1
|
Yan HF, Tuo QZ, Lei P. Cell density impacts the susceptibility to ferroptosis by modulating IRP1-mediated iron homeostasis. J Neurochem 2024; 168:1359-1373. [PMID: 38382918 DOI: 10.1111/jnc.16085] [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: 08/21/2023] [Revised: 01/28/2024] [Accepted: 02/06/2024] [Indexed: 02/23/2024]
Abstract
Ferroptosis has been implicated in several neurological disorders and may be therapeutically targeted. However, the susceptibility to ferroptosis varies in different cells, and inconsistent results have been reported even using the same cell line. Understanding the effects of key variables of in vitro studies on ferroptosis susceptibility is of critical importance to facilitate drug discoveries targeting ferroptosis. Here, we showed that increased cell seeding density leads to enhanced resistance to ferroptosis by reducing intracellular iron levels. We further identified iron-responsive protein 1 (IRP1) as the key protein affected by cell density, which affects the expression of ferroportin or transferrin receptor and results in altered iron levels. Such observations were consistent across different cell lines, indicating that cell density should be tightly controlled in studies of ferroptosis. Since cell densities vary in different brain regions, these results may also shed light on selective regional vulnerability observed in neurological disorders.
Collapse
Affiliation(s)
- Hong-Fa Yan
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qing-Zhang Tuo
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Langley J, Bennett IJ, Hu XP. Examining iron-related off-target binding effects of 18F-AV1451 PET in the cortex of Aβ+ individuals. Eur J Neurosci 2024; 60:3614-3628. [PMID: 38722153 DOI: 10.1111/ejn.16362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 12/22/2023] [Accepted: 04/01/2024] [Indexed: 07/06/2024]
Abstract
The presence of neurofibrillary tangles containing hyper-phosphorylated tau is a characteristic of Alzheimer's disease (AD) pathology. The positron emission tomography (PET) radioligand sensitive to tau neurofibrillary tangles (18F-AV1451) also binds with iron. This off-target binding effect may be enhanced in older adults on the AD spectrum, particularly those with amyloid-positive biomarkers. Here, we examined group differences in 18F-AV1451 PET after controlling for iron-sensitive measures from magnetic resonance imaging (MRI) and its relationships to tissue microstructure and cognition in 40 amyloid beta positive (Aβ+) individuals, 20 amyloid beta negative (Aβ-) with MCI and 31 Aβ- control participants. After controlling for iron, increased 18F-AV1451 PET uptake was found in the temporal lobe and hippocampus of Aβ+ participants compared to Aβ- MCI and control participants. Within the Aβ+ group, significant correlations were seen between 18F-AV1451 PET uptake and tissue microstructure and these correlations remained significant after controlling for iron. These findings indicate that off-target binding of iron to the 18F-AV1451 ligand may not affect its sensitivity to Aβ status or cognition in early-stage AD.
Collapse
Affiliation(s)
- Jason Langley
- Center for Advanced Neuroimaging, University of California Riverside, Riverside, California, USA
| | - Ilana J Bennett
- Department of Psychology, University of California Riverside, Riverside, California, USA
| | - Xiaoping P Hu
- Center for Advanced Neuroimaging, University of California Riverside, Riverside, California, USA
- Department of Bioengineering, University of California Riverside, Riverside, California, USA
| |
Collapse
|
3
|
Su H, Masters CL, Bush AI, Barnham KJ, Reid GE, Vella LJ. Exploring the significance of lipids in Alzheimer's disease and the potential of extracellular vesicles. Proteomics 2024; 24:e2300063. [PMID: 37654087 DOI: 10.1002/pmic.202300063] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/07/2023] [Accepted: 08/14/2023] [Indexed: 09/02/2023]
Abstract
Lipids play a significant role in maintaining central nervous system (CNS) structure and function, and the dysregulation of lipid metabolism is known to occur in many neurological disorders, including Alzheimer's disease. Here we review what is currently known about lipid dyshomeostasis in Alzheimer's disease. We propose that small extracellular vesicle (sEV) lipids may provide insight into the pathophysiology and progression of Alzheimer's disease. This stems from the recognition that sEV likely contributes to disease pathogenesis, but also an understanding that sEV can serve as a source of potential biomarkers. While the protein and RNA content of sEV in the CNS diseases have been studied extensively, our understanding of the lipidome of sEV in the CNS is still in its infancy.
Collapse
Affiliation(s)
- Huaqi Su
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Ashley I Bush
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Kevin J Barnham
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Gavin E Reid
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - Laura J Vella
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
4
|
LeVine SM. Exploring Potential Mechanisms Accounting for Iron Accumulation in the Central Nervous System of Patients with Alzheimer's Disease. Cells 2024; 13:689. [PMID: 38667304 PMCID: PMC11049304 DOI: 10.3390/cells13080689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/12/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
Elevated levels of iron occur in both cortical and subcortical regions of the CNS in patients with Alzheimer's disease. This accumulation is present early in the disease process as well as in more advanced stages. The factors potentially accounting for this increase are numerous, including: (1) Cells increase their uptake of iron and reduce their export of iron, as iron becomes sequestered (trapped within the lysosome, bound to amyloid β or tau, etc.); (2) metabolic disturbances, such as insulin resistance and mitochondrial dysfunction, disrupt cellular iron homeostasis; (3) inflammation, glutamate excitotoxicity, or other pathological disturbances (loss of neuronal interconnections, soluble amyloid β, etc.) trigger cells to acquire iron; and (4) following neurodegeneration, iron becomes trapped within microglia. Some of these mechanisms are also present in other neurological disorders and can also begin early in the disease course, indicating that iron accumulation is a relatively common event in neurological conditions. In response to pathogenic processes, the directed cellular efforts that contribute to iron buildup reflect the importance of correcting a functional iron deficiency to support essential biochemical processes. In other words, cells prioritize correcting an insufficiency of available iron while tolerating deposited iron. An analysis of the mechanisms accounting for iron accumulation in Alzheimer's disease, and in other relevant neurological conditions, is put forward.
Collapse
Affiliation(s)
- Steven M LeVine
- Department of Cell Biology and Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd., Mail Stop 3043, Kansas City, KS 66160, USA
| |
Collapse
|
5
|
Lee S, Kovacs GG. The Irony of Iron: The Element with Diverse Influence on Neurodegenerative Diseases. Int J Mol Sci 2024; 25:4269. [PMID: 38673855 PMCID: PMC11049980 DOI: 10.3390/ijms25084269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Iron accumulation in the brain is a common feature of many neurodegenerative diseases. Its involvement spans across the main proteinopathies involving tau, amyloid-beta, alpha-synuclein, and TDP-43. Accumulating evidence supports the contribution of iron in disease pathologies, but the delineation of its pathogenic role is yet challenged by the complex involvement of iron in multiple neurotoxicity mechanisms and evidence supporting a reciprocal influence between accumulation of iron and protein pathology. Here, we review the major proteinopathy-specific observations supporting four distinct hypotheses: (1) iron deposition is a consequence of protein pathology; (2) iron promotes protein pathology; (3) iron protects from or hinders protein pathology; and (4) deposition of iron and protein pathology contribute parallelly to pathogenesis. Iron is an essential element for physiological brain function, requiring a fine balance of its levels. Understanding of disease-related iron accumulation at a more intricate and systemic level is critical for advancements in iron chelation therapies.
Collapse
Affiliation(s)
- Seojin Lee
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5T 0S8, Canada;
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gabor G. Kovacs
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5T 0S8, Canada;
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Edmond J. Safra Program in Parkinson’s Disease, Rossy Program for PSP Research and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON M5T 2S8, Canada
| |
Collapse
|
6
|
Huang Z, Jordan JD, Zhang Q. Myelin Pathology in Alzheimer's Disease: Potential Therapeutic Opportunities. Aging Dis 2024; 15:698-713. [PMID: 37548935 PMCID: PMC10917545 DOI: 10.14336/ad.2023.0628] [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: 04/27/2023] [Accepted: 06/28/2023] [Indexed: 08/08/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by memory loss and cognitive decline. Despite significant efforts over several decades, our understanding of the pathophysiology of this disease is still incomplete. Myelin is a multi-layered membrane structure ensheathing neuronal axons, which is essential for the fast and effective propagation of action potentials along the axons. Recent studies highlight the critical involvement of myelin in memory consolidation and reveal its vulnerability in various pathological conditions. Notably, apart from the classic amyloid hypothesis, myelin degeneration has been proposed as another critical pathophysiological feature of AD, which could occur prior to the development of amyloid pathology. Here, we review recent works supporting the critical role of myelin in cognition and myelin pathology during AD progression, with a focus on the mechanisms underlying myelin degeneration in AD. We also discuss the complex intersections between myelin pathology and typical AD pathophysiology, as well as the therapeutic potential of pro-myelinating approaches for this disease. Overall, these findings implicate myelin degeneration as a critical contributor to AD-related cognitive deficits and support targeting myelin repair as a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Zhihai Huang
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71103 USA
| | - J. Dedrick Jordan
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71103 USA
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71103 USA
| |
Collapse
|
7
|
Cha WJ, Yi D, Ahn H, Byun MS, Chang YY, Choi JM, Kim K, Choi H, Jung G, Kang KM, Sohn CH, Lee YS, Kim YK, Lee DY. Association between brain amyloid deposition and longitudinal changes of white matter hyperintensities. Alzheimers Res Ther 2024; 16:50. [PMID: 38454444 PMCID: PMC10918927 DOI: 10.1186/s13195-024-01417-8] [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: 11/04/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Growing evidence suggests that not only cerebrovascular disease but also Alzheimer's disease (AD) pathological process itself cause cerebral white matter degeneration, resulting in white matter hyperintensities (WMHs). Some preclinical evidence also indicates that white matter degeneration may precede or affect the development of AD pathology. This study aimed to clarify the direction of influence between in vivo AD pathologies, particularly beta-amyloid (Aβ) and tau deposition, and WMHs through longitudinal approach. METHODS Total 282 older adults including cognitively normal and cognitively impaired individuals were recruited from the Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer's Disease (KBASE) cohort. The participants underwent comprehensive clinical and neuropsychological assessment, [11C] Pittsburgh Compound B PET for measuring Aβ deposition, [18F] AV-1451 PET for measuring tau deposition, and MRI scans with fluid-attenuated inversion recovery image for measuring WMH volume. The relationships between Aβ or tau deposition and WMH volume were examined using multiple linear regression analysis. In this analysis, baseline Aβ or tau were used as independent variables, and change of WMH volume over 2 years was used as dependent variable to examine the effect of AD pathology on increase of WMH volume. Additionally, we set baseline WMH volume as independent variable and longitudinal change of Aβ or tau deposition for 2 years as dependent variables to investigate whether WMH volume could precede AD pathologies. RESULTS Baseline Aβ deposition, but not tau deposition, had significant positive association with longitudinal change of WMH volume over 2 years. Baseline WMH volume was not related with any of longitudinal change of Aβ or tau deposition for 2 years. We also found a significant interaction effect between baseline Aβ deposition and sex on longitudinal change of WMH volume. Subsequent subgroup analyses showed that high baseline Aβ deposition was associated with increase of WMH volume over 2 years in female, but not in male. CONCLUSIONS Our findings suggest that Aβ deposition accelerates cerebral WMHs, particularly in female, whereas white matter degeneration appears not influence on longitudinal Aβ increase. The results also did not support any direction of influence between tau deposition and WMHs.
Collapse
Affiliation(s)
- Woo-Jin Cha
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Hyejin Ahn
- Interdisciplinary program of cognitive science, Seoul National University College of Humanities, Seoul, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yoon Young Chang
- Department of Psychiatry, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Jung-Min Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyungtae Kim
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyeji Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea.
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea.
- Interdisciplinary program of cognitive science, Seoul National University College of Humanities, Seoul, Republic of Korea.
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Rubinski A, Dewenter A, Zheng L, Franzmeier N, Stephenson H, Deming Y, Duering M, Gesierich B, Denecke J, Pham AV, Bendlin B, Ewers M. Florbetapir PET-assessed demyelination is associated with faster tau accumulation in an APOE ε4-dependent manner. Eur J Nucl Med Mol Imaging 2024; 51:1035-1049. [PMID: 38049659 PMCID: PMC10881623 DOI: 10.1007/s00259-023-06530-8] [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: 05/18/2023] [Accepted: 11/14/2023] [Indexed: 12/06/2023]
Abstract
PURPOSE The main objectives were to test whether (1) a decrease in myelin is associated with enhanced rate of fibrillar tau accumulation and cognitive decline in Alzheimer's disease, and (2) whether apolipoprotein E (APOE) ε4 genotype is associated with worse myelin decrease and thus tau accumulation. METHODS To address our objectives, we repurposed florbetapir-PET as a marker of myelin in the white matter (WM) based on previous validation studies showing that beta-amyloid (Aβ) PET tracers bind to WM myelin. We assessed 43 Aβ-biomarker negative (Aβ-) cognitively normal participants and 108 Aβ+ participants within the AD spectrum with florbetapir-PET at baseline and longitudinal flortaucipir-PET as a measure of fibrillar tau (tau-PET) over ~ 2 years. In linear regression analyses, we tested florbetapir-PET in the whole WM and major fiber tracts as predictors of tau-PET accumulation in a priori defined regions of interest (ROIs) and fiber-tract projection areas. In mediation analyses we tested whether tau-PET accumulation mediates the effect of florbetapir-PET in the whole WM on cognition. Finally, we assessed the role of myelin alteration on the association between APOE and tau-PET accumulation. RESULTS Lower florbetapir-PET in the whole WM or at a given fiber tract was predictive of faster tau-PET accumulation in Braak stages or the connected grey matter areas in Aβ+ participants. Faster tau-PET accumulation in higher cortical brain areas mediated the association between a decrease in florbetapir-PET in the WM and a faster rate of decline in global cognition and episodic memory. APOE ε4 genotype was associated with a worse decrease in the whole WM florbetapir-PET and thus enhanced tau-PET accumulation. CONCLUSION Myelin alterations are associated in an APOE ε4 dependent manner with faster tau progression and cognitive decline, and may therefore play a role in the etiology of AD.
Collapse
Affiliation(s)
- Anna Rubinski
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Anna Dewenter
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Lukai Zheng
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Henry Stephenson
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, WI, USA
| | - Yuetiva Deming
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, WI, USA
| | - Marco Duering
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
- Medical Image Analysis Center (MIAC) and Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Benno Gesierich
- Medical Image Analysis Center (MIAC) and Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Jannis Denecke
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
| | - An-Vi Pham
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
- Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Barbara Bendlin
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, WI, USA
| | - Michael Ewers
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| |
Collapse
|
9
|
Perera Molligoda Arachchige AS, Garner AK. Seven Tesla MRI in Alzheimer's disease research: State of the art and future directions: A narrative review. AIMS Neurosci 2023; 10:401-422. [PMID: 38188012 PMCID: PMC10767068 DOI: 10.3934/neuroscience.2023030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Seven tesla magnetic resonance imaging (7T MRI) is known to offer a superior spatial resolution and a signal-to-noise ratio relative to any other non-invasive imaging technique and provides the possibility for neuroimaging researchers to observe disease-related structural changes, which were previously only apparent on post-mortem tissue analyses. Alzheimer's disease is a natural and widely used subject for this technology since the 7T MRI allows for the anticipation of disease progression, the evaluation of secondary prevention measures thought to modify the disease trajectory, and the identification of surrogate markers for treatment outcome. In this editorial, we discuss the various neuroimaging biomarkers for Alzheimer's disease that have been studied using 7T MRI, which include morphological alterations, molecular characterization of cerebral T2*-weighted hypointensities, the evaluation of cerebral microbleeds and microinfarcts, biochemical changes studied with MR spectroscopy, as well as some other approaches. Finally, we discuss the limitations of the 7T MRI regarding imaging Alzheimer's disease and we provide our outlook for the future.
Collapse
|
10
|
Moallemian S, Salmon E, Bahri MA, Beliy N, Delhaye E, Balteau E, Degueldre C, Phillips C, Bastin C. Multimodal imaging of microstructural cerebral alterations and loss of synaptic density in Alzheimer's disease. Neurobiol Aging 2023; 132:24-35. [PMID: 37717552 DOI: 10.1016/j.neurobiolaging.2023.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/01/2023] [Accepted: 08/05/2023] [Indexed: 09/19/2023]
Abstract
Multiple neuropathological events are involved in Alzheimer's disease (AD). The current study investigated the concurrence of neurodegeneration, increased iron content, atrophy, and demyelination in AD. Quantitative multiparameter magnetic resonance imaging (MRI) maps providing neuroimaging biomarkers for myelination and iron content along with synaptic density measurements using [18F] UCB-H PET were acquired in 24 AD and 19 Healthy controls (19 males). The whole brain voxel-wise group comparison revealed demyelination in the right hippocampus, while no significant iron content difference was detected. Bilateral atrophy and synaptic density loss were observed in the hippocampus and amygdala. The multivariate GLM (mGLM) analysis shows a bilateral difference in the hippocampus and amygdala, right pallidum, left fusiform and temporal lobe suggesting that these regions are the most affected despite the diverse differences in brain tissue properties in AD. Demyelination was identified as the most affecting factor in the observed differences. Here, the mGLM is introduced as an alternative for multiple comparisons between different modalities, reducing the risk of false positives while informing about the co-occurrence of neuropathological processes in AD.
Collapse
Affiliation(s)
- Soodeh Moallemian
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Eric Salmon
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Mohamed Ali Bahri
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Nikita Beliy
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Emma Delhaye
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Evelyne Balteau
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Christian Degueldre
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Christophe Phillips
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| | - Christine Bastin
- GIGA-Cyclotron Research Centre-In Vivo Imaging, University of Liège, Liège, Belgium.
| |
Collapse
|
11
|
Kruggel F, Solodkin A. Analyzing the cortical fine structure as revealed by ex-vivo anatomical MRI. J Comp Neurol 2023; 531:2146-2161. [PMID: 37522626 DOI: 10.1002/cne.25532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 04/15/2023] [Accepted: 06/21/2023] [Indexed: 08/01/2023]
Abstract
The human cortex has a rich fiber structure as revealed by myelin-staining of histological slices. Myelin also contributes to the image contrast in Magnetic Resonance Imaging (MRI). Recent advances in Magnetic Resonance (MR) scanner and imaging technology allowed the acquisition of an ex-vivo data set at an isotropic resolution of 100 µm. This study focused on a computational analysis of this data set with the aim of bridging between histological knowledge and MRI-based results. This work highlights: (1) the design and implementation of a processing chain that extracts intracortical features from a high-resolution MR image; (2) a demonstration of the correspondence between MRI-based cortical intensity profiles and the myelo-architectonic layering of the cortex; (3) the characterization and classification of four basic myelo-architectonic profile types; (4) the distinction of cortical regions based on myelo-architectonic features; and (5) the segmentation of cortical modules in the entorhinal cortex.
Collapse
Affiliation(s)
- Frithjof Kruggel
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA
| | - Ana Solodkin
- School of Behavioral and Brain Sciences, University of Texas, Richardson, Texas, USA
| |
Collapse
|
12
|
Baringer SL, Lukacher AS, Palsa K, Kim H, Lippmann ES, Spiegelman VS, Simpson IA, Connor JR. Amyloid-β exposed astrocytes induce iron transport from endothelial cells at the blood-brain barrier by altering the ratio of apo- and holo-transferrin. J Neurochem 2023; 167:248-261. [PMID: 37667496 PMCID: PMC10592116 DOI: 10.1111/jnc.15954] [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: 05/17/2023] [Revised: 07/30/2023] [Accepted: 08/21/2023] [Indexed: 09/06/2023]
Abstract
Excessive brain iron accumulation is observed early in the onset of Alzheimer's disease, notably prior to widespread proteinopathy. These findings suggest that increases in brain iron levels are due to a dysregulation of the iron transport mechanism at the blood-brain barrier. Astrocytes release signals (apo- and holo-transferrin) that communicate brain iron needs to endothelial cells in order to modulate iron transport. Here we use iPSC-derived astrocytes and endothelial cells to investigate how early-disease levels of amyloid-β disrupt iron transport signals secreted by astrocytes to stimulate iron transport from endothelial cells. We demonstrate that conditioned media from astrocytes treated with amyloid-β stimulates iron transport from endothelial cells and induces changes in iron transport pathway proteins. The mechanism underlying this response begins with increased iron uptake and mitochondrial activity by the astrocytes, which in turn increases levels of apo-transferrin in the amyloid-β conditioned astrocyte media leading to increased iron transport from endothelial cells. These novel findings offer a potential explanation for the initiation of excessive iron accumulation in early stages of Alzheimer's disease. What's more, these data provide the first example of how the mechanism of iron transport regulation by apo- and holo-transferrin becomes misappropriated in disease that can lead to iron accumulation. The clinical benefit from understanding early dysregulation in brain iron transport in AD cannot be understated. If therapeutics can target this early process, they could possibly prevent the detrimental cascade that occurs with excessive iron accumulation.
Collapse
Affiliation(s)
- Stephanie L. Baringer
- Department of Neurosurgery, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033
| | - Avraham S. Lukacher
- Department of Neurosurgery, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033
| | - Kondaiah Palsa
- Department of Neurosurgery, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033
| | - Hyosung Kim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, 2201 West End Ave, Nashville, TN, USA, 37235
| | - Ethan S. Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, 2201 West End Ave, Nashville, TN, USA, 37235
| | - Vladimir S. Spiegelman
- Department of Pediatrics, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033
| | - Ian A. Simpson
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033
| | - James R. Connor
- Department of Neurosurgery, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033
| |
Collapse
|
13
|
Baringer SL, Lukacher AS, Palsa K, Kim H, Lippmann ES, Spiegelman VS, Simpson IA, Connor JR. Amyloid-β exposed astrocytes induce iron transport from endothelial cells at the blood-brain barrier by altering the ratio of apo- and holo-transferrin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.15.540795. [PMID: 37292926 PMCID: PMC10245582 DOI: 10.1101/2023.05.15.540795] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Excessive brain iron accumulation is observed in early in the onset of Alzheimer's disease, notably prior to widespread proteinopathy. These findings suggest that increases in brain iron levels are due to a dysregulation of the iron transport mechanism at the blood-brain barrier. Astrocytes release signals (apo- and holo-transferrin) that communicate brain iron needs to endothelial cells in order to modulate iron transport. Here we use iPSC-derived astrocytes and endothelial cells to investigate how early-disease levels of amyloid-β disrupt iron transport signals secreted by astrocytes to stimulate iron transport from endothelial cells. We demonstrate that conditioned media from astrocytes treated with amyloid-β stimulates iron transport from endothelial cells and induces changes in iron transport pathway protein levels. The mechanism underlying this response begins with increased iron uptake and mitochondrial activity by the astrocytes which in turn increases levels of apo-transferrin in the amyloid-β conditioned astrocyte media leading to increased iron transport from endothelial cells. These novel findings offer a potential explanation for the initiation of excessive iron accumulation in early stages of Alzheimer's disease. What's more, these data provide the first example of how the mechanism of iron transport regulation by apo- and holo-transferrin becomes misappropriated in disease to detrimental ends. The clinical benefit from understanding early dysregulation in brain iron transport in AD cannot be understated. If therapeutics can target this early process, they could possibly prevent the detrimental cascade that occurs with excessive iron accumulation. Significance Statement Excessive brain iron accumulation is hallmark pathology of Alzheimer's disease that occurs early in the disease staging and before widespread proteinopathy deposition. This overabundance of brain iron has been implicated to contribute to disease progression, thus understandingthe mechanism of early iron accumulation has significant therapeutic potential to slow to halt disease progression. Here, we show that in response to low levels of amyloid-β exposure, astrocytes increase their mitochondrial activity and iron uptake, resulting in iron deficient conditions. Elevated levels of apo (iron free)-transferrin stimulate iron release from endothelial cells. These data are the first to propose a mechanism for the initiation of iron accumulation and the misappropriation of iron transport signaling leading to dysfunctional brain iron homeostasis and resultant disease pathology.
Collapse
|
14
|
Wu L, Xian X, Tan Z, Dong F, Xu G, Zhang M, Zhang F. The Role of Iron Metabolism, Lipid Metabolism, and Redox Homeostasis in Alzheimer's Disease: from the Perspective of Ferroptosis. Mol Neurobiol 2023; 60:2832-2850. [PMID: 36735178 DOI: 10.1007/s12035-023-03245-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/26/2023] [Indexed: 02/04/2023]
Abstract
In the development of Alzheimer's disease (AD), cell death is common. Novel cell death form-ferroptosis is discovered in recent years. Ferroptosis is an iron-regulated programmed cell death mechanism and has been identified in AD clinical samples. Typical characteristics of ferroptosis involve the specific changes in cell morphology, iron-dependent aggregation of reactive oxygen species (ROS) and lipid peroxides, loss of glutathione (GSH), inactivation of glutathione peroxidase 4 (GPX4), and a unique group of regulatory genes. Increasing evidence demonstrates that ferroptosis may be associated with neurological dysfunction in AD. However, the underlying mechanisms have not been fully elucidated. This article reviews the potential role of ferroptosis in AD, the involvement of ferroptosis in the pathological progression of AD through the mechanisms of iron metabolism, lipid metabolism, and redox homeostasis, as well as a range of potential therapies targeting ferroptosis for AD. Intervention strategies based on ferroptosis are promising for Alzheimer's disease treatment at present, but further researches are still needed.
Collapse
Affiliation(s)
- Linyu Wu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Xiaohui Xian
- Department of Pathophysiology, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050051, Hebei, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050051, People's Republic of China
| | - Zixuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, People's Republic of China
| | - Guangyu Xu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050051, Hebei, People's Republic of China.
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050051, People's Republic of China.
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China.
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050051, People's Republic of China.
| |
Collapse
|
15
|
Li Z, Feng R, Liu Q, Feng J, Lao G, Zhang M, Li J, Zhang Y, Wei H. APART-QSM: an improved sub-voxel quantitative susceptibility mapping for susceptibility source separation using an iterative data fitting method. Neuroimage 2023; 274:120148. [PMID: 37127191 DOI: 10.1016/j.neuroimage.2023.120148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/06/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023] Open
Abstract
The brain tissue phase contrast in MRI sequences reflects the spatial distributions of multiple substances, such as iron, myelin, calcium, and proteins. These substances with paramagnetic and diamagnetic susceptibilities often colocalize in one voxel in brain regions. Both opposing susceptibilities play vital roles in brain development and neurodegenerative diseases. Conventional QSM methods only provide voxel-averaged susceptibility value and cannot disentangle intravoxel susceptibilities with opposite signs. Advanced susceptibility imaging methods have been recently developed to distinguish the contributions of opposing susceptibility sources for QSM. The basic concept of separating paramagnetic and diamagnetic susceptibility proportions is to include the relaxation rate R2* with R2' in QSM. The magnitude decay kernel, describing the proportionality coefficient between R2' and susceptibility, is an essential reconstruction coefficient for QSM separation methods. In this study, we proposed a more comprehensive complex signal model that describes the relationship between 3D GRE signal and the contributions of paramagnetic and diamagnetic susceptibility to the frequency shift and R2* relaxation. The algorithm is implemented as a constrained minimization problem in which the voxel-wise magnitude decay kernel and sub-voxel susceptibilities are determined alternately in each iteration until convergence. The calculated voxel-wise magnitude decay kernel could realistically model the relationship between the R2' relaxation and the volume susceptibility. Thus, the proposed method effectively prevents the errors of the magnitude decay kernel from propagating to the final susceptibility separation reconstruction. Phantom studies, ex vivo macaque brain experiments, and in vivo human brain imaging studies were conducted to evaluate the ability of the proposed method to distinguish paramagnetic and diamagnetic susceptibility sources. The results demonstrate that the proposed method provides state-of-the-art performances for quantifying brain iron and myelin compared to previous QSM separation methods. Our results show that the proposed method has the potential to simultaneously quantify whole brain iron and myelin during brain development and aging. The proposed model was also deployed with multiple-orientation complex GRE data input measurements, resulting in high-quality QSM separation maps with more faithful tissue delineation between brain structures compared to those reconstructed by single-orientation QSM separation methods.
Collapse
Affiliation(s)
- Zhenghao Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ruimin Feng
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qiangqiang Liu
- Department of Neurosurgery, Clinical Neuroscience Center Comprehensive Epilepsy Unit, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Feng
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guoyan Lao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Li
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yuyao Zhang
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hongjiang Wei
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
16
|
Sun 孙意冉 Y, Yan C, He L, Xiang S, Wang P, Li Z, Chen Y, Zhao J, Yuan Y, Wang W, Zhang X, Su P, Su Y, Ma J, Xu J, Peng Q, Ma H, Xie Z, Zhang Z. Inhibition of ferroptosis through regulating neuronal calcium homeostasis: An emerging therapeutic target for Alzheimer's disease. Ageing Res Rev 2023; 87:101899. [PMID: 36871781 DOI: 10.1016/j.arr.2023.101899] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/22/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Alzheimer's disease (AD), a chronic and progressive neurodegenerative disease, generates a serious threat to the health of the elderly. The AD brain is microscopically characterized by amyloid plaques and neurofibrillary tangles. There are still no effective therapeutic drugs to restrain the progression of AD though much attention has been paid to exploit AD treatments. Ferroptosis, a type of programmed cell death, has been reported to promote the pathological occurrence and development of AD, and inhibition of neuronal ferroptosis can effectively improve the cognitive impairment of AD. Studies have shown that calcium (Ca2+) dyshomeostasis is closely related to the pathology of AD, and can drive the occurrence of ferroptosis through several pathways, such as interacting with iron, and regulating the crosstalk between endoplasmic reticulum (ER) and mitochondria. This paper mainly reviews the roles of ferroptosis and Ca2+ in the pathology of AD, and highlights that restraining ferroptosis through maintaining the homeostasis of Ca2+ may be an innovative target for the treatment of AD.
Collapse
Affiliation(s)
- Yiran Sun 孙意冉
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Chenchen Yan
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Libo He
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, Sichuan, China
| | - Shixie Xiang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Pan Wang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Zhonghua Li
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yuanzhao Chen
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jie Zhao
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Ye Yuan
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Wang Wang
- School of basic medicine, Nanchang Medical College, Nanchang 330052, Jiangxi, China
| | - Xiaowei Zhang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Pan Su
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yunfang Su
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jinlian Ma
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jiangyan Xu
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Quekun Peng
- School of Biosciences and Technology, Chengdu Medical College, Chengdu 610500, China.
| | - Huifen Ma
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Zhishen Xie
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Zhenqiang Zhang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| |
Collapse
|
17
|
Monteiro AR, Barbosa DJ, Remião F, Silva R. Alzheimer’s disease: insights and new prospects in disease pathophysiology, biomarkers and disease-modifying drugs. Biochem Pharmacol 2023; 211:115522. [PMID: 36996971 DOI: 10.1016/j.bcp.2023.115522] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases that affect millions of people worldwide, with both prevalence and incidence increasing with age. It is characterized by cognitive decline associated, specifically, with degeneration of cholinergic neurons. The problem of this disease is even more fundamental as the available therapies remain fairly limited and mainly focused on symptoms' relief. Although the aetiology of the disease remains elusive, two main pathological hallmarks are described: i) presence of neurofibrillary tangles formed by unfolded protein aggregates (hyperphosphorylated Tau protein) and ii) presence of extracellular aggregates of amyloid-beta peptide. Given the complexity surrounding the pathogenesis of the disease, several potential targets have been highlighted and interrelated upon its progression, such as oxidative stress and the accumulation of metal ions. Thus, advances have been made on the development of innovative multitarget therapeutical compounds to delay the disease progression and restore cell function. This review focuses the ongoing research on new insights and emerging disease-modifying drugs for AD treatment. Furthermore, classical and novel potential biomarkers for early diagnosis of the disease, and their role in assisting on the improvement of targeted therapies will also be approached.
Collapse
Affiliation(s)
- Ana R Monteiro
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, Porto University, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Daniel J Barbosa
- TOXRUN - Toxicology Research Unit, Department of Sciences, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Fernando Remião
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, Porto University, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Renata Silva
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, Porto University, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
18
|
Paquola C, Hong SJ. The Potential of Myelin-Sensitive Imaging: Redefining Spatiotemporal Patterns of Myeloarchitecture. Biol Psychiatry 2023; 93:442-454. [PMID: 36481065 DOI: 10.1016/j.biopsych.2022.08.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/12/2022] [Accepted: 08/30/2022] [Indexed: 02/07/2023]
Abstract
Recent advances in magnetic resonance imaging (MRI) have paved the way for approximation of myelin content in vivo. In this review, our main goal was to determine how to best capitalize on myelin-sensitive imaging. First, we briefly overview the theoretical and empirical basis for the myelin sensitivity of different MRI markers and, in doing so, highlight how multimodal imaging approaches are important for enhancing specificity to myelin. Then, we discuss recent studies that have probed the nonuniform distribution of myelin across cortical layers and along white matter tracts. These approaches, collectively known as myelin profiling, have provided detailed depictions of myeloarchitecture in both the postmortem and living human brain. Notably, MRI-based profiling studies have recently focused on investigating whether it can capture interindividual variability in myelin characteristics as well as trajectories across the lifespan. Finally, another line of recent evidence emphasizes the contribution of region-specific myelination to large-scale organization, demonstrating the impact of myelination on global brain networks. In conclusion, we suggest that combining well-validated MRI markers with profiling techniques holds strong potential to elucidate individual differences in myeloarchitecture, which has important implications for understanding brain function and disease.
Collapse
Affiliation(s)
- Casey Paquola
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany.
| | - Seok-Jun Hong
- Center for Neuroscience Imaging Research, Institute for Basic Science, Sungkyunkwan University, Suwon, South Korea; Center for the Developing Brain, Child Mind Institute, New York, New York; Department of Biomedical Engineering, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
19
|
Li K, Rashid T, Li J, Honnorat N, Nirmala AB, Fadaee E, Wang D, Charisis S, Liu H, Franklin C, Maybrier M, Katragadda H, Abazid L, Ganapathy V, Valaparla VL, Badugu P, Vasquez E, Solano L, Clarke G, Maestre G, Richardson T, Walker J, Fox PT, Bieniek K, Seshadri S, Habes M. Postmortem Brain Imaging in Alzheimer's Disease and Related Dementias: The South Texas Alzheimer's Disease Research Center Repository. J Alzheimers Dis 2023; 96:1267-1283. [PMID: 37955086 PMCID: PMC10693476 DOI: 10.3233/jad-230389] [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] [Accepted: 09/24/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Neuroimaging bears the promise of providing new biomarkers that could refine the diagnosis of dementia. Still, obtaining the pathology data required to validate the relationship between neuroimaging markers and neurological changes is challenging. Existing data repositories are focused on a single pathology, are too small, or do not precisely match neuroimaging and pathology findings. OBJECTIVE The new data repository introduced in this work, the South Texas Alzheimer's Disease research center repository, was designed to address these limitations. Our repository covers a broad diversity of dementias, spans a wide age range, and was specifically designed to draw exact correspondences between neuroimaging and pathology data. METHODS Using four different MRI sequences, we are reaching a sample size that allows for validating multimodal neuroimaging biomarkers and studying comorbid conditions. Our imaging protocol was designed to capture markers of cerebrovascular disease and related lesions. Quantification of these lesions is currently underway with MRI-guided histopathological examination. RESULTS A total of 139 postmortem brains (70 females) with mean age of 77.9 years were collected, with 71 brains fully analyzed. Of these, only 3% showed evidence of AD-only pathology and 76% had high prevalence of multiple pathologies contributing to clinical diagnosis. CONCLUSION This repository has a significant (and increasing) sample size consisting of a wide range of neurodegenerative disorders and employs advanced imaging protocols and MRI-guided histopathological analysis to help disentangle the effects of comorbid disorders to refine diagnosis, prognosis and better understand neurodegenerative disorders.
Collapse
Affiliation(s)
- Karl Li
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Tanweer Rashid
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jinqi Li
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Nicolas Honnorat
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Anoop Benet Nirmala
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Elyas Fadaee
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Di Wang
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sokratis Charisis
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hangfan Liu
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Crystal Franklin
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Mallory Maybrier
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Haritha Katragadda
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Leen Abazid
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Vinutha Ganapathy
- Department of Neurology, University of Texas Health Science Center, San Antonio, TX, USA
| | | | - Pradeepthi Badugu
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Eliana Vasquez
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Leigh Solano
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Geoffrey Clarke
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Gladys Maestre
- Department of Neuroscience, School of Medicine, University of Texas Rio Grande Valley, Harlingen, TX, USA
- Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Tim Richardson
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jamie Walker
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter T. Fox
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kevin Bieniek
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Pathology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Mohamad Habes
- Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
20
|
Ji Y, Zheng K, Li S, Ren C, Shen Y, Tian L, Zhu H, Zhou Z, Jiang Y. Insight into the potential role of ferroptosis in neurodegenerative diseases. Front Cell Neurosci 2022; 16:1005182. [PMID: 36385946 PMCID: PMC9647641 DOI: 10.3389/fncel.2022.1005182] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/14/2022] [Indexed: 11/30/2022] Open
Abstract
Ferroptosis is a newly discovered way of programmed cell death, mainly caused by the accumulation of iron-dependent lipid peroxides in cells, which is morphologically, biochemically and genetically different from the previously reported apoptosis, necrosis and autophagy. Studies have found that ferroptosis plays a key role in the occurrence and development of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease and vascular dementia, which suggest that ferroptosis may be involved in regulating the progression of neurodegenerative diseases. At present, on the underlying mechanism of ferroptosis in neurodegenerative diseases is still unclear, and relevant research is urgently needed to clarify the regulatory mechanism and provide the possibility for the development of agents targeting ferroptosis. This review focused on the regulatory mechanism of ferroptosis and its various effects in neurodegenerative diseases, in order to provide reference for the research on ferroptosis in neurodegenerative diseases.
Collapse
Affiliation(s)
- Yingying Ji
- The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
| | - Kai Zheng
- The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
| | - Shiming Li
- The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
| | - Caili Ren
- The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
| | - Ying Shen
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lin Tian
- The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
| | - Haohao Zhu
- The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
- *Correspondence: Haohao Zhu
| | - Zhenhe Zhou
- The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
- Zhenhe Zhou
| | - Ying Jiang
- The Affiliated Wuxi Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
- Ying Jiang
| |
Collapse
|
21
|
Rubinski A, Franzmeier N, Dewenter A, Luan Y, Smith R, Strandberg O, Ossenkoppele R, Dichgans M, Hansson O, Ewers M. Higher levels of myelin are associated with higher resistance against tau pathology in Alzheimer's disease. Alzheimers Res Ther 2022; 14:139. [PMID: 36153607 PMCID: PMC9508747 DOI: 10.1186/s13195-022-01074-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/28/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND In Alzheimer's disease (AD), fibrillar tau initially occurs locally and progresses preferentially between closely connected regions. However, the underlying sources of regional vulnerability to tau pathology remain unclear. Previous brain-autopsy findings suggest that the myelin levels-which differ substantially between white matter tracts in the brain-are a key modulating factor of region-specific susceptibility to tau deposition. Here, we investigated whether myelination differences between fiber tracts of the human connectome are predictive of the interregional spreading of tau pathology in AD. METHODS We included two independently recruited samples consisting of amyloid-PET-positive asymptomatic and symptomatic elderly individuals, in whom tau-PET was obtained at baseline (ADNI: n = 275; BioFINDER-1: n = 102) and longitudinally in a subset (ADNI: n = 123, mean FU = 1.53 [0.69-3.95] years; BioFINDER-1: n = 39, mean FU = 1.87 [1.21-2.78] years). We constructed MRI templates of the myelin water fraction (MWF) in 200 gray matter ROIs and connecting fiber tracts obtained from adult cognitively normal participants. Using the same 200 ROI brain-parcellation atlas, we obtained tau-PET ROI values from each individual in ADNI and BioFINDER-1. In a spatial regression analysis, we first tested the association between cortical myelin and group-average tau-PET signal in the amyloid-positive and control groups. Secondly, employing a previously established approach of modeling tau-PET spreading based on functional connectivity between ROIs, we estimated in a linear regression analysis, whether the level of fiber-tract myelin modulates the association between functional connectivity and longitudinal tau-PET spreading (i.e., covariance) between ROIs. RESULTS We found that higher myelinated cortical regions show lower tau-PET uptake (ADNI: rho = - 0.267, p < 0.001; BioFINDER-1: rho = - 0.175, p = 0.013). Fiber-tract myelin levels modulated the association between functional connectivity and tau-PET spreading, such that at higher levels of fiber-tract myelin, the association between stronger connectivity and higher covariance of tau-PET between the connected ROIs was attenuated (interaction fiber-tract myelin × functional connectivity: ADNI: β = - 0.185, p < 0.001; BioFINDER-1: β = - 0.166, p < 0.001). CONCLUSION Higher levels of myelin are associated with lower susceptibility of the connected regions to accumulate fibrillar tau. These results enhance our understanding of brain substrates that explain regional variation in tau accumulation and encourage future studies to investigate potential underlying mechanisms.
Collapse
Affiliation(s)
- Anna Rubinski
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Anna Dewenter
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Ying Luan
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Ruben Smith
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Michael Ewers
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| |
Collapse
|
22
|
O’Neill E, Mela V, Gaban AS, Bechet S, McGrath A, Walsh A, McIntosh A, Lynch MA. Sex-Related Microglial Perturbation Is Related to Mitochondrial Changes in a Model of Alzheimer’s Disease. Front Cell Neurosci 2022; 16:939830. [PMID: 35875349 PMCID: PMC9297004 DOI: 10.3389/fncel.2022.939830] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/09/2022] [Indexed: 11/29/2022] Open
Abstract
Many studies implicate microglia in the pathogenesis of Alzheimer’s disease (AD) but precisely how these cells make their impact has not been determined to date. One contributory factor is likely to be the enhanced production of inflammatory mediators and it is now known that microglia with this secretory phenotype exhibit other adaptations including in their morphology, function, and metabolism. AD, like many neurological disorders, demonstrates a sex bias and recent evidence indicates that the sexual dimorphism in microglial function, which has been recognized for many years in early development, persists into adulthood and aging. Here, we demonstrate sex-related differences in microglia from post mortem tissue of male and female AD patients and a marked increase in the number of dystrophic and rod-shaped microglia in tissue from female AD patients compared with males. Furthermore, there was an increase in iron-laden microglia in tissue from female AD patients and this has been reported to reflect mitochondrial changes. To address this further, we assessed changes in microglia from male and female APP/PS1 mice and demonstrate that iron accumulation in microglia is increased to a greater extent in tissue prepared from females compared with males. This was associated with altered expression of genes coding for proteins that modulate mitochondrial function. The findings suggest that sex-related differences in the severity and perhaps incidence of AD may, at least in part, arise from sexual dimorphism in microglia.
Collapse
|
23
|
Cerebral Iron Deposition in Neurodegeneration. Biomolecules 2022; 12:biom12050714. [PMID: 35625641 PMCID: PMC9138489 DOI: 10.3390/biom12050714] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Disruption of cerebral iron regulation appears to have a role in aging and in the pathogenesis of various neurodegenerative disorders. Possible unfavorable impacts of iron accumulation include reactive oxygen species generation, induction of ferroptosis, and acceleration of inflammatory changes. Whole-brain iron-sensitive magnetic resonance imaging (MRI) techniques allow the examination of macroscopic patterns of brain iron deposits in vivo, while modern analytical methods ex vivo enable the determination of metal-specific content inside individual cell-types, sometimes also within specific cellular compartments. The present review summarizes the whole brain, cellular, and subcellular patterns of iron accumulation in neurodegenerative diseases of genetic and sporadic origin. We also provide an update on mechanisms, biomarkers, and effects of brain iron accumulation in these disorders, focusing on recent publications. In Parkinson’s disease, Friedreich’s disease, and several disorders within the neurodegeneration with brain iron accumulation group, there is a focal siderosis, typically in regions with the most pronounced neuropathological changes. The second group of disorders including multiple sclerosis, Alzheimer’s disease, and amyotrophic lateral sclerosis shows iron accumulation in the globus pallidus, caudate, and putamen, and in specific cortical regions. Yet, other disorders such as aceruloplasminemia, neuroferritinopathy, or Wilson disease manifest with diffuse iron accumulation in the deep gray matter in a pattern comparable to or even more extensive than that observed during normal aging. On the microscopic level, brain iron deposits are present mostly in dystrophic microglia variably accompanied by iron-laden macrophages and in astrocytes, implicating a role of inflammatory changes and blood–brain barrier disturbance in iron accumulation. Options and potential benefits of iron reducing strategies in neurodegeneration are discussed. Future research investigating whether genetic predispositions play a role in brain Fe accumulation is necessary. If confirmed, the prevention of further brain Fe uptake in individuals at risk may be key for preventing neurodegenerative disorders.
Collapse
|
24
|
Holz TG, Kunzler FA, Carra Forte G, Miranda Difini JP, Bernardi Soder R, Watte G, Hochhegger B. In vivo brain iron concentration in healthy individuals at 3.0 T magnetic resonance imaging: a prospective cross-sectional study. Br J Radiol 2022; 95:20210809. [PMID: 35119909 PMCID: PMC10993970 DOI: 10.1259/bjr.20210809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/01/2021] [Accepted: 01/17/2022] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To quantify iron deposits in the basal ganglia and to evaluate its relation to age, sex, body mass index and brain laterality. METHODS Prospective observational study. Data were collected from the patients' electronic medical records. The concentration of iron deposits in the brain was assessed using whole-brain MRI at 3.0 Tesla. RESULTS 138 participants were selected, 69.6% were female and the mean age was 47 ± 19 years. The κ coefficient was very strong (k = 0.92, p < 0.001). Age showed a moderate correlation between iron deposits in the caudate and putamen nuclei, on both right and left sides. In overall and right-handed individuals, a significantly higher iron concentration was observed on the left side for the caudate nucleus, putamen, thalamus, globus pallidus, and centrum semiovale, and for left-handed individuals, it was also observed in the left side-for the putamen and centrum semiovale. A weak correlation was shown between body mass index and left and right substantia nigra, left caudate nuclei, left putamen and right globus pallidus. CONCLUSION Our results showed a significantly higher iron deposit on the left side in most brain regions. In addition, the body mass index may also be related to iron overload, especially in the caudate nucleus. ADVANCES IN KNOWLEDGE Brain iron deposits may be normal, owing to aging, or be pathological, such as neurodegeneration. Thus, it is important to know how much is expected of iron deposition in the brain of healthy populations.
Collapse
Affiliation(s)
- Tiago Garcia Holz
- School of Medicine, Graduate Program in Medicine and Health
Sciences, Pontifical Catholic University of Rio Grande do
Sul, Porto Alegre,
Brazil
| | - Felipe Augusto Kunzler
- School of Medicine, Graduate Program in Medicine and Health
Sciences, Pontifical Catholic University of Rio Grande do
Sul, Porto Alegre,
Brazil
| | - Gabriele Carra Forte
- School of Medicine, Graduate Program in Medicine and Health
Sciences, Pontifical Catholic University of Rio Grande do
Sul, Porto Alegre,
Brazil
| | - João Pedro Miranda Difini
- School of Medicine, Graduate Program in Medicine and Health
Sciences, Pontifical Catholic University of Rio Grande do
Sul, Porto Alegre,
Brazil
| | - Ricardo Bernardi Soder
- School of Medicine, Graduate Program in Medicine and Health
Sciences, Pontifical Catholic University of Rio Grande do
Sul, Porto Alegre,
Brazil
| | - Guilherme Watte
- School of Medicine, Graduate Program in Medicine and Health
Sciences, Pontifical Catholic University of Rio Grande do
Sul, Porto Alegre,
Brazil
| | - Bruno Hochhegger
- School of Medicine, Graduate Program in Medicine and Health
Sciences, Pontifical Catholic University of Rio Grande do
Sul, Porto Alegre,
Brazil
| |
Collapse
|
25
|
Cerebrospinal fluid tau levels are associated with abnormal neuronal plasticity markers in Alzheimer's disease. Mol Neurodegener 2022; 17:27. [PMID: 35346299 PMCID: PMC8962234 DOI: 10.1186/s13024-022-00521-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/13/2022] [Indexed: 12/15/2022] Open
Abstract
Background Increased total tau (t-tau) in cerebrospinal fluid (CSF) is a key characteristic of Alzheimer’s disease (AD) and is considered to result from neurodegeneration. T-tau levels, however, can be increased in very early disease stages, when neurodegeneration is limited, and can be normal in advanced disease stages. This suggests that t-tau levels may be driven by other mechanisms as well. Because tau pathophysiology is emerging as treatment target for AD, we aimed to clarify molecular processes associated with CSF t-tau levels. Methods We performed a proteomic, genomic, and imaging study in 1380 individuals with AD, in the preclinical, prodromal, and mild dementia stage, and 380 controls from the Alzheimer’s Disease Neuroimaging Initiative and EMIF-AD Multimodality Biomarker Discovery study. Results We found that, relative to controls, AD individuals with increased t-tau had increased CSF concentrations of over 400 proteins enriched for neuronal plasticity processes. In contrast, AD individuals with normal t-tau had decreased levels of these plasticity proteins and showed increased concentrations of proteins indicative of blood–brain barrier and blood-CSF barrier dysfunction, relative to controls. The distinct proteomic profiles were already present in the preclinical AD stage and persisted in prodromal and dementia stages implying that they reflect disease traits rather than disease states. Dysregulated plasticity proteins were associated with SUZ12 and REST signaling, suggesting aberrant gene repression. GWAS analyses contrasting AD individuals with and without increased t-tau highlighted several genes involved in the regulation of gene expression. Targeted analyses of SNP rs9877502 in GMNC, associated with t-tau levels previously, correlated in individuals with AD with CSF concentrations of 591 plasticity associated proteins. The number of APOE-e4 alleles, however, was not associated with the concentration of plasticity related proteins. Conclusions CSF t-tau levels in AD are associated with altered levels of proteins involved in neuronal plasticity and blood–brain and blood-CSF barrier dysfunction. Future trials may need to stratify on CSF t-tau status, as AD individuals with increased t-tau and normal t-tau are likely to respond differently to treatment, given their opposite CSF proteomic profiles. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-022-00521-3.
Collapse
|
26
|
Tisdall MD, Ohm DT, Lobrovich R, Das SR, Mizsei G, Prabhakaran K, Ittyerah R, Lim S, McMillan CT, Wolk DA, Gee J, Trojanowski JQ, Lee EB, Detre JA, Yushkevich P, Grossman M, Irwin DJ. Ex vivo MRI and histopathology detect novel iron-rich cortical inflammation in frontotemporal lobar degeneration with tau versus TDP-43 pathology. Neuroimage Clin 2022; 33:102913. [PMID: 34952351 PMCID: PMC8715243 DOI: 10.1016/j.nicl.2021.102913] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/28/2021] [Accepted: 12/08/2021] [Indexed: 02/08/2023]
Abstract
Comparative study of whole-hemisphere ex vivo T2*-weighted MRI and histopathology. Sample of FTLD-Tau and FTLD-TDP subtypes with reference to healthy and AD brain. Novel focal upper cortical-layer iron-rich pathology distinguishes FTLD-TDP from clinically-similar FTLD-Tau and AD. Distinct novel iron-rich FTLD-Tau pathology in mid-to-deep cortical-layers and WM. T2*-weighted MRI signatures offer in vivo biomarker targets for FTLD proteinopathy.
Frontotemporal lobar degeneration (FTLD) is a heterogeneous spectrum of age-associated neurodegenerative diseases that include two main pathologic categories of tau (FTLD-Tau) and TDP-43 (FTLD-TDP) proteinopathies. These distinct proteinopathies are often clinically indistinguishable during life, posing a major obstacle for diagnosis and emerging therapeutic trials tailored to disease-specific mechanisms. Moreover, MRI-derived measures have had limited success to date discriminating between FTLD-Tau or FTLD-TDP. T2*-weighted (T2*w) ex vivo MRI has previously been shown to be sensitive to non-heme iron in healthy intracortical lamination and myelin, and to pathological iron deposits in amyloid-beta plaques and activated microglia in Alzheimer’s disease neuropathologic change (ADNC). However, an integrated, ex vivo MRI and histopathology approach is understudied in FTLD. We apply joint, whole-hemisphere ex vivo MRI at 7 T and histopathology to the study autopsy-confirmed FTLD-Tau (n = 4) and FTLD-TDP (n = 3), relative to ADNC disease-control brains with antemortem clinical symptoms of frontotemporal dementia (n = 2), and an age-matched healthy control. We detect distinct laminar patterns of novel iron-laden glial pathology in both FTLD-Tau and FTLD-TDP brains. We find iron-positive ameboid and hypertrophic microglia and astrocytes largely in deeper GM and adjacent WM in FTLD-Tau. In contrast, FTLD-TDP presents prominent superficial cortical layer iron reactivity in astrocytic processes enveloping small blood vessels with limited involvement of adjacent WM, as well as more diffuse distribution of punctate iron-rich dystrophic microglial processes across all GM lamina. This integrated MRI/histopathology approach reveals ex vivo MRI features that are consistent with these pathological observations distinguishing FTLD-Tau and FTLD-TDP subtypes, including prominent irregular hypointense signal in deeper cortex in FTLD-Tau whereas FTLD-TDP showed upper cortical layer hypointense bands and diffuse cortical speckling. Moreover, differences in adjacent WM degeneration and iron-rich gliosis on histology between FTLD-Tau and FTLD-TDP were also readily apparent on MRI as hyperintense signal and irregular areas of hypointensity, respectively that were more prominent in FTLD-Tau compared to FTLD-TDP. These unique histopathological and radiographic features were distinct from healthy control and ADNC brains, suggesting that iron-sensitive T2*w MRI, adapted to in vivo application at sufficient resolution, may eventually offer an opportunity to improve antemortem diagnosis of FTLD proteinopathies using tissue-validated methods.
Collapse
Affiliation(s)
- M Dylan Tisdall
- Radiology, Perelman School of Medicine, University of Pennsylvania, United States.
| | - Daniel T Ohm
- Neurology, Perelman School of Medicine, University of Pennsylvania, United States
| | - Rebecca Lobrovich
- Neurology, Perelman School of Medicine, University of Pennsylvania, United States
| | - Sandhitsu R Das
- Neurology, Perelman School of Medicine, University of Pennsylvania, United States
| | - Gabor Mizsei
- Radiology, Perelman School of Medicine, University of Pennsylvania, United States
| | - Karthik Prabhakaran
- Neurology, Perelman School of Medicine, University of Pennsylvania, United States
| | - Ranjit Ittyerah
- Radiology, Perelman School of Medicine, University of Pennsylvania, United States
| | - Sydney Lim
- Radiology, Perelman School of Medicine, University of Pennsylvania, United States
| | - Corey T McMillan
- Neurology, Perelman School of Medicine, University of Pennsylvania, United States
| | - David A Wolk
- Neurology, Perelman School of Medicine, University of Pennsylvania, United States
| | - James Gee
- Radiology, Perelman School of Medicine, University of Pennsylvania, United States
| | - John Q Trojanowski
- Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, United States
| | - Edward B Lee
- Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, United States
| | - John A Detre
- Radiology, Perelman School of Medicine, University of Pennsylvania, United States; Neurology, Perelman School of Medicine, University of Pennsylvania, United States
| | - Paul Yushkevich
- Radiology, Perelman School of Medicine, University of Pennsylvania, United States
| | - Murray Grossman
- Neurology, Perelman School of Medicine, University of Pennsylvania, United States
| | - David J Irwin
- Neurology, Perelman School of Medicine, University of Pennsylvania, United States; Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, United States.
| |
Collapse
|
27
|
Chourrout M, Roux M, Boisvert C, Gislard C, Legland D, Arganda-Carreras I, Olivier C, Peyrin F, Boutin H, Rama N, Baron T, Meyronet D, Brun E, Rositi H, Wiart M, Chauveau F. Brain virtual histology with X-ray phase-contrast tomography Part II:3D morphologies of amyloid- β plaques in Alzheimer's disease models. BIOMEDICAL OPTICS EXPRESS 2022; 13:1640-1653. [PMID: 35414980 PMCID: PMC8973161 DOI: 10.1364/boe.438890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 05/15/2023]
Abstract
While numerous transgenic mouse strains have been produced to model the formation of amyloid-β (Aβ) plaques in the brain, efficient methods for whole-brain 3D analysis of Aβ deposits have to be validated and standardized. Moreover, routine immunohistochemistry performed on brain slices precludes any shape analysis of Aβ plaques, or require complex procedures for serial acquisition and reconstruction. The present study shows how in-line (propagation-based) X-ray phase-contrast tomography (XPCT) combined with ethanol-induced brain sample dehydration enables hippocampus-wide detection and morphometric analysis of Aβ plaques. Performed in three distinct Alzheimer mouse strains, the proposed workflow identified differences in signal intensity and 3D shape parameters: 3xTg displayed a different type of Aβ plaques, with a larger volume and area, greater elongation, flatness and mean breadth, and more intense average signal than J20 and APP/PS1. As a label-free non-destructive technique, XPCT can be combined with standard immunohistochemistry. XPCT virtual histology could thus become instrumental in quantifying the 3D spreading and the morphological impact of seeding when studying prion-like properties of Aβ aggregates in animal models of Alzheimer's disease. This is Part II of a series of two articles reporting the value of in-line XPCT for virtual histology of the brain; Part I shows how in-line XPCT enables 3D myelin mapping in the whole rodent brain and in human autopsy brain tissue.
Collapse
Affiliation(s)
- Matthieu Chourrout
- Univ. Lyon, Lyon Neuroscience Research Center, CNRS UMR5292, Inserm U1028, Université Claude Bernard Lyon 1, Lyon, France
| | - Margaux Roux
- Univ. Lyon, Lyon Neuroscience Research Center, CNRS UMR5292, Inserm U1028, Université Claude Bernard Lyon 1, Lyon, France
| | - Carlie Boisvert
- Univ. Lyon, Lyon Neuroscience Research Center, CNRS UMR5292, Inserm U1028, Université Claude Bernard Lyon 1, Lyon, France
- Current affiliation: Faculty of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ontario, Canada
| | - Coralie Gislard
- Univ. Lyon, Lyon Neuroscience Research Center, CNRS UMR5292, Inserm U1028, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Ignacio Arganda-Carreras
- University of the Basque Country (UPV/EHU), San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Donostia International Physics Center (DIPC), San Sebastian, Spain
| | - Cécile Olivier
- Univ. Lyon, CREATIS; CNRS UMR5220; INSERM U1044; INSA-Lyon; Univ. Lyon 1, Lyon, France
| | - Françoise Peyrin
- Univ. Lyon, CREATIS; CNRS UMR5220; INSERM U1044; INSA-Lyon; Univ. Lyon 1, Lyon, France
| | - Hervé Boutin
- Univ. Manchester, Faculty of Biology Medicine and Health, Wolfson Molecular Imaging Centre, Manchester, UK
| | - Nicolas Rama
- Univ. Lyon, CRCL; INSERM U1052; CNRS UMR5286; Univ. Lyon 1; Centre Léon Bérard, Lyon, France
| | | | | | - Emmanuel Brun
- Univ. Grenoble Alpes, Inserm UA07 Strobe Grenoble, France
| | - Hugo Rositi
- Univ. Clermont Auvergne, Institut Pascal; CNRS UMR 6602; SIGMA Clermont, Clermont-Ferrand, France
| | - Marlène Wiart
- Univ. Lyon, CarMeN Laboratory; INSERM U1060; INRA U1397; Hospices Civils de Lyon, Lyon, France
- CNRS, Lyon, France
- These authors contributed equally to this work
| | - Fabien Chauveau
- Univ. Lyon, Lyon Neuroscience Research Center, CNRS UMR5292, Inserm U1028, Université Claude Bernard Lyon 1, Lyon, France
- CNRS, Lyon, France
- These authors contributed equally to this work
| |
Collapse
|
28
|
Majerníková N, den Dunnen WFA, Dolga AM. The Potential of Ferroptosis-Targeting Therapies for Alzheimer's Disease: From Mechanism to Transcriptomic Analysis. Front Aging Neurosci 2022; 13:745046. [PMID: 34987375 PMCID: PMC8721139 DOI: 10.3389/fnagi.2021.745046] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/18/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer’s disease (AD), the most common form of dementia, currently affects 40–50 million people worldwide. Despite the extensive research into amyloid β (Aβ) deposition and tau protein hyperphosphorylation (p-tau), an effective treatment to stop or slow down the progression of neurodegeneration is missing. Emerging evidence suggests that ferroptosis, an iron-dependent and lipid peroxidation-driven type of programmed cell death, contributes to neurodegeneration in AD. Therefore, how to intervene against ferroptosis in the context of AD has become one of the questions addressed by studies aiming to develop novel therapeutic strategies. However, the underlying molecular mechanism of ferroptosis in AD, when ferroptosis occurs in the disease course, and which ferroptosis-related genes are differentially expressed in AD remains to be established. In this review, we summarize the current knowledge on cell mechanisms involved in ferroptosis, we discuss how these processes relate to AD, and we analyze which ferroptosis-related genes are differentially expressed in AD brain dependant on cell type, disease progression and gender. In addition, we point out the existing targets for therapeutic options to prevent ferroptosis in AD. Future studies should focus on developing new tools able to demonstrate where and when cells undergo ferroptosis in AD brain and build more translatable AD models for identifying anti-ferroptotic agents able to slow down neurodegeneration.
Collapse
Affiliation(s)
- Nad'a Majerníková
- Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, Netherlands.,Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands.,Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Wilfred F A den Dunnen
- Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands.,Research Institute Brain and Cognition, Molecular Neuroscience and Aging Research (MOLAR), University Medical Centre Groningen, Groningen, Netherlands
| | - Amalia M Dolga
- Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, Netherlands.,Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| |
Collapse
|
29
|
Folarin OR, Olopade FE, Olopade JO. Essential Metals in the Brain and the Application of Laser Ablation-Inductively Coupled Plasma-Mass Spectrometry for their Detection. Niger J Physiol Sci 2021; 36:123-147. [PMID: 35947740 DOI: 10.54548/njps.v36i2.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 06/15/2023]
Abstract
Metals are natural component of the ecosystem present throughout the layers of atmosphere; their abundant expression in the brain indicates their importance in the central nervous system (CNS). Within the brain tissue, their distribution is highly compartmentalized, the pattern of which is determined by their primary roles. Bio-imaging of the brain to reveal spatial distribution of metals within specific regions has provided a unique understanding of brain biochemistry and architecture, linking both the structures and the functions through several metal mediated activities. Bioavailability of essential trace metal is needed for normal brain function. However, disrupted metal homeostasis can influence several biochemical pathways in different fields of metabolism and cause characteristic neurological disorders with a typical disease process usually linked with aberrant metal accumulations. In this review we give a brief overview of roles of key essential metals (Iron, Copper and Zinc) including their molecular mechanisms and bio-distribution in the brain as well as their possible involvement in the pathogenesis of related neurodegenerative diseases. In addition, we also reviewed recent applications of Laser Ablation Inductively Couple Plasma Mass Spectrophotometry (LA-ICP-MS) in the detection of both toxic and essential metal dyshomeostasis in neuroscience research and other related brain diseases.
Collapse
|
30
|
Pini L, Wennberg AM, Salvalaggio A, Vallesi A, Pievani M, Corbetta M. Breakdown of specific functional brain networks in clinical variants of Alzheimer's disease. Ageing Res Rev 2021; 72:101482. [PMID: 34606986 DOI: 10.1016/j.arr.2021.101482] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is characterized by different clinical entities. Although AD phenotypes share a common molecular substrate (i.e., amyloid beta and tau accumulation), several clinicopathological differences exist. Brain functional networks might provide a macro-scale scaffolding to explain this heterogeneity. In this review, we summarize the evidence linking different large-scale functional network abnormalities to distinct AD phenotypes. Specifically, executive deficits in early-onset AD link with the dysfunction of networks that support sustained attention and executive functions. Posterior cortical atrophy relates to the breakdown of visual and dorsal attentional circuits, while the primary progressive aphasia variant of AD may be associated with the dysfunction of the left-lateralized language network. Additionally, network abnormalities might provide in vivo signatures for distinguishing proteinopathies that mimic AD, such as TAR DNA binding protein 43 related pathologies. These network differences vis-a-vis clinical syndromes are more evident in the earliest stage of AD. Finally, we discuss how these findings might pave the way for new tailored interventions targeting the most vulnerable brain circuit at the optimal time window to maximize clinical benefits.
Collapse
|
31
|
Radulović S, Sunkara S, Maurer C, Leitinger G. Digging Deeper: Advancements in Visualization of Inhibitory Synapses in Neurodegenerative Disorders. Int J Mol Sci 2021; 22:12470. [PMID: 34830352 PMCID: PMC8623765 DOI: 10.3390/ijms222212470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/02/2022] Open
Abstract
Recent research has provided strong evidence that neurodegeneration may develop from an imbalance between synaptic structural components in the brain. Lately, inhibitory synapses communicating via the neurotransmitters GABA or glycine have come to the center of attention. Increasing evidence suggests that imbalance in the structural composition of inhibitory synapses affect deeply the ability of neurons to communicate effectively over synaptic connections. Progressive failure of synaptic plasticity and memory are thus hallmarks of neurodegenerative diseases. In order to prove that structural changes at synapses contribute to neurodegeneration, we need to visualize single-molecule interactions at synaptic sites in an exact spatial and time frame. This visualization has been restricted in terms of spatial and temporal resolution. New developments in electron microscopy and super-resolution microscopy have improved spatial and time resolution tremendously, opening up numerous possibilities. Here we critically review current and recently developed methods for high-resolution visualization of inhibitory synapses in the context of neurodegenerative diseases. We present advantages, strengths, weaknesses, and current limitations for selected methods in research, as well as present a future perspective. A range of new options has become available that will soon help understand the involvement of inhibitory synapses in neurodegenerative disorders.
Collapse
Affiliation(s)
- Snježana Radulović
- Gottfried Schatz Research Center, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria; (S.R.); (S.S.)
| | - Sowmya Sunkara
- Gottfried Schatz Research Center, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria; (S.R.); (S.S.)
| | - Christa Maurer
- Gottfried Schatz Research Center, Division of Macroscopic and Clinical Anatomy, Medical University of Graz, 8010 Graz, Austria;
| | - Gerd Leitinger
- Gottfried Schatz Research Center, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria; (S.R.); (S.S.)
| |
Collapse
|
32
|
Veale T, Malone IB, Poole T, Parker TD, Slattery CF, Paterson RW, Foulkes AJM, Thomas DL, Schott JM, Zhang H, Fox NC, Cash DM. Loss and dispersion of superficial white matter in Alzheimer's disease: a diffusion MRI study. Brain Commun 2021; 3:fcab272. [PMID: 34859218 PMCID: PMC8633427 DOI: 10.1093/braincomms/fcab272] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/24/2021] [Accepted: 10/18/2021] [Indexed: 11/22/2022] Open
Abstract
Pathological cerebral white matter changes in Alzheimer's disease have been shown using diffusion tensor imaging. Superficial white matter changes are relatively understudied despite their importance in cortico-cortical connections. Measuring superficial white matter degeneration using diffusion tensor imaging is challenging due to its complex organizational structure and proximity to the cortex. To overcome this, we investigated diffusion MRI changes in young-onset Alzheimer's disease using standard diffusion tensor imaging and Neurite Orientation Dispersion and Density Imaging to distinguish between disease-related changes that are degenerative (e.g. loss of myelinated fibres) and organizational (e.g. increased fibre dispersion). Twenty-nine young-onset Alzheimer's disease patients and 22 healthy controls had both single-shell and multi-shell diffusion MRI. We calculated fractional anisotropy, mean diffusivity, neurite density index, orientation dispersion index and tissue fraction (1-free water fraction). Diffusion metrics were sampled in 15 a priori regions of interest at four points along the cortical profile: cortical grey matter, grey/white boundary, superficial white matter (1 mm below grey/white boundary) and superficial/deeper white matter (2 mm below grey/white boundary). To estimate cross-sectional group differences, we used average marginal effects from linear mixed effect models of participants' diffusion metrics along the cortical profile. The superficial white matter of young-onset Alzheimer's disease individuals had lower neurite density index compared to controls in five regions (superior and inferior parietal, precuneus, entorhinal and parahippocampus) (all P < 0.05), and higher orientation dispersion index in three regions (fusiform, entorhinal and parahippocampus) (all P < 0.05). Young-onset Alzheimer's disease individuals had lower fractional anisotropy in the entorhinal and parahippocampus regions (both P < 0.05) and higher fractional anisotropy within the postcentral region (P < 0.05). Mean diffusivity was higher in the young-onset Alzheimer's disease group in the parahippocampal region (P < 0.05) and lower in the postcentral, precentral and superior temporal regions (all P < 0.05). In the overlying grey matter, disease-related changes were largely consistent with superficial white matter findings when using neurite density index and fractional anisotropy, but appeared at odds with orientation dispersion and mean diffusivity. Tissue fraction was significantly lower across all grey matter regions in young-onset Alzheimer's disease individuals (all P < 0.001) but group differences reduced in magnitude and coverage when moving towards the superficial white matter. These results show that microstructural changes occur within superficial white matter and along the cortical profile in individuals with young-onset Alzheimer's disease. Lower neurite density and higher orientation dispersion suggests underlying fibres undergo neurodegeneration and organizational changes, two effects previously indiscernible using standard diffusion tensor metrics in superficial white matter.
Collapse
Affiliation(s)
- Thomas Veale
- The Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Ian B Malone
- The Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Teresa Poole
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK
| | - Thomas D Parker
- The Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Catherine F Slattery
- The Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Ross W Paterson
- The Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Alexander J M Foulkes
- The Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - David L Thomas
- The Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, London, UK
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, UK
| | - Jonathan M Schott
- The Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Hui Zhang
- Department of Computer Science and Centre for Medical Image Computing, UCL, London, UK
| | - Nick C Fox
- The Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - David M Cash
- The Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| |
Collapse
|
33
|
Jakubowski H, Zioła-Frankowska A, Frankowski M, Perła-Kaján J, Refsum H, de Jager CA, Smith AD. B Vitamins Prevent Iron-Associated Brain Atrophy and Domain-Specific Effects of Iron, Copper, Aluminum, and Silicon on Cognition in Mild Cognitive Impairment. J Alzheimers Dis 2021; 84:1039-1055. [PMID: 34602484 PMCID: PMC8673493 DOI: 10.3233/jad-215085] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Metals, silicon, and homocysteine are linked to Alzheimer’s disease. B vitamin therapy lowers homocysteine and slows brain atrophy and cognitive decline in mild cognitive impairment (MCI). Objective: Examine metals and silicon as predictors of cognition/brain atrophy in MCI, their interaction with homocysteine and cysteine, and how B vitamins affect these relationships. Methods: MCI participants (n = 266, 77.6-year-old, 60.7% female) in VITACOG trial were randomized to receive daily folic acid (0.8 mg)/vitamin B12 (0.5 mg)/vitamin B6 (20 mg) (n = 133) or placebo for two years. At baseline and end-of-study, cranial MRIs were obtained from 168 participants, cognition was analyzed by neuropsychological tests, and serum iron, copper, arsenic, aluminum, and silicon quantified by inductively-coupled plasma mass spectrometry in 196 participants. Data were analyzed by bivariate and multiple regression. Results: Baseline iron, cysteine, and homocysteine were significantly associated with brain atrophy rate. Homocysteine effects on brain atrophy rate were modified by iron and cysteine. At baseline, iron, copper, aluminum, and silicon were significantly associated with one or more domains of cognition: semantic memory, verbal episodic memory, attention/processing speed, and executive function. At end-of-study, baseline iron, copper, aluminum, and silicon predicted cognition in at least one domain: semantic memory, verbal episodic memory, visuospatial episodic memory, attention/processing speed, and global cognition in the placebo but not the B vitamin group. Conclusion: Disparate effects of serum iron, copper, aluminum, silicon, and homocysteine on cognition and brain atrophy in MCI suggest that cognitive impairment is independent of brain atrophy. These factors showed domain-specific associations with cognition, which were abrogated by B vitamin therapy.
Collapse
Affiliation(s)
- Hieronim Jakubowski
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, International Center for Public Health, Rutgers University, Newark, NJ, USA.,Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland
| | - Anetta Zioła-Frankowska
- Department of Analytical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Poznań, Poland
| | - Marcin Frankowski
- Department of Analytical and Environmental Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Poznań, Poland
| | - Joanna Perła-Kaján
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Celeste A de Jager
- Oxford Project to Investigate Memory and Aging (OPTIMA), Department of Pharmacology, University of Oxford, Oxford, UK.,Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
| | - A David Smith
- Oxford Project to Investigate Memory and Aging (OPTIMA), Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
34
|
Jakaria M, Belaidi AA, Bush AI, Ayton S. Ferroptosis as a mechanism of neurodegeneration in Alzheimer's disease. J Neurochem 2021; 159:804-825. [PMID: 34553778 DOI: 10.1111/jnc.15519] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/07/2021] [Accepted: 09/14/2021] [Indexed: 01/19/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, with complex pathophysiology that is not fully understood. While β-amyloid plaque and neurofibrillary tangles define the pathology of the disease, the mechanism of neurodegeneration is uncertain. Ferroptosis is an iron-mediated programmed cell death mechanism characterised by phospholipid peroxidation that has been observed in clinical AD samples. This review will outline the growing molecular and clinical evidence implicating ferroptosis in the pathogenesis of AD, with implications for disease-modifying therapies.
Collapse
Affiliation(s)
- Md Jakaria
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Abdel Ali Belaidi
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
35
|
Lier J, Streit WJ, Bechmann I. Beyond Activation: Characterizing Microglial Functional Phenotypes. Cells 2021; 10:cells10092236. [PMID: 34571885 PMCID: PMC8464670 DOI: 10.3390/cells10092236] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/18/2021] [Accepted: 08/26/2021] [Indexed: 12/20/2022] Open
Abstract
Classically, the following three morphological states of microglia have been defined: ramified, amoeboid and phagocytic. While ramified cells were long regarded as “resting”, amoeboid and phagocytic microglia were viewed as “activated”. In aged human brains, a fourth, morphologically novel state has been described, i.e., dystrophic microglia, which are thought to be senescent cells. Since microglia are not replenished by blood-borne mononuclear cells under physiological circumstances, they seem to have an “expiration date” limiting their capacity to phagocytose and support neurons. Identifying factors that drive microglial aging may thus be helpful to delay the onset of neurodegenerative diseases, such as Alzheimer’s disease (AD). Recent progress in single-cell deep sequencing methods allowed for more refined differentiation and revealed regional-, age- and sex-dependent differences of the microglial population, and a growing number of studies demonstrate various expression profiles defining microglial subpopulations. Given the heterogeneity of pathologic states in the central nervous system, the need for accurately describing microglial morphology and expression patterns becomes increasingly important. Here, we review commonly used microglial markers and their fluctuations in expression in health and disease, with a focus on IBA1 low/negative microglia, which can be found in individuals with liver disease.
Collapse
Affiliation(s)
- Julia Lier
- Institute of Anatomy, University of Leipzig, 04109 Leipzig, Germany;
- Department of Neurology, University of Leipzig, 04109 Leipzig, Germany
- Correspondence:
| | - Wolfgang J. Streit
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32611, USA;
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, 04109 Leipzig, Germany;
| |
Collapse
|
36
|
Vachha B, Huang SY. MRI with ultrahigh field strength and high-performance gradients: challenges and opportunities for clinical neuroimaging at 7 T and beyond. Eur Radiol Exp 2021; 5:35. [PMID: 34435246 PMCID: PMC8387544 DOI: 10.1186/s41747-021-00216-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
Research in ultrahigh magnetic field strength combined with ultrahigh and ultrafast gradient technology has provided enormous gains in sensitivity, resolution, and contrast for neuroimaging. This article provides an overview of the technical advantages and challenges of performing clinical neuroimaging studies at ultrahigh magnetic field strength combined with ultrahigh and ultrafast gradient technology. Emerging clinical applications of 7-T MRI and state-of-the-art gradient systems equipped with up to 300 mT/m gradient strength are reviewed, and the impact and benefits of such advances to anatomical, structural and functional MRI are discussed in a variety of neurological conditions. Finally, an outlook and future directions for ultrahigh field MRI combined with ultrahigh and ultrafast gradient technology in neuroimaging are examined.
Collapse
Affiliation(s)
- Behroze Vachha
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Susie Y Huang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Room 2301, Charlestown, MA, 02129, USA.
| |
Collapse
|
37
|
Perosa V, Scherlek AA, Kozberg MG, Smith L, Westerling-Bui T, Auger CA, Vasylechko S, Greenberg SM, van Veluw SJ. Deep learning assisted quantitative assessment of histopathological markers of Alzheimer's disease and cerebral amyloid angiopathy. Acta Neuropathol Commun 2021; 9:141. [PMID: 34419154 PMCID: PMC8380352 DOI: 10.1186/s40478-021-01235-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/26/2021] [Indexed: 12/20/2022] Open
Abstract
Traditionally, analysis of neuropathological markers in neurodegenerative diseases has relied on visual assessments of stained sections. Resulting semiquantitative scores often vary between individual raters and research centers, limiting statistical approaches. To overcome these issues, we have developed six deep learning-based models, that identify some of the most characteristic markers of Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA). The deep learning-based models are trained to differentially detect parenchymal amyloid β (Aβ)-plaques, vascular Aβ-deposition, iron and calcium deposition, reactive astrocytes, microglia, as well as fibrin extravasation. The models were trained on digitized histopathological slides from brains of patients with AD and CAA, using a workflow that allows neuropathology experts to train convolutional neural networks (CNNs) on a cloud-based graphical interface. Validation of all models indicated a very good to excellent performance compared to three independent expert human raters. Furthermore, the Aβ and iron models were consistent with previously acquired semiquantitative scores in the same dataset and allowed the use of more complex statistical approaches. For example, linear mixed effects models could be used to confirm the previously described relationship between leptomeningeal CAA severity and cortical iron accumulation. A similar approach enabled us to explore the association between neuroinflammation and disparate Aβ pathologies. The presented workflow is easy for researchers with pathological expertise to implement and is customizable for additional histopathological markers. The implementation of deep learning-assisted analyses of histopathological slides is likely to promote standardization of the assessment of neuropathological markers across research centers, which will allow specific pathophysiological questions in neurodegenerative disease to be addressed in a harmonized way and on a larger scale.
Collapse
|
38
|
Bauer CE, Zachariou V, Seago E, Gold BT. White Matter Hyperintensity Volume and Location: Associations With WM Microstructure, Brain Iron, and Cerebral Perfusion. Front Aging Neurosci 2021; 13:617947. [PMID: 34290597 PMCID: PMC8287527 DOI: 10.3389/fnagi.2021.617947] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 05/31/2021] [Indexed: 11/29/2022] Open
Abstract
Cerebral white matter hyperintensities (WMHs) represent macrostructural brain damage associated with various etiologies. However, the relative contributions of various etiologies to WMH volume, as assessed via different neuroimaging measures, is not well-understood. Here, we explored associations between three potential early markers of white matter hyperintensity volume. Specifically, the unique variance in total and regional WMH volumes accounted for by white matter microstructure, brain iron concentration and cerebral blood flow (CBF) was assessed. Regional volumes explored were periventricular and deep regions. Eighty healthy older adults (ages 60–86) were scanned at 3 Tesla MRI using fluid-attenuated inversion recovery, diffusion tensor imaging (DTI), multi-echo gradient-recalled echo and pseudo-continuous arterial spin labeling sequences. In a stepwise regression model, DTI-based radial diffusivity accounted for significant variance in total WMH volume (adjusted R2 change = 0.136). In contrast, iron concentration (adjusted R2 change = 0.043) and CBF (adjusted R2 change = 0.027) made more modest improvements to the variance accounted for in total WMH volume. However, there was an interaction between iron concentration and location on WMH volume such that iron concentration predicted deep (p = 0.034) but not periventricular (p = 0.414) WMH volume. Our results suggest that WM microstructure may be a better predictor of WMH volume than either brain iron or CBF but also draws attention to the possibility that some early WMH markers may be location-specific.
Collapse
Affiliation(s)
- Christopher E Bauer
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Valentinos Zachariou
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Elayna Seago
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Brian T Gold
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
39
|
Ayton S, Portbury S, Kalinowski P, Agarwal P, Diouf I, Schneider JA, Morris MC, Bush AI. Regional brain iron associated with deterioration in Alzheimer's disease: A large cohort study and theoretical significance. Alzheimers Dement 2021; 17:1244-1256. [PMID: 33491917 PMCID: PMC9701539 DOI: 10.1002/alz.12282] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE This paper is a proposal for an update of the iron hypothesis of Alzheimer's disease (AD), based on large-scale emerging evidence. BACKGROUND Iron featured historically early in AD research efforts for its involvement in the amyloid and tau proteinopathies, APP processing, genetics, and one clinical trial, yet iron neurochemistry remains peripheral in mainstream AD research. Much of the effort investigating iron in AD has focused on the potential for iron to provoke the onset of disease, by promoting proteinopathy though increased protein expression, phosphorylation, and aggregation. NEW/UPDATED HYPOTHESIS We provide new evidence from a large post mortem cohort that brain iron levels within the normal range were associated with accelerated ante mortem disease progression in cases with underlying proteinopathic neuropathology. These results corroborate recent findings that argue for an additional downstream role for iron as an effector of neurodegeneration, acting independently of tau or amyloid pathologies. We hypothesize that the level of tissue iron is a trait that dictates the probability of neurodegeneration in AD by ferroptosis, a regulated cell death pathway that is initiated by signals such as glutathione depletion and lipid peroxidation. MAJOR CHALLENGES FOR THE HYPOTHESIS While clinical biomarkers of ferroptosis are still in discovery, the demonstration of additional ferroptotic correlates (genetic or biomarker derived) of disease progression is required to test this hypothesis. The genes implicated in familial AD are not known to influence ferroptosis, although recent reports on APP mutations and apolipoprotein E allele (APOE) have shown impact on cellular iron retention. Familial AD mutations will need to be tested for their impact on ferroptotic vulnerability. Ultimately, this hypothesis will be substantiated, or otherwise, by a clinical trial of an anti-ferroptotic/iron compound in AD patients. LINKAGE TO OTHER MAJOR THEORIES Iron has historically been linked to the amyloid and tau proteinopathies of AD. Tau, APP, and apoE have been implicated in physiological iron homeostasis in the brain. Iron is biochemically the origin of most chemical radicals generated in biochemistry and thus closely associated with the oxidative stress theory of AD. Iron accumulation is also a well-established consequence of aging and inflammation, which are major theories of disease pathogenesis.
Collapse
Affiliation(s)
- Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, Australia
| | - Stuart Portbury
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, Australia
| | - Pawel Kalinowski
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, Australia
| | - Puja Agarwal
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, USA
| | - Ibrahima Diouf
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, Australia
- CSIRO Health and Biosecurity, Australian E-Health Research Centre, Brisbane, Australia
| | - Julie A Schneider
- Rush Alzheimer Disease Center, Rush University Medical Center, Chicago, USA
| | - Martha Clare Morris
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, USA
| | - Ashley I. Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, and The University of Melbourne, Parkville, Australia
| |
Collapse
|
40
|
Zhang PF, Hu H, Tan L, Yu JT. Microglia Biomarkers in Alzheimer's Disease. Mol Neurobiol 2021; 58:3388-3404. [PMID: 33713018 DOI: 10.1007/s12035-021-02348-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 03/03/2021] [Indexed: 12/13/2022]
Abstract
Early detection and clinical diagnosis of Alzheimer's disease (AD) have become an extremely important link in the prevention and treatment of AD. Because of the occult onset, the diagnosis and treatment of AD based on clinical symptoms are increasingly challenged by current severe situations. Therefore, molecular diagnosis models based on early AD pathological markers have received more attention. Among the possible pathological mechanisms, microglia which are necessary for normal brain function are highly expected and have been continuously studied in various models. Several AD biomarkers already exist, but currently there is a paucity of specific and sensitive microglia biomarkers which can accurately measure preclinical AD. Bringing microglia biomarkers into the molecular diagnostic system which is based on fluid and neuroimaging will play an important role in future scientific research and clinical practice. Furthermore, developing novel, more specific, and sensitive microglia biomarkers will make it possible to pharmaceutically target chemical pathways that preserve beneficial microglial functions in response to AD pathology. This review discusses microglia biomarkers in the context of AD.
Collapse
Affiliation(s)
- Peng-Fei Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
41
|
Tuzzi E, Balla DZ, Loureiro JRA, Neumann M, Laske C, Pohmann R, Preische O, Scheffler K, Hagberg GE. Ultra-High Field MRI in Alzheimer's Disease: Effective Transverse Relaxation Rate and Quantitative Susceptibility Mapping of Human Brain In Vivo and Ex Vivo compared to Histology. J Alzheimers Dis 2021; 73:1481-1499. [PMID: 31958079 DOI: 10.3233/jad-190424] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. So far, diagnosis of AD is only unequivocally defined through postmortem histology. Amyloid plaques are a classical hallmark of AD and amyloid load is currently quantified by Positron Emission tomography (PET) in vivo. Ultra-high field magnetic resonance imaging (UHF-MRI) can potentially provide a non-invasive biomarker for AD by allowing imaging of pathological processes at a very-high spatial resolution. The first aim of this work was to reproduce the characteristic cortical pattern previously observed in vivo in AD patients using weighted-imaging at 7T. We extended these findings using quantitative susceptibility mapping (QSM) and quantification of the effective transverse relaxation rate (R2*) at 9.4T. The second aim was to investigate the origin of the contrast patterns observed in vivo in the cortex of AD patients at 9.4T by comparing quantitative UHF-MRI (9.4T and 14.1T) of postmortem samples with histology. We observed a distinctive cortical pattern in vivo in patients compared to healthy controls (HC), and these findings were confirmed ex vivo. Specifically, we found a close link between the signal changes detected by QSM in the AD sample at 14.1T and the distribution pattern of amyloid plaques in the histological sections of the same specimen. Our findings showed that QSM and R2* maps can distinguish AD from HC at UHF by detecting cortical alterations directly related to amyloid plaques in AD patients. Furthermore, we provided a method to quantify amyloid plaque load in AD patients at UHF non-invasively.
Collapse
Affiliation(s)
- Elisa Tuzzi
- Department for High Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany.,Department for Biomedical Magnetic Resonance, Eberhard Karl's University, Tübingen and University Hospital, Tübingen, Germany
| | - David Z Balla
- Department for Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Joana R A Loureiro
- Department for High Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany.,Department for Biomedical Magnetic Resonance, Eberhard Karl's University, Tübingen and University Hospital, Tübingen, Germany.,Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California, Los Angeles, CA, USA
| | - Manuela Neumann
- Department of Neuropathology, University Hospital, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE) Tübingen, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE) Tübingen, Germany.,Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Rolf Pohmann
- Department for High Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Oliver Preische
- German Center for Neurodegenerative Diseases (DZNE) Tübingen, Germany.,Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Klaus Scheffler
- Department for High Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany.,Department for Biomedical Magnetic Resonance, Eberhard Karl's University, Tübingen and University Hospital, Tübingen, Germany
| | - Gisela E Hagberg
- Department for High Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany.,Department for Biomedical Magnetic Resonance, Eberhard Karl's University, Tübingen and University Hospital, Tübingen, Germany
| |
Collapse
|
42
|
Lim D, Jeong JH, Song J. Lipocalin 2 regulates iron homeostasis, neuroinflammation, and insulin resistance in the brains of patients with dementia: Evidence from the current literature. CNS Neurosci Ther 2021; 27:883-894. [PMID: 33945675 PMCID: PMC8265939 DOI: 10.1111/cns.13653] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/13/2021] [Accepted: 04/17/2021] [Indexed: 12/24/2022] Open
Abstract
Dementia accompanied by memory loss is considered one of the most common neurodegenerative diseases worldwide, and its prevalence is gradually increasing. Known risk factors for dementia include genetic background, certain lifestyle and dietary patterns, smoking, iron overload, insulin resistance, and impaired glucose metabolism in the brain. Here, we review recent evidence on the regulatory role of lipocalin 2 (LCN2) in dementia from various perspectives. LCN2 is a neutrophil gelatinase-associated protein that influences diverse cellular processes, including the immune system, iron homeostasis, lipid metabolism, and inflammatory responses. Although its functions within the peripheral system are most widely recognized, recent findings have revealed links between LCN2 and central nervous system diseases, as well as novel roles for LCN2 in neurons and glia. Furthermore, LCN2 may modulate diverse pathological mechanisms involved in dementia. Taken together, LCN2 is a promising therapeutic target with which to address the neuropathology of dementia.
Collapse
Affiliation(s)
- Daejin Lim
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Korea
| | - Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Chonnam National University, Gwangju, Korea
| |
Collapse
|
43
|
Bulk M, van Harten T, Kenkhuis B, Inglese F, Hegeman I, van Duinen S, Ercan E, Magro-Checa C, Goeman J, Mawrin C, van Buchem M, Steup-Beekman G, Huizinga T, van der Weerd L, Ronen I. Quantitative susceptibility mapping in the thalamus and basal ganglia of systemic lupus erythematosus patients with neuropsychiatric complaints. Neuroimage Clin 2021; 30:102637. [PMID: 33812303 PMCID: PMC8053812 DOI: 10.1016/j.nicl.2021.102637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 11/30/2022]
Abstract
Systemic lupus erythematosus (SLE) is an auto-immune disease characterized by multi-organ involvement. Although uncommon, central nervous system involvement in SLE, termed neuropsychiatric SLE (NPSLE), is not an exception. Current knowledge on underlying pathogenic mechanisms is incomplete, however, neuroinflammation is thought to play a critical role. Evidence from neurodegenerative diseases and multiple sclerosis suggests that neuroinflammation is correlated with brain iron accumulation, making quantitative susceptibility mapping (QSM) a potential hallmark for neuroinflammation in vivo. This study assessed susceptibility values of the thalamus and basal ganglia in (NP)SLE patients and further investigated the in vivo findings with histological analyses of postmortem brain tissue derived from SLE patients. We used a 3T MRI scanner to acquire single-echo T2*-weighted images of 44 SLE patients and 20 age-matched healthy controls. Of the 44 patients with SLE, all had neuropsychiatric complaints, of which 29 were classified as non-NPSLE and 15 as NPSLE (seven as inflammatory NPSLE and eight as ischemic NPSLE). Mean susceptibility values of the thalamus, caudate nucleus, putamen, and globus pallidus were calculated. Formalin-fixed paraffin-embedded post-mortem brain tissue including the putamen and globus pallidus of three additional SLE patients was obtained and stained for iron, microglia and astrocytes. Susceptibility values of SLE patients and age-matched controls showed that iron levels in the thalamus and basal ganglia were not changed due to the disease. No subgroup of SLE showed higher susceptibility values. No correlation was found with disease activity or damage due to SLE. Histological examination of the post-mortem brain showed no increased iron accumulation. Our results suggest that neuroinflammation in NPSLE does not necessarily go hand in hand with iron accumulation, and that the inflammatory pathomechanism in SLE may differ from the one observed in neurodegenerative diseases and in multiple sclerosis.
Collapse
Affiliation(s)
- Marjolein Bulk
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thijs van Harten
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Boyd Kenkhuis
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands; Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Francesca Inglese
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ingrid Hegeman
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd van Duinen
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ece Ercan
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - César Magro-Checa
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands; Department of Rheumatology, Zuyderland Medical Center, Heerlen, The Netherlands
| | - Jelle Goeman
- Department of Medical Statistics, Leiden University Medical Center, Leiden, The Netherlands
| | - Christian Mawrin
- Department of Neuropathology, Otto-von-Guericke University, Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Mark van Buchem
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerda Steup-Beekman
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Louise van der Weerd
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands; Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Itamar Ronen
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
44
|
Qiu Y, She S, Zhang S, Wu F, Liang Q, Peng Y, Yuan H, Ning Y, Wu H, Huang R. Cortical myelin content mediates differences in affective temperaments. J Affect Disord 2021; 282:1263-1271. [PMID: 33601705 DOI: 10.1016/j.jad.2021.01.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/29/2020] [Accepted: 01/09/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND Affective temperaments are regarded as subclinical forms and precursors of mental disorders. It may serve as candidates to facilitate the diagnosis and prediction of mental disorders. Cortical myelination likely characterizes the neurodevelopment and the evolution of cognitive functions and reflects brain functional demand. However, little is known about the relationship between affective temperaments and myelin plasticity. This study aims to analyze the association between the affective temperaments and cortical myelin content (CMC) in human brain. METHODS We measured affective temperaments using the Temperament Evaluation of Memphis, Pisa, Paris and San Diego Autoquestionnaire (TEMPS-A) on 106 healthy adults and used the ratio of T1- and T2-weighted images as the proxy for CMC. Using the unsupervised k-means clustering algorithm, we classified the cortical gray matter into heavily, intermediately, and lightly myelinated regions. The correlation between affective temperaments and CMC was calculated separately for different myelinated regions. RESULTS Hyperthymic temperament correlated negatively with CMC in the heavily myelinated (right postcentral gyrus and bilateral precentral gyrus) and lightly myelinated (bilateral frontal and lateral temporal) regions. Cyclothymic temperament showed a downward parabola-like correlation with CMC across the heavily, intermediately, and lightly myel0inated areas of the bilateral parietal-temporal regions. LIMITATIONS The analysis was constrained to cortical regions. The results were obtained from healthy subjects and we did not acquired data from patients of affective disorder, which may compromise the generalizability of the present findings. CONCLUSION The findings suggest that hyperthymic and cyclothymic temperaments have a CMC basis in extensive brain regions.
Collapse
Affiliation(s)
- Yidan Qiu
- Key Laboratory of Brain, Cognition and Education Sciences (South China Normal University), Ministry of Education; School of Psychology, Center for Studies of Psychological Application; Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, China
| | - Shenglin She
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Shufei Zhang
- Key Laboratory of Brain, Cognition and Education Sciences (South China Normal University), Ministry of Education; School of Psychology, Center for Studies of Psychological Application; Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, China
| | - Fengchun Wu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Qunjun Liang
- Key Laboratory of Brain, Cognition and Education Sciences (South China Normal University), Ministry of Education; School of Psychology, Center for Studies of Psychological Application; Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, China
| | - Yongjun Peng
- Department of Medical Imaging, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, China
| | - Haishan Yuan
- Key Laboratory of Brain, Cognition and Education Sciences (South China Normal University), Ministry of Education; School of Psychology, Center for Studies of Psychological Application; Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, China
| | - Yuping Ning
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Huawang Wu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, 510370, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China.
| | - Ruiwang Huang
- Key Laboratory of Brain, Cognition and Education Sciences (South China Normal University), Ministry of Education; School of Psychology, Center for Studies of Psychological Application; Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
45
|
Ficiarà E, Boschi S, Ansari S, D'Agata F, Abollino O, Caroppo P, Di Fede G, Indaco A, Rainero I, Guiot C. Machine Learning Profiling of Alzheimer's Disease Patients Based on Current Cerebrospinal Fluid Markers and Iron Content in Biofluids. Front Aging Neurosci 2021; 13:607858. [PMID: 33692679 PMCID: PMC7937894 DOI: 10.3389/fnagi.2021.607858] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by a complex etiology that makes therapeutic strategies still not effective. A true understanding of key pathological mechanisms and new biomarkers are needed, to identify alternative disease-modifying therapies counteracting the disease progression. Iron is an essential element for brain metabolism and its imbalance is implicated in neurodegeneration, due to its potential neurotoxic effect. However, the role of iron in different stages of dementia is not clearly established. This study aimed to investigate the potential impact of iron both in cerebrospinal fluid (CSF) and in serum to improve early diagnosis and the related therapeutic possibility. In addition to standard clinical method to detect iron in serum, a precise quantification of total iron in CSF was performed using graphite-furnace atomic absorption spectrometry in patients affected by AD, mild cognitive impairment, frontotemporal dementia, and non-demented neurological controls. The application of machine learning techniques, such as clustering analysis and multiclassification algorithms, showed a new potential stratification of patients exploiting iron-related data. The results support the involvement of iron dysregulation and its potential interaction with biomarkers (Tau protein and Amyloid-beta) in the pathophysiology and progression of dementia.
Collapse
Affiliation(s)
- Eleonora Ficiarà
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Torino, Italy
| | - Silvia Boschi
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Torino, Italy.,Department NEUROFARBA, University of Firenze, Firenze, Italy
| | - Shoeb Ansari
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Torino, Italy
| | - Federico D'Agata
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Torino, Italy
| | - Ornella Abollino
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Paola Caroppo
- Unit of Neurology 5 and Neuropathology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuseppe Di Fede
- Unit of Neurology 5 and Neuropathology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologico Carlo Besta, Milan, Italy
| | - Antonio Indaco
- Unit of Neurology 5 and Neuropathology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologico Carlo Besta, Milan, Italy
| | - Innocenzo Rainero
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Torino, Italy
| | - Caterina Guiot
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Torino, Italy
| |
Collapse
|
46
|
Kenkhuis B, Somarakis A, de Haan L, Dzyubachyk O, IJsselsteijn ME, de Miranda NFCC, Lelieveldt BPF, Dijkstra J, van Roon-Mom WMC, Höllt T, van der Weerd L. Iron loading is a prominent feature of activated microglia in Alzheimer's disease patients. Acta Neuropathol Commun 2021; 9:27. [PMID: 33597025 PMCID: PMC7887813 DOI: 10.1186/s40478-021-01126-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 01/30/2021] [Indexed: 12/19/2022] Open
Abstract
Brain iron accumulation has been found to accelerate disease progression in amyloid-β(Aβ) positive Alzheimer patients, though the mechanism is still unknown. Microglia have been identified as key players in the disease pathogenesis, and are highly reactive cells responding to aberrations such as increased iron levels. Therefore, using histological methods, multispectral immunofluorescence and an automated in-house developed microglia segmentation and analysis pipeline, we studied the occurrence of iron-accumulating microglia and the effect on its activation state in human Alzheimer brains. We identified a subset of microglia with increased expression of the iron storage protein ferritin light chain (FTL), together with increased Iba1 expression, decreased TMEM119 and P2RY12 expression. This activated microglia subset represented iron-accumulating microglia and appeared morphologically dystrophic. Multispectral immunofluorescence allowed for spatial analysis of FTL+Iba1+-microglia, which were found to be the predominant Aβ-plaque infiltrating microglia. Finally, an increase of FTL+Iba1+-microglia was seen in patients with high Aβ load and Tau load. These findings suggest iron to be taken up by microglia and to influence the functional phenotype of these cells, especially in conjunction with Aβ.
Collapse
Affiliation(s)
- Boyd Kenkhuis
- Department of Human Genetics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Antonios Somarakis
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lorraine de Haan
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Oleh Dzyubachyk
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | - Jouke Dijkstra
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Willeke M C van Roon-Mom
- Department of Human Genetics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Thomas Höllt
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Intelligent Systems, Delft University of Technology, Delft, The Netherlands
| | - Louise van der Weerd
- Department of Human Genetics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
47
|
Lopes de Andrade V, Marreilha dos Santos AP, Aschner M. NEUROTOXICITY OF METAL MIXTURES. ADVANCES IN NEUROTOXICOLOGY 2021; 5:329-364. [PMID: 34263093 PMCID: PMC8276944 DOI: 10.1016/bs.ant.2020.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Environmental exposures and/or alterations in the homeostasis of essential transition metals (ETM), such as Fe, Cu, Zn or Mn, are known to contribute to neurodegenerative diseases (ND), such as Alzheimer's Disease (AD) and Parkinson's Disease (PD). Aberrant ETM homeostasis leads to altered distributions, as significant amounts may accumulate in specific brain areas, while causing metal deficiency in others. The disruption of processes reliant on the interplay between these ETM, may lead to loss of metal balance and the ensuing neurotoxicity via shared mechanisms, such as the induction of oxidative stress (OS). Both ETM imbalance and OS may play a role, via complex positive loop processes, in primary neuropathological signatures of AD, such as the accumulation of amyloid plaques and neurofibrillary tangles (NTF), and in PD, α-Syn aggregation and loss of dopamine(DA)rgic neurons. The association between ETM imbalance and ND is rarely approached under the view that metals such as Fe, Cu, Zn and Mn, can act as dangerous endogenous neurotoxic mixtures when their control mechanisms became disrupted. In fact, their presence as mixtures implies intricacies, which should be kept in mind when developing therapies for complex disorders of metal dyshomeostasis, which commonly occur in ND.
Collapse
Affiliation(s)
- Vanda Lopes de Andrade
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa. Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Ana Paula Marreilha dos Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa. Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Michael Aschner
- Albert Einstein College of Medicine. Einstein Center of Toxicology.1300 Morris Park Avenue. Bronx, NY 10461
| |
Collapse
|
48
|
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
|
49
|
Meysami S, Raji CA, Merrill DA, Porter VR, Mendez MF. Quantitative MRI Differences Between Early versus Late Onset Alzheimer's Disease. Am J Alzheimers Dis Other Demen 2021; 36:15333175211055325. [PMID: 34814740 PMCID: PMC10623969 DOI: 10.1177/15333175211055325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Investigators report greater parietal tau deposition and alternate frontoparietal network involvement in early onset Alzheimer's Disease (EOAD) with onset <65 years as compared with typical late onset AD (LOAD). To determine whether clinical brain MRI volumes reflect these differences in EOAD compared with LOAD. This study investigated the clinical MRI scans of 45 persons with Clinically Probable AD with onset <65 years, and compared them to 32 with Clinically Probable AD with onset ≥65 years. Brain volumes on their T1 MRI scans were quantified with a volumetric program. Receiver operating curve analyses were performed. Persons with EOAD had significantly smaller parietal lobes (volumetric percentiles) than LOAD. Late onset Alzheimer's Disease had a smaller left putamen and hippocampus. Area Under the Curve was 96.5% with brain region delineation of EOAD compared to LOAD. This study indicates parietal atrophy less than 30% of normal on clinical MRI scans is suggestive of EOAD compared to LOAD.
Collapse
Affiliation(s)
- Somayeh Meysami
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Cyrus A. Raji
- Mallinckrodt Institute of Radiology, Division of Neuroradiology, Washington University in St Louis, St Louis, MO, USA
| | - David A. Merrill
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
- Providence and St Johns Health Center, Pacific Neuroscience Institute, Santa Monica, CA, USA
| | - Verna R. Porter
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
- Providence and St Johns Health Center, Pacific Neuroscience Institute, Santa Monica, CA, USA
| | - Mario F. Mendez
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
- V.A. Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
50
|
Payoux P, Ranjeva JP. Contributions of PET and MRI imaging in the evaluation of CNS drugs in human neurodegenerative diseases. Therapie 2020; 76:121-126. [PMID: 33563477 DOI: 10.1016/j.therap.2020.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/29/2020] [Indexed: 11/19/2022]
Abstract
This manuscript reviews the contributions of the neuroimaging methods including PET, conventional and advanced MRI methods to monitor the effect of new disease modifying drugs in neurodegenerative diseases. It now seems obvious that in many pathologies these two techniques are more and more complementary.
Collapse
Affiliation(s)
- Pierre Payoux
- Inserm, UPS, ToNIC, Nuclear Medicine Department, Toulouse NeuroImaging Center, University Hospital of Toulouse France, Université de Toulouse, 31000 Toulouse, France.
| | - Jean-Philippe Ranjeva
- CNRS, CRMBM, Aix-Marseille University, 13385 Marseille, France; CEMEREM, AP-HM, University Hospital Timone, 13385 Marseille, France
| |
Collapse
|