This study provides evidence that RNA-sequencing can be used to predict protein-level changes in cortical tissues and that RSIPs can be further investigated to identify new biomarkers of the tissue response that influence signal quality. STATEMENT OF SIGNIFICANCE: : Microelectrode arrays implanted into the brain are useful tools that can be used to study neuroscience and to treat pathological conditions in a clinical setting. The tissue response to these devices, however, can severely limit their functional longevity. Transcriptomics has deepened the understandings of the tissue response by revealing numerous genes which are differentially expressed following device insertion. This manuscript provides validation for the use of transcriptomics to characterize the tissue response by evaluating a subset of known differentially expressed genes at the protein level around implanted electrodes over time. In additional to validating mRNA-to-protein relationships at the device interface, this study has identified emerging trends in the spatiotemporal distribution of proteins involved with glial activation, neuronal remodeling, and essential iron binding proteins around implanted silicon devices. This study additionally provides a new MATLAB based methodology to quantify protein distribution within discrete cell types at the device interface which may be used as biomarkers for further study or therapeutic intervention in the future.
Collapse
Affiliation(s)
- Cort Thompson
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Blake Evans
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Dorothy Zhao
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Erin Purcell
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America.
| |
Collapse
|
7
|
Tortuyaux R, Avila-Gutierrez K, Oudart M, Mazaré N, Mailly P, Deschemin JC, Vaulont S, Escartin C, Cohen-Salmon M. Physiopathological changes of ferritin mRNA density and distribution in hippocampal astrocytes in the mouse brain. J Neurochem 2022; 164:847-857. [PMID: 36562685 DOI: 10.1111/jnc.15747] [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: 07/18/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
Astrocytes are thought to play a crucial role in brain iron homeostasis. How they accomplish this regulation in vivo is unclear. In a recent transcriptomic analysis, we showed that polysomal Ftl1 and Fth1 mRNAs, encoding the ferritin light (Ftl) and heavy (Fth) chains that assemble into ferritin, a critical complex for iron storage and reduction, are enriched in perisynaptic astrocytic processes as compared to astrocytic soma. These data suggested that ferritin translation plays a specific role at the perisynaptic astrocytic interface and is tighly regulated by local translation. Here, we used our recently described AstroDot 3D in situ methodology to study the density and localization of ferritin mRNAs in astrocytes in the hippocampus in three different contexts in which local or systemic iron overload has been documented: aging, the hepcidin knock-out mouse model of hemochromatosis and the APP/PS1dE9 mouse model of Alzheimer's disease (AD). Our results showed that in wild type mice, Fth1 mRNA density was higher than Ftl1 and that both mRNAs were mostly distributed in astrocyte fine processes. Aging and absence of hepcidin caused an increased Fth1/Ftl1 ratio in astrocytes and in the case of aging, led to a redistribution of Fth1 mRNAs in astrocytic fine processes. In contrast, in AD mice, we observed a lower Fth1/Ftl1 ratio. Fth1 mRNAs became more somatic and Ftl1 mRNAs redistributed in large processes of astrocytes proximal to Amyloid beta (Aß) deposits. Hence, we propose that regulation of ferritin mRNA density and distribution in astrocytes contribute to iron homeostasis in physiology and pathophysiology.
Collapse
Affiliation(s)
- Romain Tortuyaux
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France.,Intensive Care Unit, CHU Lille, Lille, France
| | - Katia Avila-Gutierrez
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Marc Oudart
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Noémie Mazaré
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Philippe Mailly
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | | | - Sophie Vaulont
- CNRS, INSERM, Institut Cochin, Université Paris Cité, Paris, France
| | - Carole Escartin
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Martine Cohen-Salmon
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
| |
Collapse
|
8
|
Casella C, Chamberland M, Laguna PL, Parker GD, Rosser AE, Coulthard E, Rickards H, Berry SC, Jones DK, Metzler‐Baddeley C. Mutation-related magnetization-transfer, not axon density, drives white matter differences in premanifest Huntington disease: Evidence from in vivo ultra-strong gradient MRI. Hum Brain Mapp 2022; 43:3439-3460. [PMID: 35396899 PMCID: PMC9248323 DOI: 10.1002/hbm.25859] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/07/2022] [Accepted: 03/27/2022] [Indexed: 11/10/2022] Open
Abstract
White matter (WM) alterations have been observed in Huntington disease (HD) but their role in the disease-pathophysiology remains unknown. We assessed WM changes in premanifest HD by exploiting ultra-strong-gradient magnetic resonance imaging (MRI). This allowed to separately quantify magnetization transfer ratio (MTR) and hindered and restricted diffusion-weighted signal fractions, and assess how they drove WM microstructure differences between patients and controls. We used tractometry to investigate region-specific alterations across callosal segments with well-characterized early- and late-myelinating axon populations, while brain-wise differences were explored with tract-based cluster analysis (TBCA). Behavioral measures were included to explore disease-associated brain-function relationships. We detected lower MTR in patients' callosal rostrum (tractometry: p = .03; TBCA: p = .03), but higher MTR in their splenium (tractometry: p = .02). Importantly, patients' mutation-size and MTR were positively correlated (all p-values < .01), indicating that MTR alterations may directly result from the mutation. Further, MTR was higher in younger, but lower in older patients relative to controls (p = .003), suggesting that MTR increases are detrimental later in the disease. Finally, patients showed higher restricted diffusion signal fraction (FR) from the composite hindered and restricted model of diffusion (CHARMED) in the cortico-spinal tract (p = .03), which correlated positively with MTR in the posterior callosum (p = .033), potentially reflecting compensatory mechanisms. In summary, this first comprehensive, ultra-strong gradient MRI study in HD provides novel evidence of mutation-driven MTR alterations at the premanifest disease stage which may reflect neurodevelopmental changes in iron, myelin, or a combination of these.
Collapse
Affiliation(s)
- Chiara Casella
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of PsychologyCardiff UniversityCardiffUK
- Department of Perinatal Imaging and Health, School of Biomedical Engineering & Imaging SciencesKing's College London, St Thomas' HospitalLondonUK
| | - Maxime Chamberland
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of PsychologyCardiff UniversityCardiffUK
- Donders Institute for Brain, Cognition and BehaviorRadboud UniversityNijmegenThe Netherlands
| | - Pedro L. Laguna
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of PsychologyCardiff UniversityCardiffUK
| | - Greg D. Parker
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of PsychologyCardiff UniversityCardiffUK
| | - Anne E. Rosser
- Department of Neurology and Psychological MedicineHayden Ellis BuildingCardiffUK
- School of BiosciencesCardiff UniversityCardiffUK
| | | | - Hugh Rickards
- Birmingham and Solihull Mental Health NHS Foundation TrustBirminghamUK
- Institute of Clinical Sciences, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Samuel C. Berry
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of PsychologyCardiff UniversityCardiffUK
| | - Derek K. Jones
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of PsychologyCardiff UniversityCardiffUK
| | - Claudia Metzler‐Baddeley
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of PsychologyCardiff UniversityCardiffUK
| |
Collapse
|
9
|
Baldacchino K, Peveler WJ, Lemgruber L, Smith RS, Scharler C, Hayden L, Komarek L, Lindsay SL, Barnett SC, Edgar JM, Linington C, Thümmler K. Myelinated axons are the primary target of hemin-mediated oxidative damage in a model of the central nervous system. Exp Neurol 2022; 354:114113. [PMID: 35569511 DOI: 10.1016/j.expneurol.2022.114113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/29/2022] [Accepted: 05/08/2022] [Indexed: 12/01/2022]
Abstract
Iron released from oligodendrocytes during demyelination or derived from haemoglobin breakdown products is believed to amplify oxidative tissue injury in multiple sclerosis (MS). However, the pathophysiological significance of iron-containing haemoglobin breakdown products themselves is rarely considered in the context of MS and their cellular specificity and mode of action remain unclear. Using myelinating cell cultures, we now report the cytotoxic potential of hemin (ferriprotoporphyrin IX chloride), a major degradation product of haemoglobin, is 25-fold greater than equimolar concentrations of free iron in myelinating cultures; a model that reproduces the complex multicellular environment of the CNS. At low micro molar concentrations (3.3 - 10 μM) we observed hemin preferentially binds to myelin and axons to initiate a complex detrimental response that results in targeted demyelination and axonal loss but spares neuronal cell bodies, astrocytes and the majority of oligodendroglia. Demyelination and axonal loss in this context are executed by a combination of mechanisms that include iron-dependent peroxidation by reactive oxygen species (ROS) and ferroptosis. These effects are microglial-independent, do not require any initiating inflammatory insult and represent a direct effect that compromises the structural integrity of myelinated axons in the CNS. Our data identify hemin-mediated demyelination and axonal loss as a novel mechanism by which intracerebral degradation of haemoglobin may contribute to lesion development in MS.
Collapse
Affiliation(s)
- Karl Baldacchino
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - William J Peveler
- WestCHEM, School of Chemistry, University of Glasgow, Joseph Black Building, G12 8QQ Glasgow, UK
| | - Leandro Lemgruber
- Glasgow Imaging Facility, Institute of Infection, Immunity and Inflammation, University of Glasgow, University Avenue, Glasgow G12 8QQ, UK
| | - Rebecca Sherrard Smith
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Cornelia Scharler
- Institute of Experimental and Clinical Cell Therapy, Paracelsus Medical University, Salzburg, Austria
| | - Lorna Hayden
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Lina Komarek
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Susan L Lindsay
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Susan C Barnett
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Julia M Edgar
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Christopher Linington
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Katja Thümmler
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom.
| |
Collapse
|
10
|
Iqubal A, Iqubal MK, Sharma S, Wasim M, Alfaleh MA, Md S, Baboota S, Ali J, Haque SE. Pathogenic mechanisms and therapeutic promise of phytochemicals and nanocarriers based drug delivery against radiotherapy-induced neurotoxic manifestations. Drug Deliv 2022; 29:1492-1511. [PMID: 35543534 PMCID: PMC9103628 DOI: 10.1080/10717544.2022.2064562] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Radiotherapy is one of the extensively used therapeutic modalities in glioblastoma and other types of cancers. Radiotherapy is either used as a first-line approach or combined with pharmacotherapy or surgery to manage and treat cancer. Although the use of radiotherapy significantly increased the survival time of patients, but its use has been reported with marked neuroinflammation and cognitive dysfunction that eventually reduced the quality of life of patients. Based on the preclinical and clinical investigations, the profound role of increased oxidative stress, nuclear translocation of NF-kB, production of proinflammatory cytokines such as TNF-α, IL-6, IL-β, increased level of MMPs, increased apoptosis, reduced angiogenesis, neurogenesis, and histological aberrations in CA1, CA2, CA3 and DG region of the hippocampus have been reported. Various pharmacotherapeutic drugs are being used as an adjuvant to counteract this neurotoxic manifestation. Still, most of these drugs suffer from systemic adverse effect, causes interference to ongoing chemotherapy, and exhibit pharmacokinetic limitations in crossing the blood-brain barrier. Therefore, various phytoconstituents, their nano carrier-based drug delivery systems and miRNAs have been explored to overcome the aforementioned limitations. The present review is focused on the mechanism and evidence of radiotherapy-induced neuroinflammation and cognitive dysfunction, pathological and molecular changes in the brain homeostasis, available adjuvants, their limitations. Additionally, the potential role and mechanism of neuroprotection of various nanocarrier based natural products and miRNAs have been discussed.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.,Product Development Department, Sentiss Research Centre, Sentiss Pharma Pvt Ltd, Gurugram, India
| | - Sumit Sharma
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mohd Wasim
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mohamed A Alfaleh
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia.,Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia.,Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
11
|
Jang J, Kang J, Nam Y. [Brain Iron Imaging in Aging and Cognitive Disorders: MRI Approaches]. TAEHAN YONGSANG UIHAKHOE CHI 2022; 83:527-537. [PMID: 36238502 PMCID: PMC9514519 DOI: 10.3348/jksr.2022.0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/09/2022] [Accepted: 05/16/2022] [Indexed: 11/23/2022]
Abstract
Iron has a vital role in the human body, including the central nervous system. Increased deposition of iron in the brain has been reported in aging and important neurodegenerative diseases. Owing to the unique magnetic resonance properties of iron, MRI has great potential for in vivo assessment of iron deposition, distribution, and non-invasive quantification. In this paper, we will review the MRI methods for iron assessment and their changes in aging and neurodegenerative diseases, focusing on Alzheimer's disease. In addition, we will summarize the limitations of current approaches and introduce new areas and MRI methods for iron imaging that are expected in the future.
Collapse
|
12
|
Lee RX, Tang FR. Radiation-induced neuropathological changes in the oligodendrocyte lineage with relevant clinical manifestations and therapeutic strategies. Int J Radiat Biol 2022; 98:1519-1531. [PMID: 35311621 DOI: 10.1080/09553002.2022.2055804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE With technological advancements in radiation therapy for tumors of the central nervous system (CNS), high doses of ionizing radiation can be delivered to the tumors with improved accuracy. Despite the reduction of ionizing radiation-induced toxicity to surrounding tissues of the CNS, a wide array of side effects still occurs, particularly late-delayed changes. These alterations, such as white matter damages and neurocognitive impairments, are often debilitative and untreatable, significantly affecting the quality of life of these patients, especially children. Oligodendrocytes, a major class of glial cells, have been identified to be one of the targets of radiation toxicity and are recognized be involved in late-delayed radiation-induced neuropathological changes. These cells are responsible for forming the myelin sheaths that surround and insulate axons within the CNS. Here, the effects of ionizing radiation on the oligodendrocyte lineage as well as the common clinical manifestations resulting from radiation-induced damage to oligodendrocytes will be discussed. Potential prophylactic and therapeutic strategies against radiation-induced oligodendrocyte damage will also be considered. CONCLUSION Oligodendrocytes and oligodendrocyte progenitor cells (OPCs) are radiosensitive cells of the CNS. Here, general responses of these cells to radiation exposure have been outlined. However, several findings have not been consistent across various studies. For instance, cognitive decline in irradiated animals was observed to be accompanied by obvious demyelination or white matter changes in several studies but not in others. Hence, further studies have to be conducted to elucidate the level of contribution of the oligodendrocyte lineage to the development of late-delayed effects of radiation exposure, as well as to classify the dose and brain region-specific responses of the oligodendrocyte lineage to radiation. Several potential therapeutic approaches against late-delayed changes have been discussed, such as the transplantation of OPCs into irradiated regions and implementation of exercise. Many of these approaches show promising results. Further elucidation of the mechanisms involved in radiation-induced death of oligodendrocytes and OPCs would certainly aid in the development of novel protective and therapeutic strategies against the late-delayed effects of radiation.
Collapse
Affiliation(s)
- Rui Xue Lee
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore, Singapore
| | - Feng Ru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore, Singapore
| |
Collapse
|
13
|
Scalabrino G. Newly Identified Deficiencies in the Multiple Sclerosis Central Nervous System and Their Impact on the Remyelination Failure. Biomedicines 2022; 10:biomedicines10040815. [PMID: 35453565 PMCID: PMC9026986 DOI: 10.3390/biomedicines10040815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of multiple sclerosis (MS) remains enigmatic and controversial. Myelin sheaths in the central nervous system (CNS) insulate axons and allow saltatory nerve conduction. MS brings about the destruction of myelin sheaths and the myelin-producing oligodendrocytes (ODCs). The conundrum of remyelination failure is, therefore, crucial in MS. In this review, the roles of epidermal growth factor (EGF), normal prions, and cobalamin in CNS myelinogenesis are briefly summarized. Thereafter, some findings of other authors and ourselves on MS and MS-like models are recapitulated, because they have shown that: (a) EGF is significantly decreased in the CNS of living or deceased MS patients; (b) its repeated administration to mice in various MS-models prevents demyelination and inflammatory reaction; (c) as was the case for EGF, normal prion levels are decreased in the MS CNS, with a strong correspondence between liquid and tissue levels; and (d) MS cobalamin levels are increased in the cerebrospinal fluid, but decreased in the spinal cord. In fact, no remyelination can occur in MS if these molecules (essential for any form of CNS myelination) are lacking. Lastly, other non-immunological MS abnormalities are reviewed. Together, these results have led to a critical reassessment of MS pathogenesis, partly because EGF has little or no role in immunology.
Collapse
Affiliation(s)
- Giuseppe Scalabrino
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| |
Collapse
|
14
|
MR susceptibility imaging for detection of tumor-associated macrophages in glioblastoma. J Neurooncol 2022; 156:645-653. [PMID: 35043276 DOI: 10.1007/s11060-022-03947-3] [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: 10/29/2021] [Accepted: 01/04/2022] [Indexed: 10/19/2022]
Abstract
PURPOSE Tumor-associated macrophages (TAMs) are a key component of glioblastoma (GBM) microenvironment. Considering the differential role of different TAM phenotypes in iron metabolism with the M1 phenotype storing intracellular iron, and M2 phenotype releasing iron in the tumor microenvironment, we investigated MRI to quantify iron as an imaging biomarker for TAMs in GBM patients. METHODS 21 adult patients with GBM underwent a 3D single echo gradient echo MRI sequence and quantitative susceptibility maps were generated. In 3 subjects, ex vivo imaging of surgical specimens was performed on a 9.4 Tesla MRI using 3D multi-echo GRE scans, and R2* (1/T2*) maps were generated. Each specimen was stained with hematoxylin and eosin, as well as CD68, CD86, CD206, and L-Ferritin. RESULTS Significant positive correlation was observed between mean susceptibility for the tumor enhancing zone and the L-ferritin positivity percent (r = 0.56, p = 0.018) and the combination of tumor's enhancing zone and necrotic core and the L-Ferritin positivity percent (r = 0.72; p = 0.001). The mean susceptibility significantly correlated with positivity percent for CD68 (ρ = 0.52, p = 0.034) and CD86 (r = 0.7 p = 0.001), but not for CD206 (ρ = 0.09; p = 0.7). There was a positive correlation between mean R2* values and CD68 positive cell counts (r = 0.6, p = 0.016). Similarly, mean R2* values significantly correlated with CD86 (r = 0.54, p = 0.03) but not with CD206 (r = 0.15, p = 0.5). CONCLUSIONS This study demonstrated the potential of MR quantitative susceptibility mapping as a non-invasive method for in vivo TAM quantification and phenotyping. Validation of these findings with large multicenter studies is needed.
Collapse
|
15
|
Cheng R, Dhorajia VV, Kim J, Kim Y. Mitochondrial iron metabolism and neurodegenerative diseases. Neurotoxicology 2022; 88:88-101. [PMID: 34748789 PMCID: PMC8748425 DOI: 10.1016/j.neuro.2021.11.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 01/03/2023]
Abstract
Iron is a key element for mitochondrial function and homeostasis, which is also crucial for maintaining the neuronal system, but too much iron promotes oxidative stress. A large body of evidence has indicated that abnormal iron accumulation in the brain is associated with various neurodegenerative diseases such as Huntington's disease, Alzheimer's disease, Parkinson's disease, and Friedreich's ataxia. However, it is still unclear how irregular iron status contributes to the development of neuronal disorders. Hence, the current review provides an update on the causal effects of iron overload in the development and progression of neurodegenerative diseases and discusses important roles of mitochondrial iron homeostasis in these disease conditions. Furthermore, this review discusses potential therapeutic targets for the treatments of iron overload-linked neurodegenerative diseases.
Collapse
Affiliation(s)
- Ruiying Cheng
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, USA
| | | | - Jonghan Kim
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, USA.
| | - Yuho Kim
- Department of Physical Therapy and Kinesiology, University of Massachusetts Lowell, USA.
| |
Collapse
|
16
|
Morgan ML, Teo W, Hernandez Y, Brideau C, Cummins K, Kuipers HF, Stys PK. Cuprizone-induced Demyelination in Mouse Brain is not due to Depletion of Copper. ASN Neuro 2022; 14:17590914221126367. [PMID: 36114624 PMCID: PMC9483969 DOI: 10.1177/17590914221126367] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
The cuprizone (CPZ) model allows the study of the biochemical processes underlying
nonautoimmune-mediated demyelination, remyelination, and chronic white matter disease
progression. CPZ is a copper (Cu) chelator that chiefly causes oligodendrocyte apoptosis
in the corpus callosum and cerebellum when administered in the mouse diet. While
disruption of Cu homeostasis is known to cause neurodegeneration (as is observed in
Wilson’s and Menkes disease), no consensus exists to date as to CPZ’s mechanism of action.
We sought to determine whether CPZ-induced pathology is due to Cu depletion as is
generally believed. Cu supplementation in chow, in stoichiometric excess to the added CPZ,
did not reduce CPZ-induced demyelination in C57Bl/6 mice. Moreover, equivalent doses of
other known Cu chelators neocuproine and D-penicillamine (D-Pen) failed to induce central
nervous system (CNS) demyelination. Since administration of D-Pen in the treatment of
Wilson’s disease can induce hypocupremia, we next sought to recreate penicillamine-induced
Cu deficiency to compare with purported CPZ-induced Cu deficiency. The resulting clinical
phenotype and histopathology were unlike that of CPZ. D-Pen-treated mice exhibited digit
paralysis, tail flaccidity, subcutaneous hemorrhaging, and optic and sciatic neuropathy,
all of which were prevented with Cu supplementation. No demyelination of the corpus
callosum or cerebellum was observed, even with D-Pen doses tenfold higher than CPZ.
Intriguingly, addition of D-Pen to the CPZ diet paradoxically prevented demyelination in a
dose-dependent manner.
Collapse
Affiliation(s)
- Megan L. Morgan
- Cumming School of Medicine, Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, 3330 Hospital Drive N.W. HRIC 1B37A, Calgary, AB, T2N 4N1, Canada
| | - Wulin Teo
- Cumming School of Medicine, Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, 3330 Hospital Drive N.W. HRIC 1B37A, Calgary, AB, T2N 4N1, Canada
| | - Yda Hernandez
- Cumming School of Medicine, Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, 3330 Hospital Drive N.W. HRIC 1B37A, Calgary, AB, T2N 4N1, Canada
| | - Craig Brideau
- Cumming School of Medicine, Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, 3330 Hospital Drive N.W. HRIC 1B37A, Calgary, AB, T2N 4N1, Canada
| | - Karen Cummins
- Cumming School of Medicine, Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, 3330 Hospital Drive N.W. HRIC 1B37A, Calgary, AB, T2N 4N1, Canada
| | - Hedwich F. Kuipers
- Cumming School of Medicine, Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, 3330 Hospital Drive N.W. HRIC 1B37A, Calgary, AB, T2N 4N1, Canada
| | - Peter K. Stys
- Cumming School of Medicine, Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, 3330 Hospital Drive N.W. HRIC 1B37A, Calgary, AB, T2N 4N1, Canada
| |
Collapse
|
17
|
Riederer P, Monoranu C, Strobel S, Iordache T, Sian-Hülsmann J. Iron as the concert master in the pathogenic orchestra playing in sporadic Parkinson's disease. J Neural Transm (Vienna) 2021; 128:1577-1598. [PMID: 34636961 PMCID: PMC8507512 DOI: 10.1007/s00702-021-02414-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/29/2021] [Indexed: 02/07/2023]
Abstract
About 60 years ago, the discovery of a deficiency of dopamine in the nigro-striatal system led to a variety of symptomatic therapeutic strategies to supplement dopamine and to substantially improve the quality of life of patients with Parkinson's disease (PD). Since these seminal developments, neuropathological, neurochemical, molecular biological and genetic discoveries contributed to elucidate the pathology of PD. Oxidative stress, the consequences of reactive oxidative species, reduced antioxidative capacity including loss of glutathione, excitotoxicity, mitochondrial dysfunction, proteasomal dysfunction, apoptosis, lysosomal dysfunction, autophagy, suggested to be causal for ɑ-synuclein fibril formation and aggregation and contributing to neuroinflammation and neural cell death underlying this devastating disorder. However, there are no final conclusions about the triggered pathological mechanism(s) and the follow-up of pathological dysfunctions. Nevertheless, it is a fact, that iron, a major component of oxidative reactions, as well as neuromelanin, the major intraneuronal chelator of iron, undergo an age-dependent increase. And ageing is a major risk factor for PD. Iron is significantly increased in the substantia nigra pars compacta (SNpc) of PD. Reasons for this finding include disturbances in iron-related import and export mechanisms across the blood-brain barrier (BBB), localized opening of the BBB at the nigro-striatal tract including brain vessel pathology. Whether this pathology is of primary or secondary importance is not known. We assume that there is a better fit to the top-down hypotheses and pathogens entering the brain via the olfactory system, then to the bottom-up (gut-brain) hypothesis of PD pathology. Triggers for the bottom-up, the dual-hit and the top-down pathologies include chemicals, viruses and bacteria. If so, hepcidin, a regulator of iron absorption and its distribution into tissues, is suggested to play a major role in the pathogenesis of iron dyshomeostasis and risk for initiating and progressing ɑ-synuclein pathology. The role of glial components to the pathology of PD is still unknown. However, the dramatic loss of glutathione (GSH), which is mainly synthesized in glia, suggests dysfunction of this process, or GSH uptake into neurons. Loss of GSH and increase in SNpc iron concentration have been suggested to be early, may be even pre-symptomatic processes in the pathology of PD, despite the fact that they are progression factors. The role of glial ferritin isoforms has not been studied so far in detail in human post-mortem brain tissue and a close insight into their role in PD is called upon. In conclusion, "iron" is a major player in the pathology of PD. Selective chelation of excess iron at the site of the substantia nigra, where a dysfunction of the BBB is suggested, with peripherally acting iron chelators is suggested to contribute to the portfolio and therapeutic armamentarium of anti-Parkinson medications.
Collapse
Affiliation(s)
- P Riederer
- Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy, University Hospital Wuerzburg, University of Wuerzburg, Wuerzburg, Germany. .,Department of Psychiatry, University of Southern Denmark, Odense, Denmark.
| | - C Monoranu
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, Wuerzburg, Germany
| | - S Strobel
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, Wuerzburg, Germany
| | - T Iordache
- George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, Târgu Mureș, Romania
| | - J Sian-Hülsmann
- Department of Medical Physiology, University of Nairobi, P.O. Box 30197, Nairobi, 00100, Kenya
| |
Collapse
|
18
|
Lloret A, Esteve D, Lloret MA, Monllor P, López B, León JL, Cervera-Ferri A. Is Oxidative Stress the Link Between Cerebral Small Vessel Disease, Sleep Disruption, and Oligodendrocyte Dysfunction in the Onset of Alzheimer's Disease? Front Physiol 2021; 12:708061. [PMID: 34512381 PMCID: PMC8424010 DOI: 10.3389/fphys.2021.708061] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/28/2021] [Indexed: 01/07/2023] Open
Abstract
Oxidative stress is an early occurrence in the development of Alzheimer’s disease (AD) and one of its proposed etiologic hypotheses. There is sufficient experimental evidence supporting the theory that impaired antioxidant enzymatic activity and increased formation of reactive oxygen species (ROS) take place in this disease. However, the antioxidant treatments fail to stop its advancement. Its multifactorial condition and the diverse toxicological cascades that can be initiated by ROS could possibly explain this failure. Recently, it has been suggested that cerebral small vessel disease (CSVD) contributes to the onset of AD. Oxidative stress is a central hallmark of CSVD and is depicted as an early causative factor. Moreover, data from various epidemiological and clinicopathological studies have indicated a relationship between CSVD and AD where endothelial cells are a source of oxidative stress. These cells are also closely related to oligodendrocytes, which are, in particular, sensitive to oxidation and lead to myelination being compromised. The sleep/wake cycle is another important control in the proliferation, migration, and differentiation of oligodendrocytes, and sleep loss reduces myelin thickness. Moreover, sleep plays a crucial role in resistance against CSVD, and poor sleep quality increases the silent markers of this vascular disease. Sleep disruption is another early occurrence in AD and is related to an increase in oxidative stress. In this study, the relationship between CSVD, oligodendrocyte dysfunction, and sleep disorders is discussed while focusing on oxidative stress as a common occurrence and its possible role in the onset of AD.
Collapse
Affiliation(s)
- Ana Lloret
- INCLIVA, CIBERFES, Department of Physiology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Daniel Esteve
- INCLIVA, CIBERFES, Department of Physiology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Maria Angeles Lloret
- Department of Clinical Neurophysiology, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Paloma Monllor
- INCLIVA, CIBERFES, Department of Physiology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Begoña López
- Department of Neurology, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - José Luis León
- Departament of Neuroradiology, Ascires Biomedical Group, Hospital Clinico Universitario, Valencia, Spain
| | - Ana Cervera-Ferri
- Department of Anatomy and Human Embryology, University of Valencia, Valencia, Spain
| |
Collapse
|
19
|
Jung SY, Kang JW. Is it really a seizure? The challenge of paroxysmal nonepileptic events in young infants. Clin Exp Pediatr 2021; 64:384-392. [PMID: 32972054 PMCID: PMC8342880 DOI: 10.3345/cep.2020.00451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 09/03/2020] [Indexed: 12/27/2022] Open
Abstract
Paroxysmal nonepileptic events (PNE) comprise of a variety of nonepileptic behaviors and are divided into various types. A more accurate diagnosis is possible by examining the video clip provided by the caregiver. In infants, physiologic PNE accounts for the majority of the PNE. It is important to exclude epilepsy, for which blood tests, electroencephalography, and imaging tests can facilitate differential diagnosis. Since most PNE have a benign progress, symptoms often improve with age and without special treatment. Therefore, it is important to reassure the caregivers after making an accurate diagnosis.
Collapse
Affiliation(s)
- Seung Yeon Jung
- Department of Pediatrics, Chungnam National University Hospital, Daejeon, Korea
| | - Joon Won Kang
- Department of Pediatrics & Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| |
Collapse
|
20
|
Gozt A, Hellewell S, Ward PGD, Bynevelt M, Fitzgerald M. Emerging Applications for Quantitative Susceptibility Mapping in the Detection of Traumatic Brain Injury Pathology. Neuroscience 2021; 467:218-236. [PMID: 34087394 DOI: 10.1016/j.neuroscience.2021.05.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/16/2022]
Abstract
Traumatic brain injury (TBI) is a common but heterogeneous injury underpinned by numerous complex and interrelated pathophysiological mechanisms. An essential trace element, iron is abundant within the brain and involved in many fundamental neurobiological processes, including oxygen transportation, oxidative phosphorylation, myelin production and maintenance, as well as neurotransmitter synthesis and metabolism. Excessive levels of iron are neurotoxic and thus iron homeostasis is tightly regulated in the brain, however, many details about the mechanisms by which this is achieved are yet to be elucidated. A key mediator of oxidative stress, mitochondrial dysfunction and neuroinflammatory response, iron dysregulation is an important contributor to secondary injury in TBI. Advances in neuroimaging that leverage magnetic susceptibility properties have enabled increasingly comprehensive investigations into the distribution and behaviour of iron in the brain amongst healthy individuals as well as disease states such as TBI. Quantitative Susceptibility Mapping (QSM) is an advanced neuroimaging technique that promises quantitative estimation of local magnetic susceptibility at the voxel level. In this review, we provide an overview of brain iron and its homeostasis, describe recent advances enabling applications of QSM within the context of TBI and summarise the current state of the literature. Although limited, the emergent research suggests that QSM is a promising neuroimaging technique that can be used to investigate a host of pathophysiological changes that are associated with TBI.
Collapse
Affiliation(s)
- Aleksandra Gozt
- Curtin University, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Bentley, WA Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA Australia
| | - Sarah Hellewell
- Curtin University, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Bentley, WA Australia
| | - Phillip G D Ward
- Australian Research Council Centre of Excellence for Integrative Brain Function, VIC Australia; Turner Institute for Brain and Mental Health, Monash University, VIC Australia
| | - Michael Bynevelt
- Neurological Intervention and Imaging Service of Western Australia, Sir Charles Gairdner Hospital, Nedlands, WA Australia
| | - Melinda Fitzgerald
- Curtin University, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Bentley, WA Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA Australia.
| |
Collapse
|
21
|
Greenwood EK, Brown DR. Senescent Microglia: The Key to the Ageing Brain? Int J Mol Sci 2021; 22:4402. [PMID: 33922383 PMCID: PMC8122783 DOI: 10.3390/ijms22094402] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
Ageing represents the single biggest risk factor for development of neurodegenerative disease. Despite being such long-lived cells, microglia have been relatively understudied for their role in the ageing process. Reliably identifying aged microglia has proven challenging, not least due to the diversity of cell populations, and the limitations of available models, further complicated by differences between human and rodent cells. Consequently, the literature contains multiple descriptions and categorisations of microglia with neurotoxic phenotypes, including senescence, without any unifying markers. The role of microglia in brain homeostasis, particularly iron storage and metabolism, may provide a key to reliable identification.
Collapse
Affiliation(s)
| | - David R. Brown
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK;
| |
Collapse
|
22
|
The Role of Butyrylcholinesterase and Iron in the Regulation of Cholinergic Network and Cognitive Dysfunction in Alzheimer's Disease Pathogenesis. Int J Mol Sci 2021; 22:ijms22042033. [PMID: 33670778 PMCID: PMC7922581 DOI: 10.3390/ijms22042033] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD), the most common form of dementia in elderly individuals, is marked by progressive neuron loss. Despite more than 100 years of research on AD, there is still no treatment to cure or prevent the disease. High levels of amyloid-β (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain are neuropathological hallmarks of AD. However, based on postmortem analyses, up to 44% of individuals have been shown to have high Aβ deposits with no clinical signs, due to having a “cognitive reserve”. The biochemical mechanism explaining the prevention of cognitive impairment in the presence of Aβ plaques is still unknown. It seems that in addition to protein aggregation, neuroinflammatory changes associated with aging are present in AD brains that are correlated with a higher level of brain iron and oxidative stress. It has been shown that iron accumulates around amyloid plaques in AD mouse models and postmortem brain tissues of AD patients. Iron is required for essential brain functions, including oxidative metabolism, myelination, and neurotransmitter synthesis. However, an imbalance in brain iron homeostasis caused by aging underlies many neurodegenerative diseases. It has been proposed that high iron levels trigger an avalanche of events that push the progress of the disease, accelerating cognitive decline. Patients with increased amyloid plaques and iron are highly likely to develop dementia. Our observations indicate that the butyrylcholinesterase (BChE) level seems to be iron-dependent, and reports show that BChE produced by reactive astrocytes can make cognitive functions worse by accelerating the decay of acetylcholine in aging brains. Why, even when there is a genetic risk, do symptoms of the disease appear after many years? Here, we discuss the relationship between genetic factors, age-dependent iron tissue accumulation, and inflammation, focusing on AD.
Collapse
|
23
|
LeVine SM, Zhu H, Tague SE. A Simplified Method for the Histochemical Detection of Iron in Paraffin Sections: Intracellular Iron Deposits in Central Nervous System Tissue. ASN Neuro 2021; 13:1759091420982169. [PMID: 33430620 PMCID: PMC7809306 DOI: 10.1177/1759091420982169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Although all cells contain iron, most histochemical methods fail to reveal the presence of iron within many cells of the central nervous system (CNS), particularly neurons. Previously, a sensitive method was developed that limited the extraction of iron in paraffin sections, and this method revealed staining within neurons. However, the staining was often too robust making it difficult to discern discrete intracellular structures. In 1970, a study incorporated acetone in an iron histochemical procedure to facilitate the demarcation of staining features. In the present study, both acetone and limits to iron extraction were included in a simplified staining procedure. This procedure was applied to paraffin sections of CNS tissue from CISD2 deficient and littermate control mice. Discrete nuclear and cytoplasmic staining features were detected in all mice. Although widely present in neurons, punctate cytoplasmic staining was particularly prominent in large neurons within the hindbrain. Evaluation of extended depth of focus images, from serial focal planes, revealed numerous stained cytoplasmic structures. Additionally, the simplified staining procedure was applied to paraffin sections from Alzheimer’s disease and control cases. Despite suboptimal processing conditions compared to mouse tissue, discrete staining of cytoplasmic structures was revealed in some neurons, although many other neurons had nondescript staining features. In addition, initial findings revealed iron deposited within some vessels from patients with Alzheimer’s disease. In summary, since paraffin sections are commonly used for histological preparations, this simplified histochemical procedure could facilitate the study of iron in various CNS conditions by revealing staining details often missed by other procedures.
Collapse
Affiliation(s)
- Steven M LeVine
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Hao Zhu
- Department of Clinical Laboratory Sciences, University of Kansas Medical Center, Kansas City, Kansas, United States.,Neuroscience Graduate Program, University of Kansas Medical Center, Kansas City, Kansas, United States.,Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Sarah E Tague
- Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, Kansas, United States
| |
Collapse
|
24
|
Kim SW, Kim Y, Kim SE, An JY. Ferroptosis-Related Genes in Neurodevelopment and Central Nervous System. BIOLOGY 2021; 10:35. [PMID: 33419148 PMCID: PMC7825574 DOI: 10.3390/biology10010035] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/01/2021] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
Ferroptosis, first introduced as a new form of regulated cell death induced by erastin, is accompanied by the accumulation of iron and lipid peroxides, thus it can be inhibited either by iron chelators or by lipophilic antioxidants. In the past decade, multiple studies have introduced the potential importance of ferroptosis in many human diseases, including cancer and neurodegenerative diseases. In this review, we will discuss the genetic association of ferroptosis with neurological disorders and development of the central nervous system.
Collapse
Affiliation(s)
- Soo-Whee Kim
- Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (S.-W.K.); (Y.K.)
- Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Yujin Kim
- Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (S.-W.K.); (Y.K.)
- Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Sung Eun Kim
- Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (S.-W.K.); (Y.K.)
- Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Joon-Yong An
- Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (S.-W.K.); (Y.K.)
- Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| |
Collapse
|
25
|
Radioprotective Effect of Flavonoids on Ionizing Radiation-Induced Brain Damage. Molecules 2020; 25:molecules25235719. [PMID: 33287417 PMCID: PMC7730479 DOI: 10.3390/molecules25235719] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/25/2020] [Accepted: 12/02/2020] [Indexed: 01/27/2023] Open
Abstract
Patients receiving brain radiotherapy may suffer acute or chronic side effects. Ionizing radiation induces the production of intracellular reactive oxygen species and pro-inflammatory cytokines in the central nervous system, leading to brain damage. Complementary Chinese herbal medicine therapy may reduce radiotherapy-induced side effects. Flavonoids are a class of natural products which can be extracted from Chinese herbal medicine and have been shown to have neuroprotective and radioprotective properties. Flavonoids are effective antioxidants and can also inhibit regulatory enzymes or transcription factors important for controlling inflammatory mediators, affect oxidative stress through interaction with DNA and enhance genomic stability. In this paper, radiation-induced brain damage and the relevant molecular mechanism were summarized. The radio-neuro-protective effect of flavonoids, i.e., antioxidant, anti-inflammatory and maintaining genomic stability, were then reviewed. We concluded that flavonoids treatment may be a promising complementary therapy to prevent radiotherapy-induced brain pathophysiological changes and cognitive impairment.
Collapse
|
26
|
Dales JP, Desplat-Jégo S. Metal Imbalance in Neurodegenerative Diseases with a Specific Concern to the Brain of Multiple Sclerosis Patients. Int J Mol Sci 2020; 21:E9105. [PMID: 33266021 PMCID: PMC7730295 DOI: 10.3390/ijms21239105] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/29/2020] [Accepted: 11/19/2020] [Indexed: 12/16/2022] Open
Abstract
There is increasing evidence that deregulation of metals contributes to a vast range of neurodegenerative diseases including multiple sclerosis (MS). MS is a chronic inflammatory disease of the central nervous system (CNS) manifesting disability and neurological symptoms. The precise origin of MS is unknown, but the disease is characterized by focal inflammatory lesions in the CNS associated with an autoimmune reaction against myelin. The treatment of this disease has mainly been based on the prescription of immunosuppressive and immune-modulating agents. However, the rate of progressive disability and early mortality is still worrisome. Metals may represent new diagnostic and predictive markers of severity and disability as well as innovative candidate drug targets for future therapies. In this review, we describe the recent advances in our understanding on the role of metals in brain disorders of neurodegenerative diseases and MS patients.
Collapse
Affiliation(s)
- Jean-Philippe Dales
- Institute of Neurophysiopathology, CNRS, INP, Aix-Marseille University, 13005 Marseille, France;
- Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Pavillon Etoile, Pôle de Biologie, Service d’anatomie-pathologie, CEDEX 20, 13915 Marseille, France
| | - Sophie Desplat-Jégo
- Institute of Neurophysiopathology, CNRS, INP, Aix-Marseille University, 13005 Marseille, France;
- Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, Pôle de Biologie, Service d’Immunologie, 13005 Marseille, France
| |
Collapse
|
27
|
Cheli VT, Correale J, Paez PM, Pasquini JM. Iron Metabolism in Oligodendrocytes and Astrocytes, Implications for Myelination and Remyelination. ASN Neuro 2020; 12:1759091420962681. [PMID: 32993319 PMCID: PMC7545512 DOI: 10.1177/1759091420962681] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Iron is a key nutrient for normal central nervous system (CNS) development and function; thus, iron deficiency as well as iron excess may result in harmful effects in the CNS. Oligodendrocytes and astrocytes are crucial players in brain iron equilibrium. However, the mechanisms of iron uptake, storage, and efflux in oligodendrocytes and astrocytes during CNS development or under pathological situations such as demyelination are not completely understood. In the CNS, iron is directly required for myelin production as a cofactor for enzymes involved in ATP, cholesterol and lipid synthesis, and oligodendrocytes are the cells with the highest iron levels in the brain which is linked to their elevated metabolic needs associated with the process of myelination. Unlike oligodendrocytes, astrocytes do not have a high metabolic requirement for iron. However, these cells are in close contact with blood vessel and have a strong iron transport capacity. In several pathological situations, changes in iron homoeostasis result in altered cellular iron distribution and accumulation and oxidative stress. In inflammatory demyelinating diseases such as multiple sclerosis, reactive astrocytes accumulate iron and upregulate iron efflux and influx molecules, which suggest that they are outfitted to take up and safely recycle iron. In this review, we will discuss the participation of oligodendrocytes and astrocytes in CNS iron homeostasis. Understanding the molecular mechanisms of iron uptake, storage, and efflux in oligodendrocytes and astrocytes is necessary for planning effective strategies for iron management during CNS development as well as for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Veronica T Cheli
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, Hunter James Kelly Research Institute, The State University of New York, University at Buffalo, Buffalo, New York, United States
| | | | - Pablo M Paez
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, Hunter James Kelly Research Institute, The State University of New York, University at Buffalo, Buffalo, New York, United States
| | - Juana M Pasquini
- CONICET, Instituto de Química y Fisicoquímica Biológicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
28
|
Casella C, Lipp I, Rosser A, Jones DK, Metzler‐Baddeley C. A Critical Review of White Matter Changes in Huntington's Disease. Mov Disord 2020; 35:1302-1311. [PMID: 32537844 PMCID: PMC9393936 DOI: 10.1002/mds.28109] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/07/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022] Open
Abstract
Huntington's disease is a genetic neurodegenerative disorder. White matter alterations have recently been identified as a relevant pathophysiological feature of Huntington's disease, but their etiology and role in disease pathogenesis and progression remain unclear. Increasing evidence suggests that white matter changes in this disorder are attributed to alterations in myelin-associated biological processes. This review first discusses evidence from neurochemical studies lending support to the demyelination hypothesis of Huntington's disease, demonstrating aberrant myelination and changes in oligodendrocytes in the Huntington's brain. Next, evidence from neuroimaging studies is reviewed, the limitations of the described methodologies are discussed, and suggested interpretations of findings from published studies are challenged. Although our understanding of Huntington's associated pathological changes in the brain will increasingly rely on neuroimaging techniques, the shortcomings of these methodologies must not be forgotten. Advances in magnetic resonance imaging techniques and tissue modeling will enable a better in vivo, longitudinal characterization of the biological properties of white matter microstructure. This in turn will facilitate identification of disease-related biomarkers and the specification of outcome measures in clinical trials. © 2020 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Chiara Casella
- Cardiff University Brain Research Imaging CentreSchool of Psychology, Cardiff UniversityCardiffUnited Kingdom
| | - Ilona Lipp
- Department of NeurophysicsMax Planck Institute for Human Cognitive and Brain SciencesLeipzigGermany
| | - Anne Rosser
- School of BiosciencesCardiff UniversityCardiffUnited Kingdom
| | - Derek K Jones
- Cardiff University Brain Research Imaging CentreSchool of Psychology, Cardiff UniversityCardiffUnited Kingdom
- Mary MacKillop Institute for Health ResearchAustralian Catholic UniversityMelbourneVictoriaAustralia
| | - Claudia Metzler‐Baddeley
- Cardiff University Brain Research Imaging CentreSchool of Psychology, Cardiff UniversityCardiffUnited Kingdom
| |
Collapse
|
29
|
Lee NJ, Ha SK, Sati P, Absinta M, Nair G, Luciano NJ, Leibovitch EC, Yen CC, Rouault TA, Silva AC, Jacobson S, Reich DS. Potential role of iron in repair of inflammatory demyelinating lesions. J Clin Invest 2020; 129:4365-4376. [PMID: 31498148 DOI: 10.1172/jci126809] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 07/16/2019] [Indexed: 12/20/2022] Open
Abstract
Inflammatory destruction of iron-rich myelin is characteristic of multiple sclerosis (MS). Although iron is needed for oligodendrocytes to produce myelin during development, its deposition has also been linked to neurodegeneration and inflammation, including in MS. We report perivascular iron deposition in multiple sclerosis lesions that was mirrored in 72 lesions from 13 marmosets with experimental autoimmune encephalomyelitis. Iron accumulated mainly inside microglia/macrophages from 6 weeks after demyelination. Consistently, expression of transferrin receptor, the brain's main iron-influx protein, increased as lesions aged. Iron was uncorrelated with inflammation and postdated initial demyelination, suggesting that iron is not directly pathogenic. Iron homeostasis was at least partially restored in remyelinated, but not persistently demyelinated, lesions. Taken together, our results suggest that iron accumulation in the weeks after inflammatory demyelination may contribute to lesion repair rather than inflammatory demyelination per se.
Collapse
Affiliation(s)
- Nathanael J Lee
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA.,Department of Neuroscience, Georgetown University Medical Center, Georgetown University, Washington, District of Columbia, USA
| | - Seung-Kwon Ha
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Pascal Sati
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Martina Absinta
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Govind Nair
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Nicholas J Luciano
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Emily C Leibovitch
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Cecil C Yen
- Cerebral Microcirculation Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Tracey A Rouault
- Section on Human Iron Metabolism, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Afonso C Silva
- Cerebral Microcirculation Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Steven Jacobson
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
30
|
Rodriguez-Callejas JD, Fuchs E, Perez-Cruz C. Increased oxidative stress, hyperphosphorylation of tau, and dystrophic microglia in the hippocampus of aged Tupaia belangeri. Glia 2020; 68:1775-1793. [PMID: 32096580 DOI: 10.1002/glia.23804] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/04/2020] [Accepted: 02/07/2020] [Indexed: 12/26/2022]
Abstract
Aging is a major risk factor for the development of neurodegenerative diseases. Alzheimer's disease and other neurodegenerative diseases are characterized by abnormal and prominent protein aggregation in the brain, partially due to deficiency in protein clearance. It has been proposed that alterations in microglia phagocytosis and debris clearance hasten the onset of neurodegeneration. Dystrophic microglia are abundant in aged humans, and it has been associated with the onset of disease. Furthermore, alterations in microglia containing ferritin are associated with neurodegenerative conditions. To further understand the process of microglia dysfunction during the aging process, we used hippocampal sections from Tupaia belangeri (tree shrews). Adult (mean age 3.8 years), old (mean age 6 years), and aged (mean age 7.5 years) tree shrews were used for histochemical and immunostaining techniques to determine ferritin and Iba1 positive microglia, iron tissue content, tau hyperphosphorylation and oxidized-RNA in dentate gyrus, subiculum, and CA1-CA3 hippocampal regions. Our results indicated that aged tree shrews presented an increased number of activated microglia containing ferritin, but microglia labeled with Iba1 with a dystrophic phenotype was more abundant in aged individuals. With aging, oxidative damage to RNA (8OHG) increased significantly in all hippocampal regions, while tau hyperphosphorylation (AT100) was enhanced in DG, CA3, and SUB in aged animals. Phagocytic inclusions of 8OHG- and AT100-damaged cells were observed in activated M2 microglia in old and aged animals. These data indicate that aged tree shrew may be a suitable model for translational research to study brain and microglia alterations during the aging process.
Collapse
Affiliation(s)
| | - Eberhard Fuchs
- German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | | |
Collapse
|
31
|
Gothié J, Vancamp P, Demeneix B, Remaud S. Thyroid hormone regulation of neural stem cell fate: From development to ageing. Acta Physiol (Oxf) 2020; 228:e13316. [PMID: 31121082 PMCID: PMC9286394 DOI: 10.1111/apha.13316] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/10/2019] [Accepted: 05/17/2019] [Indexed: 12/13/2022]
Abstract
In the vertebrate brain, neural stem cells (NSCs) generate both neuronal and glial cells throughout life. However, their neuro‐ and gliogenic capacity changes as a function of the developmental context. Despite the growing body of evidence on the variety of intrinsic and extrinsic factors regulating NSC physiology, their precise cellular and molecular actions are not fully determined. Our review focuses on thyroid hormone (TH), a vital component for both development and adult brain function that regulates NSC biology at all stages. First, we review comparative data to analyse how TH modulates neuro‐ and gliogenesis during vertebrate brain development. Second, as the mammalian brain is the most studied, we highlight the molecular mechanisms underlying TH action in this context. Lastly, we explore how the interplay between TH signalling and cell metabolism governs both neurodevelopmental and adult neurogenesis. We conclude that, together, TH and cellular metabolism regulate optimal brain formation, maturation and function from early foetal life to adult in vertebrate species.
Collapse
Affiliation(s)
- Jean‐David Gothié
- Department of Neurology & Neurosurgery Montreal Neurological Institute & Hospital, McGill University Montreal Quebec Canada
| | - Pieter Vancamp
- CNRS UMR 7221 Muséum National d’Histoire Naturelle Paris France
| | | | - Sylvie Remaud
- CNRS UMR 7221 Muséum National d’Histoire Naturelle Paris France
| |
Collapse
|
32
|
Age, But Not Repeated Exposure to Gadoterate Meglumine, Is Associated With T1- and T2-Weighted Signal Intensity Changes in the Deep Brain Nuclei of Pediatric Patients. Invest Radiol 2019; 54:537-548. [DOI: 10.1097/rli.0000000000000564] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
33
|
Monzani E, Nicolis S, Dell'Acqua S, Capucciati A, Bacchella C, Zucca FA, Mosharov EV, Sulzer D, Zecca L, Casella L. Dopamin, oxidativer Stress und Protein‐Chinonmodifikationen bei Parkinson und anderen neurodegenerativen Erkrankungen. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201811122] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Enrico Monzani
- Department of ChemistryUniversity of Pavia 27100 Pavia Italien
| | | | | | | | | | - Fabio A. Zucca
- Institute of Biomedical TechnologiesNational Research Council of Italy Segrate (Mailand) Italien
| | - Eugene V. Mosharov
- Department of PsychiatryColumbia University Medical CenterNew York State Psychiatric Institute New York NY USA
- Departments Neurology, PharmacologyColumbia University Medical Center New York NY USA
| | - David Sulzer
- Department of PsychiatryColumbia University Medical CenterNew York State Psychiatric Institute New York NY USA
- Departments Neurology, PharmacologyColumbia University Medical Center New York NY USA
| | - Luigi Zecca
- Institute of Biomedical TechnologiesNational Research Council of Italy Segrate (Mailand) Italien
- Department of PsychiatryColumbia University Medical CenterNew York State Psychiatric Institute New York NY USA
| | - Luigi Casella
- Department of ChemistryUniversity of Pavia 27100 Pavia Italien
| |
Collapse
|
34
|
Monzani E, Nicolis S, Dell'Acqua S, Capucciati A, Bacchella C, Zucca FA, Mosharov EV, Sulzer D, Zecca L, Casella L. Dopamine, Oxidative Stress and Protein-Quinone Modifications in Parkinson's and Other Neurodegenerative Diseases. Angew Chem Int Ed Engl 2019; 58:6512-6527. [PMID: 30536578 DOI: 10.1002/anie.201811122] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/10/2018] [Indexed: 12/19/2022]
Abstract
Dopamine (DA) is the most important catecholamine in the brain, as it is the most abundant and the precursor of other neurotransmitters. Degeneration of nigrostriatal neurons of substantia nigra pars compacta in Parkinson's disease represents the best-studied link between DA neurotransmission and neuropathology. Catecholamines are reactive molecules that are handled through complex control and transport systems. Under normal conditions, small amounts of cytosolic DA are converted to neuromelanin in a stepwise process involving melanization of peptides and proteins. However, excessive cytosolic or extraneuronal DA can give rise to nonselective protein modifications. These reactions involve DA oxidation to quinone species and depend on the presence of redox-active transition metal ions such as iron and copper. Other oxidized DA metabolites likely participate in post-translational protein modification. Thus, protein-quinone modification is a heterogeneous process involving multiple DA-derived residues that produce structural and conformational changes of proteins and can lead to aggregation and inactivation of the modified proteins.
Collapse
Affiliation(s)
- Enrico Monzani
- Department of Chemistry, University of Pavia, 27100, Pavia, Italy
| | - Stefania Nicolis
- Department of Chemistry, University of Pavia, 27100, Pavia, Italy
| | | | | | - Chiara Bacchella
- Department of Chemistry, University of Pavia, 27100, Pavia, Italy
| | - Fabio A Zucca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate (Milano), Italy
| | - Eugene V Mosharov
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - David Sulzer
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA.,Departments of Neurology and Pharmacology, Columbia University Medical Center, New York, NY, USA
| | - Luigi Zecca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate (Milano), Italy.,Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Luigi Casella
- Department of Chemistry, University of Pavia, 27100, Pavia, Italy
| |
Collapse
|
35
|
Liu C, Liang MC, Soong TW. Nitric Oxide, Iron and Neurodegeneration. Front Neurosci 2019; 13:114. [PMID: 30833886 PMCID: PMC6388708 DOI: 10.3389/fnins.2019.00114] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/30/2019] [Indexed: 12/25/2022] Open
Abstract
Iron is a crucial cofactor for several physiological functions in the brain including transport of oxygen, DNA synthesis, mitochondrial respiration, synthesis of myelin, and neurotransmitter metabolism. If iron concentration exceeds the capacity of cellular sequestration, excessive labile iron will be harmful by generating oxidative stress that leads to cell death. In patients suffering from Parkinson disease, the total amount of iron in the substantia nigra was reported to increase with disease severity. High concentrations of iron were also found in the amyloid plaques and neurofibrillary tangles of human Alzheimer disease brains. Besides iron, nitric oxide (NO) produced in high concentration has been associated with neurodegeneration. NO is produced as a co-product when the enzyme NO synthase converts L-arginine to citrulline, and NO has a role to support normal physiological functions. When NO is produced in a high concentration under pathological conditions such as inflammation, aberrantly S-nitrosylated proteins can initiate neurodegeneration. Interestingly, NO is closely related with iron homeostasis. Firstly, it regulates iron-related gene expression through a system involving iron regulatory protein and its cognate iron responsive element (IRP-IRE). Secondly, it modified the function of iron-related protein directly via S-nitrosylation. In this review, we examine the recent advances about the potential role of dysregulated iron homeostasis in neurodegeneration, with an emphasis on AD and PD, and we discuss iron chelation as a potential therapy. This review also highlights the changes in iron homeostasis caused by NO. An understanding of these mechanisms will help us formulate strategies to reverse or ameliorate iron-related neurodegeneration in diseases such as AD and PD.
Collapse
Affiliation(s)
- Chao Liu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- Neurobiology/Ageing Program, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
| | - Mui Cheng Liang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Neurobiology/Ageing Program, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
| | - Tuck Wah Soong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Neurobiology/Ageing Program, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- National Neuroscience Institute, Singapore, Singapore
| |
Collapse
|
36
|
Wade QW, Chiou B, Connor JR. Iron uptake at the blood-brain barrier is influenced by sex and genotype. PHARMACOLOGY OF RESTLESS LEGS SYNDROME (RLS) 2019; 84:123-145. [DOI: 10.1016/bs.apha.2019.02.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
37
|
Imaging biomarkers of epileptogenecity after traumatic brain injury - Preclinical frontiers. Neurobiol Dis 2018; 123:75-85. [PMID: 30321600 DOI: 10.1016/j.nbd.2018.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/04/2018] [Accepted: 10/11/2018] [Indexed: 02/06/2023] Open
Abstract
Posttraumatic epilepsy (PTE) is a major neurodegenerative disease accounting for 20% of symptomatic epilepsy cases. A long latent phase offers a potential window for prophylactic treatment strategies to prevent epilepsy onset, provided that the patients at risk can be identified. Some promising imaging biomarker candidates for posttraumatic epileptogenesis have been identified, but more are required to provide the specificity and sensitivity for accurate prediction. Experimental models and preclinical longitudinal, multimodal imaging studies allow follow-up of complex cascade of events initiated by traumatic brain injury, as well as monitoring of treatment effects. Preclinical imaging data from the posttraumatic brain are rich in information, yet examination of their specific relevance to epilepsy is lacking. Accumulating evidence from ongoing preclinical studies in TBI support insight into processes involved in epileptogenesis, e.g. inflammation and changes in functional and structural brain-wide connectivity. These efforts are likely to produce both new biomarkers and treatment targets for PTE.
Collapse
|
38
|
Sruthi S, Maurizi L, Nury T, Sallem F, Boudon J, Riedinger J, Millot N, Bouyer F, Lizard G. Cellular interactions of functionalized superparamagnetic iron oxide nanoparticles on oligodendrocytes without detrimental side effects: Cell death induction, oxidative stress and inflammation. Colloids Surf B Biointerfaces 2018; 170:454-462. [DOI: 10.1016/j.colsurfb.2018.06.041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/11/2018] [Accepted: 06/18/2018] [Indexed: 02/02/2023]
|
39
|
The Divalent Metal Transporter 1 (DMT1) Is Required for Iron Uptake and Normal Development of Oligodendrocyte Progenitor Cells. J Neurosci 2018; 38:9142-9159. [PMID: 30190412 DOI: 10.1523/jneurosci.1447-18.2018] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/08/2018] [Accepted: 08/27/2018] [Indexed: 01/24/2023] Open
Abstract
The divalent metal transporter 1 (DMT1) is a multimetal transporter with a primary role in iron transport. Although DMT1 has been described previously in the CNS, nothing was known about the role of this metal transporter in oligodendrocyte maturation and myelination. To determine whether DMT1 is required for oligodendrocyte progenitor cell (OPC) maturation, we used siRNAs and the Cre-lox system to knock down/knock out DMT1 expression in vitro as well as in vivo Blocking DMT1 synthesis in primary cultures of OPCs reduced oligodendrocyte iron uptake and significantly delayed OPC development. In vivo, a significant hypomyelination was found in DMT1 conditional knock-out mice in which DMT1 was postnatally deleted in NG2- or Sox10-positive OPCs. The brain of DMT1 knock-out animals presented a decrease in the expression levels of myelin proteins and a substantial reduction in the percentage of myelinated axons. This reduced postnatal myelination was accompanied by a decrease in the number of myelinating oligodendrocytes and a rise in proliferating OPCs. Furthermore, using the cuprizone model of demyelination, we established that DMT1 deletion in NG2-positive OPCs lead to less efficient remyelination of the adult brain. These results indicate that DMT1 is vital for OPC maturation and for the normal myelination of the mouse brain.SIGNIFICANCE STATEMENT To determine whether divalent metal transporter 1 (DMT1), a multimetal transporter with a primary role in iron transport, is essential for oligodendrocyte development, we created two conditional knock-out mice in which DMT1 was postnatally deleted in NG2- or Sox10-positive oligodendrocyte progenitor cells (OPCs). We have established that DMT1 is necessary for normal OPC maturation and is required for an efficient remyelination of the adult brain. Since iron accumulation by OPCs is indispensable for myelination, understanding the iron incorporation mechanism as well as the molecules involved is critical to design new therapeutic approaches to intervene in diseases in which the myelin sheath is damaged or lost.
Collapse
|
40
|
Birkl C, Carassiti D, Hussain F, Langkammer C, Enzinger C, Fazekas F, Schmierer K, Ropele S. Assessment of ferritin content in multiple sclerosis brains using temperature-induced R* 2 changes. Magn Reson Med 2018; 79:1609-1615. [PMID: 28618066 DOI: 10.1002/mrm.26780] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/13/2017] [Accepted: 05/13/2017] [Indexed: 12/23/2022]
Abstract
PURPOSE Current MRI techniques cannot reliably assess iron content in white matter due to the confounding diamagnetic effect of myelin. The purpose of this study was to validate with histology a novel iron mapping technique that uses the temperature dependency of the paramagnetic susceptibility in multiple sclerosis (MS) brains, where white matter has been reported to show significant variations in iron content. METHODS We investigated post mortem brain tissue from three MS patients and one control subject. Temperature-dependent R2* relaxometry was performed between 4°C and 37°C. The resulting temperature coefficient ( TcR2*) maps were compared with immunohistochemical stains for ferritin light chain. RESULTS Good agreement between TcR2* maps and ferritin staining was found by way of visual comparison and quantitative analysis. The highest iron concentrations were detected at the edge of MS lesions and in the basal ganglia. For all regions, except the subcortical U-fibers, there was a significant negative correlation between the TcR2* values and the ferritin count. CONCLUSION This study provides further evidence that TcR2* may be a reliable measure of white matter iron content due to the elimination of myelin-induced susceptibility changes and is well suited for further research into neurological diseases with distortions of the iron homeostasis. Magn Reson Med 79:1609-1615, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Christoph Birkl
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Daniele Carassiti
- Blizard Institute (Neuroscience), Queen Mary, University of London, London, United Kingdom
| | - Fariha Hussain
- Blizard Institute (Neuroscience), Queen Mary, University of London, London, United Kingdom
| | | | | | - Franz Fazekas
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Klaus Schmierer
- Blizard Institute (Neuroscience), Queen Mary, University of London, London, United Kingdom.,Barts Health NHS Trust, Emergency Care and Acute Medicine Neuroscience Clinical Academic Group, London, United Kingdom
| | - Stefan Ropele
- Department of Neurology, Medical University of Graz, Graz, Austria
| |
Collapse
|
41
|
Gillen KM, Mubarak M, Nguyen TD, Pitt D. Significance and In Vivo Detection of Iron-Laden Microglia in White Matter Multiple Sclerosis Lesions. Front Immunol 2018. [PMID: 29515576 PMCID: PMC5826076 DOI: 10.3389/fimmu.2018.00255] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Microglia are resident immune cells that fulfill protective and homeostatic functions in the central nervous system (CNS) but may also promote neurotoxicity in the aged brain and in chronic disease. In multiple sclerosis (MS), an autoimmune demyelinating disease of the CNS, microglia and macrophages contribute to the development of white matter lesions through myelin phagocytosis, and possibly to disease progression through diffuse activation throughout myelinated white matter. In this review, we discuss an additional compartment of myeloid cell activation in MS, i.e., the rim and normal adjacent white matter of chronic active lesions. In chronic active lesions, microglia and macrophages may contain high amounts of iron, express markers of proinflammatory polarization, are activated for an extended period of time (years), and drive chronic tissue damage. Iron-positive myeloid cells can be visualized and quantified with quantitative susceptibility mapping (QSM), a magnetic resonance imaging technique. Thus, QSM has potential as an in vivo biomarker for chronic inflammatory activity in established white matter MS lesions. Reducing chronic inflammation associated with iron accumulation using existing or novel MS therapies may impact disease severity and progression.
Collapse
Affiliation(s)
- Kelly M Gillen
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Mayyan Mubarak
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| | - Thanh D Nguyen
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - David Pitt
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
42
|
Barone E, Arena A, Head E, Butterfield DA, Perluigi M. Disturbance of redox homeostasis in Down Syndrome: Role of iron dysmetabolism. Free Radic Biol Med 2018; 114:84-93. [PMID: 28705658 PMCID: PMC5748256 DOI: 10.1016/j.freeradbiomed.2017.07.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/05/2017] [Accepted: 07/06/2017] [Indexed: 02/08/2023]
Abstract
Down Syndrome (DS) is the most common genetic form of intellectual disability that leads in the majority of cases to development of early-onset Alzheimer-like dementia (AD). The neuropathology of DS has several common features with AD including alteration of redox homeostasis, mitochondrial deficits, and inflammation among others. Interestingly, some of the genes encoded by chromosome 21 are responsible of increased oxidative stress (OS) conditions that are further exacerbated by decreased antioxidant defense. Previous studies from our groups showed that accumulation of oxidative damage is an early event in DS neurodegeneration and that oxidative modifications of selected proteins affects the integrity of the protein degradative systems, antioxidant response, neuronal integrity and energy metabolism. In particular, the current review elaborates recent findings demonstrating the accumulation of oxidative damage in DS and we focus attention on specific deregulation of iron metabolism, which affects both the central nervous system and the periphery. Iron dysmetabolism is a well-recognized factor that contributes to neurodegeneration; thus we opine that better understanding how and to what extent the concerted loss of iron dyshomeostasis and increased OS occur in DS could provide novel insights for the development of therapeutic strategies for the treatment of Alzheimer-like dementia.
Collapse
Affiliation(s)
- Eugenio Barone
- Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Rome, Italy; Universidad Autónoma de Chile, Instituto de Ciencias Biomédicas, Facultad de alud, Avenida Pedro de Valdivia 425, Providencia, Santiago, Chile
| | - Andrea Arena
- Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Rome, Italy
| | - Elizabeth Head
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506 USA; Department of Neurology, University of Kentucky, Lexington, KY 40506 USA
| | - D Allan Butterfield
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506 USA; Department of Chemistry, University of Kentucky, Lexington, KY 40506 USA
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Rome, Italy.
| |
Collapse
|
43
|
Healy S, McMahon JM, FitzGerald U. Modelling iron mismanagement in neurodegenerative disease in vitro: paradigms, pitfalls, possibilities & practical considerations. Prog Neurobiol 2017; 158:1-14. [DOI: 10.1016/j.pneurobio.2017.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/27/2017] [Accepted: 08/23/2017] [Indexed: 01/26/2023]
|
44
|
Mahajan KR, Ontaneda D. The Role of Advanced Magnetic Resonance Imaging Techniques in Multiple Sclerosis Clinical Trials. Neurotherapeutics 2017; 14:905-923. [PMID: 28770481 PMCID: PMC5722766 DOI: 10.1007/s13311-017-0561-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Magnetic resonance imaging has been crucial in the development of anti-inflammatory disease-modifying treatments. The current landscape of multiple sclerosis clinical trials is currently expanding to include testing not only of anti-inflammatory agents, but also neuroprotective, remyelinating, neuromodulating, and restorative therapies. This is especially true of therapies targeting progressive forms of the disease where neurodegeneration is a prominent feature. Imaging techniques of the brain and spinal cord have rapidly evolved in the last decade to permit in vivo characterization of tissue microstructural changes, connectivity, metabolic changes, neuronal loss, glial activity, and demyelination. Advanced magnetic resonance imaging techniques hold significant promise for accelerating the development of different treatment modalities targeting a variety of pathways in MS.
Collapse
Affiliation(s)
- Kedar R Mahajan
- Mellen Center for Multiple Sclerosis Treatment and Research, Cleveland Clinic, 9500 Euclid Avenue, U-10, Cleveland, OH, 44195, USA
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis Treatment and Research, Cleveland Clinic, 9500 Euclid Avenue, U-10, Cleveland, OH, 44195, USA.
| |
Collapse
|
45
|
Roth AD, Núñez MT. Oligodendrocytes: Functioning in a Delicate Balance Between High Metabolic Requirements and Oxidative Damage. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 949:167-181. [PMID: 27714689 DOI: 10.1007/978-3-319-40764-7_8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The study of the metabolic interactions between myelinating glia and the axons they ensheath has blossomed into an area of research much akin to the elucidation of the role of astrocytes in tripartite synapses (Tsacopoulos and Magistretti in J Neurosci 16:877-885, 1996). Still, unlike astrocytes, rich in cytochrome-P450 and other anti-oxidative defense mechanisms (Minn et al. in Brain Res Brain Res Rev 16:65-82, 1991; Wilson in Can J Physiol Pharmacol. 75:1149-1163, 1997), oligodendrocytes can be easily damaged and are particularly sensitive to both hypoxia and oxidative stress, especially during their terminal differentiation phase and while generating myelin sheaths. In the present review, we will focus in the metabolic complexity of oligodendrocytes, particularly during the processes of differentiation and myelin deposition, and with a specific emphasis in the context of oxidative stress and the intricacies of the iron metabolism of the most iron-loaded cells of the central nervous system (CNS).
Collapse
Affiliation(s)
- Alejandro D Roth
- Department of Biology, Faculty of Science, University of Chile, Santiago, Chile.
| | - Marco T Núñez
- Department of Biology, Faculty of Science, University of Chile, Santiago, Chile
| |
Collapse
|
46
|
Zucca FA, Segura-Aguilar J, Ferrari E, Muñoz P, Paris I, Sulzer D, Sarna T, Casella L, Zecca L. Interactions of iron, dopamine and neuromelanin pathways in brain aging and Parkinson's disease. Prog Neurobiol 2017; 155:96-119. [PMID: 26455458 PMCID: PMC4826627 DOI: 10.1016/j.pneurobio.2015.09.012] [Citation(s) in RCA: 441] [Impact Index Per Article: 55.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 09/14/2015] [Accepted: 09/17/2015] [Indexed: 12/11/2022]
Abstract
There are several interrelated mechanisms involving iron, dopamine, and neuromelanin in neurons. Neuromelanin accumulates during aging and is the catecholamine-derived pigment of the dopamine neurons of the substantia nigra and norepinephrine neurons of the locus coeruleus, the two neuronal populations most targeted in Parkinson's disease. Many cellular redox reactions rely on iron, however an altered distribution of reactive iron is cytotoxic. In fact, increased levels of iron in the brain of Parkinson's disease patients are present. Dopamine accumulation can induce neuronal death; however, excess dopamine can be removed by converting it into a stable compound like neuromelanin, and this process rescues the cell. Interestingly, the main iron compound in dopamine and norepinephrine neurons is the neuromelanin-iron complex, since neuromelanin is an effective metal chelator. Neuromelanin serves to trap iron and provide neuronal protection from oxidative stress. This equilibrium between iron, dopamine, and neuromelanin is crucial for cell homeostasis and in some cellular circumstances can be disrupted. Indeed, when neuromelanin-containing organelles accumulate high load of toxins and iron during aging a neurodegenerative process can be triggered. In addition, neuromelanin released by degenerating neurons activates microglia and the latter cause neurons death with further release of neuromelanin, then starting a self-propelling mechanism of neuroinflammation and neurodegeneration. Considering the above issues, age-related accumulation of neuromelanin in dopamine neurons shows an interesting link between aging and neurodegeneration.
Collapse
Affiliation(s)
- Fabio A Zucca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Juan Segura-Aguilar
- Faculty of Medicine, Molecular and Clinical Pharmacology, ICBM, University of Chile, Santiago, Chile
| | - Emanuele Ferrari
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Patricia Muñoz
- Faculty of Medicine, Molecular and Clinical Pharmacology, ICBM, University of Chile, Santiago, Chile
| | - Irmgard Paris
- Faculty of Medicine, Molecular and Clinical Pharmacology, ICBM, University of Chile, Santiago, Chile; Department of Basic Sciences, Faculty of Sciences, Santo Tomás University, Viña del Mar, Chile
| | - David Sulzer
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA; Department of Neurology, Columbia University Medical Center, New York, NY, USA; Department of Pharmacology, Columbia University Medical Center, New York, NY, USA
| | - Tadeusz Sarna
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Luigi Casella
- Department of Chemistry, University of Pavia, Pavia, Italy
| | - Luigi Zecca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy.
| |
Collapse
|
47
|
Hanini R, Chatti A, Ghorbel SB, Landoulsi A. Role of Sod Gene in Response to Static Magnetic Fields in Pseudomonas aeruginosa. Curr Microbiol 2017; 74:930-937. [PMID: 28523373 DOI: 10.1007/s00284-017-1264-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 05/10/2017] [Indexed: 12/11/2022]
Abstract
The protective role of superoxide dismutase (SOD) against non-ionizing radiation such as static electromagnetic field (200 mT) has been studied in wild-type and mutant strain of Pseudomonas aeruginosa lacking cytosolic Mn-SOD (sodM), Fe-SOD (sodB), or both SODs (sodMB). Our results showed that inactivation of sodM and/or sodB genes increases the sensitivity of P. aeruginosa toward stress induced by the static magnetic field (200 mT). Furthermore, our results showed an enhancement of SOD, catalase, and peroxidases after exposure to the magnetic field. However, wild-type cells maintained significantly higher activities of antioxidant enzymes than mutant strains. The malondialdehyde produced by the oxidative degradation of unsaturated lipids and fatty acids showed significant increase in mutant strains compared to the wild-type. The overall results showed that the SOD has a protective role against a stress induced by static electromagnetic field in P. aeruginosa.
Collapse
Affiliation(s)
- Raouia Hanini
- Unité de Biochimie des lipides et interactions des macromolécules en Biologie (03/UR/0902), Laboratoire de Biochimie et biologie moléculaire, Faculté des Sciences de Bizerte, Zarzouna 7021, Bizerte, Tunisia.
| | - Abdelwaheb Chatti
- Laboratoire de Traitement des Eaux Usées, Centre de Recherches et Technologies des Eaux, Technopole Borj Cedria, Tunis, Tunisia
| | - Selma Ben Ghorbel
- Laboratoire de Traitement des Eaux Usées, Centre de Recherches et Technologies des Eaux, Technopole Borj Cedria, Tunis, Tunisia
| | - Ahmed Landoulsi
- Unité de Biochimie des lipides et interactions des macromolécules en Biologie (03/UR/0902), Laboratoire de Biochimie et biologie moléculaire, Faculté des Sciences de Bizerte, Zarzouna 7021, Bizerte, Tunisia
| |
Collapse
|
48
|
Dulamea AO. Role of Oligodendrocyte Dysfunction in Demyelination, Remyelination and Neurodegeneration in Multiple Sclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 958:91-127. [PMID: 28093710 DOI: 10.1007/978-3-319-47861-6_7] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Oligodendrocytes (OLs) are the myelinating cells of the central nervous system (CNS) during development and throughout adulthood. They result from a complex and well controlled process of activation, proliferation, migration and differentiation of oligodendrocyte progenitor cells (OPCs) from the germinative niches of the CNS. In multiple sclerosis (MS), the complex pathological process produces dysfunction and apoptosis of OLs leading to demyelination and neurodegeneration. This review attempts to describe the patterns of demyelination in MS, the steps involved in oligodendrogenesis and myelination in healthy CNS, the different pathways leading to OLs and myelin loss in MS, as well as principles involved in restoration of myelin sheaths. Environmental factors and their impact on OLs and pathological mechanisms of MS are also discussed. Finally, we will present evidence about the potential therapeutic targets in re-myelination processes that can be accessed in order to develop regenerative therapies for MS.
Collapse
Affiliation(s)
- Adriana Octaviana Dulamea
- Neurology Clinic, University of Medicine and Pharmacy "Carol Davila", Fundeni Clinical Institute, Building A, Neurology Clinic, Room 201, 022328, Bucharest, Romania.
| |
Collapse
|
49
|
Waller R, Woodroofe MN, Wharton SB, Ince PG, Francese S, Heath PR, Cudzich-Madry A, Thomas RH, Rounding N, Sharrack B, Simpson JE. Gene expression profiling of the astrocyte transcriptome in multiple sclerosis normal appearing white matter reveals a neuroprotective role. J Neuroimmunol 2016; 299:139-146. [DOI: 10.1016/j.jneuroim.2016.09.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/12/2016] [Accepted: 09/13/2016] [Indexed: 01/03/2023]
|
50
|
Piñero DJ, Connor JR. Iron in the Brain: An Important Contributor in Normal and Diseased States. Neuroscientist 2016. [DOI: 10.1177/107385840000600607] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Iron is essential for normal neurological function because of its role in oxidative metabolism and because it is a cofactor in the synthesis of neurotransmitters and myelin. In the past several years, there has been increased attention to the importance of oxidative stress in the central nervous system. Iron is the most important inducer of reactive oxygen species, therefore, the relation of iron to neurodegenerative processes is more appreciated today than it was a few years ago. Nevertheless, despite this increased attention and awareness, our knowledge of iron metabolism in the brain at the cellular and molecular levels is still limited. Iron is distributed in a heterogeneous fashion among the different regions and cells of the brain. This regional and cellular heterogeneity is preserved across many species. Brain iron concentrations are not static; they increase with age and in many diseases and decrease when iron is deficient in the diet. In infants and children, insufficient iron in the diet is associated with decreased brain iron and with changes in behavior and cognitive functioning. Abnormal iron accumulation in the diseased brain areas and, in some cases, alterations in iron-related proteins have been reported in many neurodegenerative diseases, including Hallervorden-Spatz syndrome, Alzheimer’s disease, Parkinson’s disease, and Friedreich’s ataxia. There is strong evidence for iron-mediated oxidative damage as a primary contributor to cell death in these disorders. Demyelinating diseases, such as multiple sclerosis, especially warrant study in relation to iron availability. Myelin synthesis and maintenance have a high iron requirement, thus, oligodendrocytes must have a relatively high and constant supply of iron. However, the high oxygen utilization, high density of lipids, and high iron content of white matter all combine to increase the risk of oxidative damage. We review here the current knowledge of the normal metabolism of iron in the brain and the suspected role of iron in neuropathology.
Collapse
Affiliation(s)
- Domingo J. Piñero
- George M. Leader Family Laboratory for Alzheimer’s Disease Research, Department of Neuroscience & Anatomy, Penn State University, College of Medicine, Hershey, Pennsylvania
| | - James R. Connor
- George M. Leader Family Laboratory for Alzheimer’s Disease Research, Department of Neuroscience & Anatomy, Penn State University, College of Medicine, Hershey, Pennsylvania,
| |
Collapse
|