51
|
Bossoni L, Hegeman-Kleinn I, van Duinen SG, Bulk M, Vroegindeweij LHP, Langendonk JG, Hirschler L, Webb A, van der Weerd L. Off-resonance saturation as an MRI method to quantify mineral- iron in the post-mortem brain. Magn Reson Med 2021; 87:1276-1288. [PMID: 34655092 PMCID: PMC9293166 DOI: 10.1002/mrm.29041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022]
Abstract
Purpose To employ an off‐resonance saturation method to measure the mineral‐iron pool in the postmortem brain, which is an endogenous contrast agent that can give information on cellular iron status. Methods An off‐resonance saturation acquisition protocol was implemented on a 7 Tesla preclinical scanner, and the contrast maps were fitted to an established analytical model. The method was validated by correlation and Bland‐Altman analysis on a ferritin‐containing phantom. Mineral‐iron maps were obtained from postmortem tissue of patients with neurological diseases characterized by brain iron accumulation, that is, Alzheimer disease, Huntington disease, and aceruloplasminemia, and validated with histology. Transverse relaxation rate and magnetic susceptibility values were used for comparison. Results In postmortem tissue, the mineral‐iron contrast colocalizes with histological iron staining in all the cases. Iron concentrations obtained via the off‐resonance saturation method are in agreement with literature. Conclusions Off‐resonance saturation is an effective way to detect iron in gray matter structures and partially mitigate for the presence of myelin. If a reference region with little iron is available in the tissue, the method can produce quantitative iron maps. This method is applicable in the study of diseases characterized by brain iron accumulation and can complement existing iron‐sensitive parametric methods.
Collapse
Affiliation(s)
- Lucia Bossoni
- C. J. Gorter Center for High field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Sjoerd G van Duinen
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marjolein Bulk
- C. J. Gorter Center for High field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Neurology, Alzheimer Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Lena H P Vroegindeweij
- Department of Internal Medicine, Center for Lysosomal and Metabolic Diseases, Porphyria Center Rotterdam, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Janneke G Langendonk
- Department of Internal Medicine, Center for Lysosomal and Metabolic Diseases, Porphyria Center Rotterdam, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Lydiane Hirschler
- C. J. Gorter Center for High field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrew Webb
- C. J. Gorter Center for High field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Louise van der Weerd
- C. J. Gorter Center for High field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
52
|
Isaacs BR, Heijmans M, Kuijf ML, Kubben PL, Ackermans L, Temel Y, Keuken MC, Forstmann BU. Variability in subthalamic nucleus targeting for deep brain stimulation with 3 and 7 Tesla magnetic resonance imaging. NEUROIMAGE-CLINICAL 2021; 32:102829. [PMID: 34560531 PMCID: PMC8463907 DOI: 10.1016/j.nicl.2021.102829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/12/2021] [Accepted: 09/12/2021] [Indexed: 12/13/2022]
Abstract
Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is an effective surgical treatment for Parkinson's disease (PD). Side-effects may, however, be induced when the DBS lead is placed suboptimally. Currently, lower field magnetic resonance imaging (MRI) at 1.5 or 3 Tesla (T) is used for targeting. Ultra-high-field MRI (7 T and above) can obtain superior anatomical information and might therefore be better suited for targeting. This study aims to test whether optimized 7 T imaging protocols result in less variable targeting of the STN for DBS compared to clinically utilized 3 T images. Three DBS-experienced neurosurgeons determined the optimal STN DBS target site on three repetitions of 3 T-T2, 7 T-T2*, 7 T-R2* and 7 T-QSM images for five PD patients. The distance in millimetres between the three repetitive coordinates was used as an index of targeting variability and was compared between field strength, MRI contrast and repetition with a Bayesian ANOVA. Further, the target coordinates were registered to MNI space, and anatomical coordinates were compared between field strength, MRI contrast and repetition using a Bayesian ANOVA. The results indicate that the neurosurgeons are stable in selecting the DBS target site across MRI field strength, MRI contrast and repetitions. The analysis of the coordinates in MNI space however revealed that the actual selected location of the electrode is seemingly more ventral when using the 3 T scan compared to the 7 T scans.
Collapse
Affiliation(s)
- Bethany R Isaacs
- Integrative Model-based Cognitive Neuroscience Research Unit, University of Amsterdam, Amsterdam, The Netherlands; Translational Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Margot Heijmans
- Translational Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Mark L Kuijf
- Translational Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Neurology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Pieter L Kubben
- Translational Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Linda Ackermans
- Translational Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Yasin Temel
- Translational Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Max C Keuken
- Integrative Model-based Cognitive Neuroscience Research Unit, University of Amsterdam, Amsterdam, The Netherlands
| | - Birte U Forstmann
- Integrative Model-based Cognitive Neuroscience Research Unit, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
53
|
Chen L, Cai S, van Zijl PC, Li X. Single-step calculation of susceptibility through multiple orientation sampling. NMR IN BIOMEDICINE 2021; 34:e4517. [PMID: 33822416 PMCID: PMC8184590 DOI: 10.1002/nbm.4517] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 03/06/2021] [Accepted: 03/14/2021] [Indexed: 06/12/2023]
Abstract
Quantitative susceptibility mapping (QSM) was developed to estimate the spatial distribution of magnetic susceptibility from MR signal phase acquired using a gradient echo (GRE) sequence. The field-to-susceptibility inversion in QSM is known to be ill-posed and needs numerical stabilization through either regularization or data oversampling. The calculation of susceptibility through the multiple orientation sampling (COSMOS) method uses phase data acquired at three or more head orientations to achieve a well-conditioned field-to-susceptibility inversion and is often considered the gold standard for in vivo QSM. However, the conventional COSMOS approach, here named multistep COSMOS (MSCOSMOS), solves the dipole inversion from the local field derived from raw GRE phase through multiple steps of phase preprocessing. Error propagations between these consecutive phase processing steps can thus affect the final susceptibility quantification. On the other hand, recently proposed single-step QSM (SSQSM) methods aim to solve an integrated inversion from unprocessed or total phase to mitigate such error propagations but have been limited to single orientation QSM. This study therefore aimed to test the feasibility of using single-step COSMOS (SSCOSMOS) to jointly perform background field removal and dipole inversion with multiple orientation sampling, which could serve as a better standard for gauging SSQSM methods. We incorporated multiple spherical mean value (SMV) kernels of various radii with the dipole inversion in SSCOSMOS. QSM reconstructions with SSCOSMOS and MSCOSMOS were compared using both simulations with a numerical head phantom and in vivo human brain data. SSCOSMOS permitted integrated background removal and dipole inversion without the need to adjust any regularization parameters. In addition, with sufficiently large SMV kernels, SSCOSMOS performed consistently better than MSCOSMOS in all the tested error metrics in our simulations, giving better susceptibility quantification and smaller reconstruction error. Consistent tissue susceptibility values were obtained between SSCOSMOS and MSCOSMOS.
Collapse
Affiliation(s)
- Lin Chen
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
- Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Shuhui Cai
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | - Peter C.M. van Zijl
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
- Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Xu Li
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
- Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
54
|
Xu M, Guo Y, Cheng J, Xue K, Yang M, Song X, Feng Y, Cheng J. Brain iron assessment in patients with First-episode schizophrenia using quantitative susceptibility mapping. NEUROIMAGE-CLINICAL 2021; 31:102736. [PMID: 34186296 PMCID: PMC8254125 DOI: 10.1016/j.nicl.2021.102736] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/30/2021] [Accepted: 06/17/2021] [Indexed: 11/28/2022]
Abstract
Patients with first-episode schizophrenia had significantly decreased QSM values in the bilateral substantia nigra, left red nucleus and left thalamus. Patients with first-episode schizophrenia had significantly increased regional volumes in the bilateral putamen and bilateral substantia nigra. QSM provides superior sensitivity over R2* mapping in the evaluation of schizophrenia-related iron alterations. QSM values in regions that showed intergroup differences did not exhibited significant correlations with PANSS scores.
Purpose Decreased serum ferritin level was recently found in schizophrenia. Whether the brain iron concentration in schizophrenia exists abnormality is of research significance. Quantitative susceptibility mapping (QSM) was used in this study to assess brain iron changes in the grey matter nuclei of patients with first-episode schizophrenia. Methods The local ethics committee approved the study, and all subjects gave written informed consent. Thirty patients with first-episode schizophrenia and 30 age and gender-matched healthy controls were included in this study. QSM and effective transverse relaxation rate (R2*) maps were reconstructed from a three-dimensional multi-echo gradient-echo sequence. The inter-group differences of regional QSM values, R2* values and volumes were calculated in the grey matter nuclei, including bilateral caudate nucleus, putamen, globus pallidus, substantia nigra, red nucleus, and thalamus. The diagnostic performance of QSM and R2* was evaluated using receiver operating characteristic curve. The correlations between regional iron variations and clinical PANSS (Positive and Negative Syndrome Scale) scores were assessed using partial correlation analysis. Results Compared to healthy controls, patients with first-episode schizophrenia had significantly decreased QSM values (less paramagnetic) in the bilateral substantia nigra, left red nucleus and left thalamus (p < 0.05, FDR correction). QSM proved more sensitive than R2* regarding inter-group differences. The highest diagnostic performance for first-episode schizophrenia was observed in QSM value of the left substantia nigra (area under the curve, AUC = 0.718, p = 0.004). Regional volumes of bilateral putamen and bilateral substantia nigra were increased (p < 0.05, FDR correction) in first-episode schizophrenia. However, both QSM and R2* values did not show significant correlations with PANSS scores (p > 0.05). Conclusion This study reveals decreased iron concentration in grey matter nuclei of patients with first-episode schizophrenia. QSM provides superior sensitivity over R2* in the evaluation of schizophrenia-related brain iron changes. It demonstrated that QSM may be a potential biomarker for further understanding the pathophysiological mechanism of first-episode schizophrenia.
Collapse
Affiliation(s)
- Man Xu
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yihao Guo
- MR Collaboration, Siemens Healthcare Ltd, Guangzhou, China
| | - Junying Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kangkang Xue
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meng Yang
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xueqin Song
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanqiu Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Medical Image Processing, Key Laboratory of Mental Health of the Ministry of Education & Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China.
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
55
|
Reguera Acuña A, Suárez San Martín E, García Fernández C, Fernández Menéndez S, Blázquez Estrada M, Amorín Díaz M, Menéndez González M, Álvarez Martínez V. A series of cases with Huntington-like phenotype and intermediate repeats in HTT. J Neurol Sci 2021; 425:117452. [PMID: 33892278 DOI: 10.1016/j.jns.2021.117452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/27/2021] [Accepted: 04/12/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Intermediate Alleles (IAs) are expansions of CAG repeats in the HTT gene between 27 and 35 repeats which pathogenic meaning remains controversial. They are present in the general population but there is an increasing number of cases with Huntington-like phenotype reported. METHODS We reviewed the medical records of cases in our centre where the neurologist suspected Huntington's disease (HD) as one of the feasible diagnoses and genetic testing showed the number of CAG repeats was in the "intermediate range". We gathered the type of symptoms in all cases and the main neuroimaging findings when available. RESULTS We found 14 cases, 8 males and 6 females, with average age at onset at 64 years old. Most cases exhibited some type of extrapyramidal symptoms. Cognitive and/or behavioral symptoms were also present in most cases (being depression, anxiety and cognitive impairment the most frequent ones). In one case we found deposits of iron in the basal ganglia in the MRI, and in another case we found diffuse cortical hypometabolism with predominantly frontal bilateral involvement and bilateral focal deficit of both caudate and thalamus in the FDG-PET. CONCLUSION The clinical and neuroimaging findings of some cases with IA in this series are compatible with the clinical picture of HD but also with several other alternative diagnoses. Therefore we can not establish association between IA and HD. Larger series with more comprehensive diagnostic workout and neuropathological studies are needed to confirm or rule out whether IAs in the HTT gene may cause HD.
Collapse
Affiliation(s)
| | - Esther Suárez San Martín
- Servicio de Neurología, Hospital Universitario Central de Asturias, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Spain
| | - Ciara García Fernández
- Servicio de Neurología, Hospital Universitario Central de Asturias, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Spain
| | - Santiago Fernández Menéndez
- Servicio de Neurología, Hospital Universitario Central de Asturias, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Spain
| | - Marta Blázquez Estrada
- Servicio de Neurología, Hospital Universitario Central de Asturias, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Spain; Departamento de Medicina, Universidad de Oviedo, Spain
| | - Manuel Amorín Díaz
- Servicio de Neurología, Hospital Universitario Central de Asturias, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Spain
| | - Manuel Menéndez González
- Servicio de Neurología, Hospital Universitario Central de Asturias, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Spain; Departamento de Medicina, Universidad de Oviedo, Spain.
| | - Victoria Álvarez Martínez
- Instituto de Investigación Sanitaria del Principado de Asturias, Spain; Servicio de Genética, Hospital Universitario Central de Asturias, Spain
| |
Collapse
|
56
|
Donley DW, Realing M, Gigley JP, Fox JH. Iron activates microglia and directly stimulates indoleamine-2,3-dioxygenase activity in the N171-82Q mouse model of Huntington's disease. PLoS One 2021; 16:e0250606. [PMID: 33989290 PMCID: PMC8121302 DOI: 10.1371/journal.pone.0250606] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/10/2021] [Indexed: 01/05/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by a dominant CAG-repeat expansion in the huntingtin gene. Microglial activation is a key feature of HD pathology, and is present before clinical disease onset. The kynurenine pathway (KP) of tryptophan degradation is activated in HD, and is thought to contribute to disease progression. Indoleamine-2,3-dioxygenase (IDO) catalyzes the first step in this pathway; this and other pathway enzymes reside with microglia. While HD brain microglia accumulate iron, the role of iron in promoting microglial activation and KP activity is unclear. Here we utilized the neonatal iron supplementation model to investigate the relationship between iron, microglial activation and neurodegeneration in adult HD mice. We show in the N171-82Q mouse model of HD microglial morphologic changes consistent with immune activation. Neonatal iron supplementation in these mice promoted neurodegeneration and resulted in additional microglial activation in adults as determined by increased soma volume and decreased process length. We further demonstrate that iron activates IDO, both in brain lysates and purified recombinant protein (EC50 = 1.24 nM). Brain IDO activity is increased by HD. Neonatal iron supplementation further promoted IDO activity in cerebral cortex, altered KP metabolite profiles, and promoted HD neurodegeneration as measured by brain weights and striatal volumes. Our results demonstrate that dietary iron is an important activator of microglia and the KP pathway in this HD model, and that this occurs in part through a direct effect on IDO. The findings are relevant to understanding how iron promotes neurodegeneration in HD.
Collapse
Affiliation(s)
- David W. Donley
- Department of Veterinary Sciences, University of Wyoming, Laramie, WY, United States of America
- Neuroscience Graduate Program, University of Wyoming, Laramie, WY, United States of America
| | - Marley Realing
- Microbiology Undergraduate Program, University of Wyoming, Laramie, WY, United States of America
| | - Jason P. Gigley
- Department of Molecular Biology, University of Wyoming, Laramie, WY, United States of America
| | - Jonathan H. Fox
- Department of Veterinary Sciences, University of Wyoming, Laramie, WY, United States of America
- Neuroscience Graduate Program, University of Wyoming, Laramie, WY, United States of America
| |
Collapse
|
57
|
Simmons DA, Mills BD, Butler Iii RR, Kuan J, McHugh TLM, Akers C, Zhou J, Syriani W, Grouban M, Zeineh M, Longo FM. Neuroimaging, Urinary, and Plasma Biomarkers of Treatment Response in Huntington's Disease: Preclinical Evidence with the p75 NTR Ligand LM11A-31. Neurotherapeutics 2021; 18:1039-1063. [PMID: 33786806 PMCID: PMC8423954 DOI: 10.1007/s13311-021-01023-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2021] [Indexed: 12/13/2022] Open
Abstract
Huntington's disease (HD) is caused by an expansion of the CAG repeat in the huntingtin gene leading to preferential neurodegeneration of the striatum. Disease-modifying treatments are not yet available to HD patients and their development would be facilitated by translatable pharmacodynamic biomarkers. Multi-modal magnetic resonance imaging (MRI) and plasma cytokines have been suggested as disease onset/progression biomarkers, but their ability to detect treatment efficacy is understudied. This study used the R6/2 mouse model of HD to assess if structural neuroimaging and biofluid assays can detect treatment response using as a prototype the small molecule p75NTR ligand LM11A-31, shown previously to reduce HD phenotypes in these mice. LM11A-31 alleviated volume reductions in multiple brain regions, including striatum, of vehicle-treated R6/2 mice relative to wild-types (WTs), as assessed with in vivo MRI. LM11A-31 also normalized changes in diffusion tensor imaging (DTI) metrics and diminished increases in certain plasma cytokine levels, including tumor necrosis factor-alpha and interleukin-6, in R6/2 mice. Finally, R6/2-vehicle mice had increased urinary levels of the p75NTR extracellular domain (ecd), a cleavage product released with pro-apoptotic ligand binding that detects the progression of other neurodegenerative diseases; LM11A-31 reduced this increase. These results are the first to show that urinary p75NTR-ecd levels are elevated in an HD mouse model and can be used to detect therapeutic effects. These data also indicate that multi-modal MRI and plasma cytokine levels may be effective pharmacodynamic biomarkers and that using combinations of these markers would be a viable and powerful option for clinical trials.
Collapse
Affiliation(s)
- Danielle A Simmons
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Brian D Mills
- Department of Radiology, Stanford University Medical Center, Stanford, CA, 94305, USA
| | - Robert R Butler Iii
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jason Kuan
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Tyne L M McHugh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Carolyn Akers
- Department of Radiology, Stanford University Medical Center, Stanford, CA, 94305, USA
| | - James Zhou
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Wassim Syriani
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Maged Grouban
- Department of Radiology, Stanford University Medical Center, Stanford, CA, 94305, USA
| | - Michael Zeineh
- Department of Radiology, Stanford University Medical Center, Stanford, CA, 94305, USA
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
58
|
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
|
59
|
Johnson EB, Parker CS, Scahill RI, Gregory S, Papoutsi M, Zeun P, Osborne-Crowley K, Lowe J, Nair A, Estevez-Fraga C, Fayer K, Rees G, Zhang H, Tabrizi SJ. Altered iron and myelin in premanifest Huntington's Disease more than 20 years before clinical onset: Evidence from the cross-sectional HD Young Adult Study. EBioMedicine 2021; 65:103266. [PMID: 33706250 PMCID: PMC7960938 DOI: 10.1016/j.ebiom.2021.103266] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 02/14/2021] [Accepted: 02/16/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Pathological processes in Huntington's disease (HD) begin many years prior to symptom onset. Recently we demonstrated that in a premanifest cohort approximately 24 years from predicted disease onset, despite intact function, there was evidence of subtle neurodegeneration. Here, we use novel imaging techniques to determine whether macro- and micro-structural changes can be detected across the whole-brain in the same cohort. METHODS 62 premanifest HD (PreHD) and 61 controls from the HD Young Adult Study (HD-YAS) were included. Grey and white matter volume, diffusion weighted imaging (DWI) measures of white matter microstructure, multiparametric maps (MPM) estimating myelin and iron content from magnetization transfer (MT), proton density (PD), longitudinal relaxation (R1) and effective transverse relaxation (R2*), and myelin g-ratio were examined. Group differences between PreHD and controls were assessed; associations between all imaging metrics and disease burden and CSF neurofilament light (NfL) were also performed. Volumetric and MPM results were corrected at a cluster-wise value of familywise error (FWE) 0.05. Diffusion and g-ratio results were corrected via threshold-free cluster enhancement at FWE 0.05. FINDINGS We showed significantly increased R1 and R2*, suggestive of increased iron, in the putamen, globus pallidum and external capsule of PreHD participants. There was also a significant association between lower cortical R2*, suggestive of reduced myelin or iron, and higher CSF NfL in the frontal lobe and the parieto-occipital cortices. No other results were significant at corrected levels. INTERPRETATION Increased iron in subcortical structures and the surrounding white matter is a feature of very early PreHD. Furthermore, increases in CSF NfL were linked to microstructural changes in the posterior parietal-occipital cortex, a region previously shown to undergo some of the earliest cortical changes in HD. These findings suggest that disease related process are occurring in both subcortical and cortical regions more than 20 years from predicted disease onset.
Collapse
Affiliation(s)
- Eileanoir B Johnson
- Huntington's Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London, UK.
| | - Christopher S Parker
- Centre for Medical Image Computing, Department of Computer Science, UCL, London, UK
| | - Rachael I Scahill
- Huntington's Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Sarah Gregory
- Huntington's Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Marina Papoutsi
- Huntington's Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London, UK; IXICO Plc, London, , UK
| | - Paul Zeun
- Huntington's Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Katherine Osborne-Crowley
- Division of Equity, Diversity and Inclusion, University of New South Wales, Sydney, New South Wales, Australia
| | - Jessica Lowe
- Huntington's Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Akshay Nair
- Huntington's Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London, UK; Max Planck University College London Centre for Computational Psychiatry and Ageing Research, UCL Queen Square Institute of Neurology, London, UK
| | - Carlos Estevez-Fraga
- Huntington's Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Kate Fayer
- Huntington's Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Geraint Rees
- University College London Institute of Cognitive Neuroscience, University College London, London, UK
| | - Hui Zhang
- Centre for Medical Image Computing, Department of Computer Science, UCL, London, UK
| | - Sarah J Tabrizi
- Huntington's Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London, UK; Dementia Research Institute at University College London, London, UK
| |
Collapse
|
60
|
Ravanfar P, Loi SM, Syeda WT, Van Rheenen TE, Bush AI, Desmond P, Cropley VL, Lane DJR, Opazo CM, Moffat BA, Velakoulis D, Pantelis C. Systematic Review: Quantitative Susceptibility Mapping (QSM) of Brain Iron Profile in Neurodegenerative Diseases. Front Neurosci 2021; 15:618435. [PMID: 33679303 PMCID: PMC7930077 DOI: 10.3389/fnins.2021.618435] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
Iron has been increasingly implicated in the pathology of neurodegenerative diseases. In the past decade, development of the new magnetic resonance imaging technique, quantitative susceptibility mapping (QSM), has enabled for the more comprehensive investigation of iron distribution in the brain. The aim of this systematic review was to provide a synthesis of the findings from existing QSM studies in neurodegenerative diseases. We identified 80 records by searching MEDLINE, Embase, Scopus, and PsycInfo databases. The disorders investigated in these studies included Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Wilson's disease, Huntington's disease, Friedreich's ataxia, spinocerebellar ataxia, Fabry disease, myotonic dystrophy, pantothenate-kinase-associated neurodegeneration, and mitochondrial membrane protein-associated neurodegeneration. As a general pattern, QSM revealed increased magnetic susceptibility (suggestive of increased iron content) in the brain regions associated with the pathology of each disorder, such as the amygdala and caudate nucleus in Alzheimer's disease, the substantia nigra in Parkinson's disease, motor cortex in amyotrophic lateral sclerosis, basal ganglia in Huntington's disease, and cerebellar dentate nucleus in Friedreich's ataxia. Furthermore, the increased magnetic susceptibility correlated with disease duration and severity of clinical features in some disorders. Although the number of studies is still limited in most of the neurodegenerative diseases, the existing evidence suggests that QSM can be a promising tool in the investigation of neurodegeneration.
Collapse
Affiliation(s)
- Parsa Ravanfar
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
| | - Samantha M Loi
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia.,Neuropsychiatry, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Warda T Syeda
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
| | - Tamsyn E Van Rheenen
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia.,Centre for Mental Health, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Patricia Desmond
- Melbourne Brain Centre Imaging Unit, Department of Medicine and Radiology, The University of Melbourne, Parkville, VIC, Australia.,Department of Radiology, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Vanessa L Cropley
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia.,Centre for Mental Health, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Carlos M Opazo
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Bradford A Moffat
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia.,Melbourne Brain Centre Imaging Unit, Department of Medicine and Radiology, The University of Melbourne, Parkville, VIC, Australia
| | - Dennis Velakoulis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia.,Neuropsychiatry, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia.,Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
61
|
Yao MY, Liu T, Zhang L, Wang MJ, Yang Y, Gao J. Role of ferroptosis in neurological diseases. Neurosci Lett 2021; 747:135614. [PMID: 33485988 DOI: 10.1016/j.neulet.2020.135614] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/14/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023]
Abstract
Ferroptosis is a newly identified form of nonapoptotic regulated cell death (RCD) characterized by iron-dependent accumulation of lipid peroxides which leads to oxidative stress and cell death. Recent studies have indicated that ferroptosis plays an essential role in the pathology of neurological diseases, such as intracerebral hemorrhage, ischemic stroke, epilepsy, neurodegenerative diseases, traumatic brain injury and brain cancer. This review focuses on the latest researches on the relationship of ferroptosis with nervous system diseases, highlighting the ferroptosis-based mechanisms, and elaborating the new perspective therapeutic targets of neurological disorders.
Collapse
Affiliation(s)
- Min-Yi Yao
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Tao Liu
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Liang Zhang
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Ming-Jian Wang
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yong Yang
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Jing Gao
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
62
|
许 欢, 孟 庆, 樊 文, 王 雪, 刘 梦, 陈 志. [Reproducibility analysis of quantitative susceptibility mapping of cerebral subcortical nuclei in healthy adults]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:1810-1815. [PMID: 33380400 PMCID: PMC7835684 DOI: 10.12122/j.issn.1673-4254.2020.12.17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVE To investigate the intra- and inter-scanner reproducibility of quantitative susceptibility mapping (QSM) of cerebral subcortical nuclei in healthy adults. METHODS QSM was performed in 21 healthy adults on two different 3.0T MR scanners, and the region of interest (ROI) method was used to measure the magnetic susceptibility value of the left subcortical nuclei (the head of the caudate, putamen, globus pallidus, thalamus, substantia nigra and red nucleus). The intraclass correlation coefficient (ICC) and Bland-Altman method were used to evaluate the inter-scanner and intra-scanner reliability. RESULTS The ICCs of the susceptibility value ranged from 0.90 to 0.99 for all the subcortical gray nuclei except for the head of the caudate nucleus measured on the same MR scanner by the same observer. Bland-Altman analysis revealed that the points with susceptibility differences for all the subcortical gray nuclei except for substantia nigra located in the 95% CI of limits of agreement for the same MR scanner. The ICCs of the susceptibility value for the inter-scanner was 0.49 (0.08-0.75) for the head of the caudate nuleus, 0.80 (0.57-0.91) for the putamen, 0.77 (0.51-0.90) for the globus pallidus, 0.78 (0.54-0.91) for the thalamus, 0.80 (0.56-0.91) for the substantia nigra and 0.93 (0.83-0.97) for the red nucleus. The points with susceptibility difference (95.2%, 20/21) located in the 95% CI of limits of agreement for the putamen and the thalamus measured on two different MR scanners. CONCLUSIONS The intra-scanner reproducibility of QSM of the subcortical gray nuclei is superior to the inter-scanner reproducibility in healthy adults.
Collapse
Affiliation(s)
- 欢 许
- 海南省儋州市人民医院放射科,海南 儋州 571700Department of Radiology, Danzhou People's Hospital, Danzhou 571700, China
| | - 庆林 孟
- 解放军总医院海南医院放射科,海南 三亚 572013Department of Radiology, Hainan Hospital Affiliated to General Hospital of PLA, Sanya 572013, China
| | - 文萍 樊
- 解放军总医院海南医院放射科,海南 三亚 572013Department of Radiology, Hainan Hospital Affiliated to General Hospital of PLA, Sanya 572013, China
| | - 雪 王
- 解放军总医院海南医院放射科,海南 三亚 572013Department of Radiology, Hainan Hospital Affiliated to General Hospital of PLA, Sanya 572013, China
| | - 梦琦 刘
- 解放军总医院海南医院放射科,海南 三亚 572013Department of Radiology, Hainan Hospital Affiliated to General Hospital of PLA, Sanya 572013, China
- 解放军总医院第一医学中心放射科,北京 100853Department of Radiology, First Medical Center of General Hospital of PLA, Beijing 100853, China
| | - 志晔 陈
- 解放军总医院海南医院放射科,海南 三亚 572013Department of Radiology, Hainan Hospital Affiliated to General Hospital of PLA, Sanya 572013, China
| |
Collapse
|
63
|
The role of transferrins and iron-related proteins in brain iron transport: applications to neurological diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 123:133-162. [PMID: 33485481 DOI: 10.1016/bs.apcsb.2020.09.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Iron transport in the central nervous system (CNS) is a highly regulated process in which several important proteins participate to ensure this important metal reaches its sites of action. However, iron accumulation has been shown to be a common factor in different neurological disorders such as Alzheimer's disease, Parkinson's disease, Huntington's disease, Multiple Sclerosis, and Sanfilippo syndrome. This review is divided into four parts. The first part describes brain iron transport in homeostasis, mentioning the main proteins involved, whereas the second part contrasts the consequences of iron dysregulation, elaborating on its role in the aforementioned neurodegenerative diseases. The third part details the functions of the main proteins involved in brain iron homeostasis and their role in neurodegeneration. In the fourth part, in order to highlight the importance of transport proteins, the focus is set on human serum transferrin, the main iron transport protein. This final part describes perspectives about the mechanisms and chemical properties of human transferrin for the development of potential targeted drug delivery systems across the blood-brain barrier (BBB) or enhancers for the treatment of neurological diseases.
Collapse
|
64
|
Bulk M, Hegeman-Kleinn I, Kenkhuis B, Suidgeest E, van Roon-Mom W, Lewerenz J, van Duinen S, Ronen I, van der Weerd L. Pathological characterization of T2*-weighted MRI contrast in the striatum of Huntington's disease patients. Neuroimage Clin 2020; 28:102498. [PMID: 33395988 PMCID: PMC7677121 DOI: 10.1016/j.nicl.2020.102498] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 01/19/2023]
Abstract
Previous MRI studies consistently reported iron accumulation within the striatum of patients with Huntington's disease (HD). However, the pattern and origin of iron accumulation is poorly understood. This study aimed to characterize the histopathological correlates of iron-sensitive ex vivo MRI contrast change in HD brains. To this end, T2*-weighted 7T MRI was performed on postmortem tissue of the striatum of three control subjects and 10 HD patients followed by histological examination. In addition, formalin-fixed paraffin-embedded material of three control subjects and 14 HD patients was selected for only histology to identify the cellular localization of iron using stainings for iron, myelin, microglia and astrocytes. As expected HD striata showed prominent atrophy. Compared to controls, the striatum of HD patients was in general more hypointense on T2*-weighted high-field MRI and showed a more intense histopathological staining for iron. In addition, T2*-weighted MRI identified large focal hypointensities within the striatum of HD patients. Upon histological examination, these large focal hypointensities frequently colocalized with enlarged perivascular spaces and iron was found within the vessel wall and reactive astrocytes. In conclusion, we show that the striatum of HD patients has a distinctive phenotype on T2*-weighted MRI compared to control subjects. On ex vivo MRI, these contrast changes are heavily biased by enlarged perivascular spaces from which it is currently unknown whether this is a fixation artefact or a disease specific observation. Clinically, the observation of iron within reactive astrocytes is of importance for the interpretation and understanding of the potential underlying mechanisms of T2*-weighted MRI results in HD patients.
Collapse
Affiliation(s)
- Marjolein Bulk
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Department of Human Genetics, 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
| | - Ernst Suidgeest
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Willeke van Roon-Mom
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan Lewerenz
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Sjoerd van Duinen
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Itamar Ronen
- Department of Radiology, 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
| |
Collapse
|
65
|
Zhou RP, Chen Y, Wei X, Yu B, Xiong ZG, Lu C, Hu W. Novel insights into ferroptosis: Implications for age-related diseases. Theranostics 2020; 10:11976-11997. [PMID: 33204324 PMCID: PMC7667696 DOI: 10.7150/thno.50663] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/29/2020] [Indexed: 12/20/2022] Open
Abstract
Rapid increase in aging populations is an urgent problem because older adults are more likely to suffer from disabilities and age-related diseases (ARDs), burdening healthcare systems and society in general. ARDs are characterized by the progressive deterioration of tissues and organs over time, eventually leading to tissue and organ failure. To date, there are no effective interventions to prevent the progression of ARDs. Hence, there is an urgent need for new treatment strategies. Ferroptosis, an iron-dependent cell death, is linked to normal development and homeostasis. Accumulating evidence, however, has highlighted crucial roles for ferroptosis in ARDs, including neurodegenerative and cardiovascular diseases. In this review, we a) summarize initiation, regulatory mechanisms, and molecular signaling pathways involved in ferroptosis, b) discuss the direct and indirect involvement of the activation and/or inhibition of ferroptosis in the pathogenesis of some important diseases, and c) highlight therapeutic targets relevant for ARDs.
Collapse
Affiliation(s)
- Ren-Peng Zhou
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Yong Chen
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Xin Wei
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Bin Yu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhi-Gang Xiong
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Chao Lu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| |
Collapse
|
66
|
Simultaneous feedback control for joint field and motion correction in brain MRI. Neuroimage 2020; 226:117286. [PMID: 32992003 DOI: 10.1016/j.neuroimage.2020.117286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/21/2020] [Accepted: 08/14/2020] [Indexed: 11/23/2022] Open
Abstract
T2*-weighted gradient-echo sequences count among the most widely used techniques in neuroimaging and offer rich magnitude and phase contrast. The susceptibility effects underlying this contrast scale with B0, making T2*-weighted imaging particularly interesting at high field. High field also benefits baseline sensitivity and thus facilitates high-resolution studies. However, enhanced susceptibility effects and high target resolution come with inherent challenges. Relying on long echo times, T2*-weighted imaging not only benefits from enhanced local susceptibility effects but also suffers from increased field fluctuations due to moving body parts and breathing. High resolution, in turn, renders neuroimaging particularly vulnerable to motion of the head. This work reports the implementation and characterization of a system that aims to jointly address these issues. It is based on the simultaneous operation of two control loops, one for field stabilization and one for motion correction. The key challenge with this approach is that the two loops both operate on the magnetic field in the imaging volume and are thus prone to mutual interference and potential instability. This issue is addressed at the levels of sensing, timing, and control parameters. Performance assessment shows the resulting system to be stable and exhibit adequate loop decoupling, precision, and bandwidth. Simultaneous field and motion control is then demonstrated in examples of T2*-weighted in vivo imaging at 7T.
Collapse
|
67
|
Zhou J, Jin Y, Lei Y, Liu T, Wan Z, Meng H, Wang H. Ferroptosis Is Regulated by Mitochondria in Neurodegenerative Diseases. NEURODEGENER DIS 2020; 20:20-34. [PMID: 32814328 DOI: 10.1159/000510083] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/10/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Neurodegenerative diseases are characterized by a gradual decline in motor and/or cognitive function caused by the selective degeneration and loss of neurons in the central nervous system, but their pathological mechanism is still unclear. Previous research has revealed that many forms of cell death, such as apoptosis and necrosis, occur in neurodegenerative diseases. Research in recent years has noticed that there is a new type of cell death in neurodegenerative diseases: ferroptosis. An increasing body of literature provides evidence for an involvement of ferroptosis in neurodegenerative diseases. SUMMARY In this article, we review a new form of cell death in neurodegenerative diseases: ferroptosis. Ferroptosis is defined as an iron-dependent form of regulated cell death, which occurs through the lethal accumulation of lipid-based reactive oxygen species when glutathione-dependent lipid peroxide repair systems are compromised. Several salient and established features of neurodegenerative diseases (including lipid peroxidation and iron dyshomeostasis) are consistent with ferroptosis, which means that ferroptosis may be involved in the progression of neurodegenerative diseases. In addition, as the center of energy metabolism in cells, mitochondria are also closely related to the regulation of iron homeostasis in the nervous system. At the same time, neurodegenerative diseases are often accompanied by degeneration of mitochondrial activity. Mitochondrial damage has been found to be involved in lipid peroxidation and iron dyshomeostasis in neurodegenerative diseases. Key Messages: Based on the summary of the related mechanisms of ferroptosis, we conclude that mitochondrial damage may affect neurodegenerative diseases by regulating many aspects of ferroptosis, including cell metabolism, iron dyshomeostasis, and lipid peroxidation.
Collapse
Affiliation(s)
- Juepu Zhou
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Yao Jin
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yuhong Lei
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Tianyi Liu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Zheng Wan
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Hao Meng
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China,
| | - Honglei Wang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
68
|
Zhang J, Liu Z, Zhang S, Zhang H, Spincemaille P, Nguyen TD, Sabuncu MR, Wang Y. Fidelity imposed network edit (FINE) for solving ill-posed image reconstruction. Neuroimage 2020; 211:116579. [PMID: 31981779 PMCID: PMC7093048 DOI: 10.1016/j.neuroimage.2020.116579] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 12/20/2019] [Accepted: 01/20/2020] [Indexed: 01/19/2023] Open
Abstract
Deep learning (DL) is increasingly used to solve ill-posed inverse problems in medical imaging, such as reconstruction from noisy and/or incomplete data, as DL offers advantages over conventional methods that rely on explicit image features and hand engineered priors. However, supervised DL-based methods may achieve poor performance when the test data deviates from the training data, for example, when it has pathologies not encountered in the training data. Furthermore, DL-based image reconstructions do not always incorporate the underlying forward physical model, which may improve performance. Therefore, in this work we introduce a novel approach, called fidelity imposed network edit (FINE), which modifies the weights of a pre-trained reconstruction network for each case in the testing dataset. This is achieved by minimizing an unsupervised fidelity loss function that is based on the forward physical model. FINE is applied to two important inverse problems in neuroimaging: quantitative susceptibility mapping (QSM) and under-sampled image reconstruction in MRI. Our experiments demonstrate that FINE can improve reconstruction accuracy.
Collapse
Affiliation(s)
- Jinwei Zhang
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, NY, USA
| | - Zhe Liu
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, NY, USA
| | - Shun Zhang
- Department of Radiology, Weill Medical College of Cornell University, New York, NY, USA
| | - Hang Zhang
- Department of Radiology, Weill Medical College of Cornell University, New York, NY, USA; Department of Electrical and Computer Engineering, Cornell University, Ithaca, NY, USA
| | - Pascal Spincemaille
- Department of Radiology, Weill Medical College of Cornell University, New York, NY, USA
| | - Thanh D Nguyen
- Department of Radiology, Weill Medical College of Cornell University, New York, NY, USA
| | - Mert R Sabuncu
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, NY, USA; Department of Electrical and Computer Engineering, Cornell University, Ithaca, NY, USA
| | - Yi Wang
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, NY, USA.
| |
Collapse
|
69
|
Eskreis-Winkler S, Simon K, Reichman M, Spincemaille P, Nguyen T, Kee Y, Cho J, Christos PJ, Drotman M, Prince MR, Morris EA, Wang Y. Dipole modeling of multispectral signal for detecting metallic biopsy markers during MRI-guided breast biopsy: a pilot study. Magn Reson Med 2020; 83:1380-1389. [PMID: 31631408 PMCID: PMC6949365 DOI: 10.1002/mrm.28017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 09/05/2019] [Accepted: 09/05/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE During MRI-guided breast biopsy, a metallic biopsy marker is deployed at the biopsy site to guide future interventions. Conventional MRI during biopsy cannot distinguish such markers from biopsy site air, and a post-biopsy mammogram is therefore performed to localize marker placement. The purpose of this pilot study is to develop dipole modeling of multispectral signal (DIMMS) as an MRI alternative to eliminate the cost, inefficiency, inconvenience, and ionizing radiation of a mammogram for biopsy marker localization. METHODS DIMMS detects and localizes the biopsy marker by fitting the measured multispectral imaging (MSI) signal to the MRI signal model and marker properties. MSI was performed on phantoms containing titanium biopsy markers and air to illustrate the clinical challenge that DIMMS addresses and on 20 patients undergoing MRI-guided breast biopsy to assess DIMMS feasibility for marker detection. DIMMS was compared to conventional MSI field map thresholding, using the post-procedure mammogram as the reference standard. RESULTS Biopsy markers were detected and localized in 20 of 20 cases using MSI with automated DIMMS post-processing (using a threshold of 0.7) and in 18 of 20 cases using MSI field mapping (using a threshold of 0.65 kHz). CONCLUSION MSI with DIMMS post-processing is a feasible technique for biopsy marker detection and localization during MRI-guided breast biopsy. With a 2-min MSI scan, DIMMS is a promising MRI alternative to the standard-of-care post-biopsy mammogram.
Collapse
Affiliation(s)
- Sarah Eskreis-Winkler
- Department of Radiology, Weill Cornell Medicine, 515 E 71 Street, Suite 104, New York, NY 10021
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Katherine Simon
- Department of Radiology, Weill Cornell Medicine, 515 E 71 Street, Suite 104, New York, NY 10021
| | - Melissa Reichman
- Department of Radiology, Weill Cornell Medicine, 515 E 71 Street, Suite 104, New York, NY 10021
| | - Pascal Spincemaille
- Department of Radiology, Weill Cornell Medicine, 515 E 71 Street, Suite 104, New York, NY 10021
| | - Thanh Nguyen
- Department of Radiology, Weill Cornell Medicine, 515 E 71 Street, Suite 104, New York, NY 10021
| | - Youngwook Kee
- Department of Radiology, Weill Cornell Medicine, 515 E 71 Street, Suite 104, New York, NY 10021
| | - Junghun Cho
- Department of Radiology, Weill Cornell Medicine, 515 E 71 Street, Suite 104, New York, NY 10021
| | - Paul J. Christos
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy & Research, Weill Cornell Medicine, New York, NY
| | - Michele Drotman
- Department of Radiology, Weill Cornell Medicine, 515 E 71 Street, Suite 104, New York, NY 10021
| | - Martin R. Prince
- Department of Radiology, Weill Cornell Medicine, 515 E 71 Street, Suite 104, New York, NY 10021
| | - Elizabeth A. Morris
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Yi Wang
- Department of Radiology, Weill Cornell Medicine, 515 E 71 Street, Suite 104, New York, NY 10021
| |
Collapse
|
70
|
Han C, Liu Y, Dai R, Ismail N, Su W, Li B. Ferroptosis and Its Potential Role in Human Diseases. Front Pharmacol 2020; 11:239. [PMID: 32256352 PMCID: PMC7090218 DOI: 10.3389/fphar.2020.00239] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/21/2020] [Indexed: 12/26/2022] Open
Abstract
Ferroptosis is a novel regulated cell death pattern discovered when studying the mechanism of erastin-killing RAS mutant tumor cells in 2012. It is an iron-dependent programmed cell death pathway mainly caused by an increased redox imbalance but with distinct biological and morphology characteristics when compared to other known cell death patterns. Ferroptosis is associated with various diseases including acute kidney injury, cancer, and cardiovascular, neurodegenerative, and hepatic diseases. Moreover, activation or inhibition of ferroptosis using a variety of ferroptosis initiators and inhibitors can modulate disease progression in animal models. In this review, we provide a comprehensive analysis of the characteristics of ferroptosis, its initiators and inhibitors, and the potential role of its main metabolic pathways in the treatment and prevention of various diseased states. We end the review with the current knowledge gaps in this area to provide direction for future research on ferroptosis.
Collapse
Affiliation(s)
- Chu Han
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China
| | - Yuanyuan Liu
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China
| | - Rongji Dai
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Nafissa Ismail
- Neuroimmunology, Stress and Endocrinology (NISE) Lab, School of Psychology, Faculty of Social Science, University of Ottawa, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Weijun Su
- School of Medicine, Nankai University, Tianjin, China
| | - Bo Li
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
71
|
Chen Y, Jakary A, Avadiappan S, Hess CP, Lupo JM. QSMGAN: Improved Quantitative Susceptibility Mapping using 3D Generative Adversarial Networks with increased receptive field. Neuroimage 2020; 207:116389. [PMID: 31760151 PMCID: PMC8081272 DOI: 10.1016/j.neuroimage.2019.116389] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/31/2019] [Accepted: 11/20/2019] [Indexed: 11/27/2022] Open
Abstract
Quantitative susceptibility mapping (QSM) is a powerful MRI technique that has shown great potential in quantifying tissue susceptibility in numerous neurological disorders. However, the intrinsic ill-posed dipole inversion problem greatly affects the accuracy of the susceptibility map. We propose QSMGAN: a 3D deep convolutional neural network approach based on a 3D U-Net architecture with increased receptive field of the input phase compared to the output and further refined the network using the WGAN with gradient penalty training strategy. Our method generates accurate QSM maps from single orientation phase maps efficiently and performs significantly better than traditional non-learning-based dipole inversion algorithms. The generalization capability was verified by applying the algorithm to an unseen pathology--brain tumor patients with radiation-induced cerebral microbleeds.
Collapse
Affiliation(s)
- Yicheng Chen
- From the UCSF/UC Berkeley Graduate Program in Bioengineering, University of California, San Francisco and Berkeley, CA, USA; From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Angela Jakary
- From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Sivakami Avadiappan
- From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Christopher P Hess
- From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Janine M Lupo
- From the UCSF/UC Berkeley Graduate Program in Bioengineering, University of California, San Francisco and Berkeley, CA, USA; From the Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
72
|
Vinayagamani S, Sheelakumari R, Sabarish S, Senthilvelan S, Ros R, Thomas B, Kesavadas C. Quantitative Susceptibility Mapping: Technical Considerations and Clinical Applications in Neuroimaging. J Magn Reson Imaging 2020; 53:23-37. [DOI: 10.1002/jmri.27058] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/28/2019] [Accepted: 12/30/2019] [Indexed: 12/12/2022] Open
Affiliation(s)
| | - R Sheelakumari
- Department of Imaging Sciences and Interventional Radiology Trivandrum India
| | - Sekar Sabarish
- Department of Imaging Sciences and Interventional Radiology Trivandrum India
| | | | - Roopa Ros
- Neurology, Sree Chitra Tirunal Institute for Medical Sciences and Technology Trivandrum India
| | - Bejoy Thomas
- Department of Imaging Sciences and Interventional Radiology Trivandrum India
| | | |
Collapse
|
73
|
Hanssen H, Prasuhn J, Heldmann M, Diesta CC, Domingo A, Göttlich M, Blood AJ, Rosales RL, Jamora RDG, Münte TF, Klein C, Brüggemann N. Imaging gradual neurodegeneration in a basal ganglia model disease. Ann Neurol 2019; 86:517-526. [DOI: 10.1002/ana.25566] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 07/29/2019] [Accepted: 07/29/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Henrike Hanssen
- Department of NeurologyUniversity Medical Center Schleswig‐Holstein, Campus Lübeck Lübeck Germany
- Institute of NeurogeneticsUniversity of Lübeck Lübeck Germany
| | - Jannik Prasuhn
- Department of NeurologyUniversity Medical Center Schleswig‐Holstein, Campus Lübeck Lübeck Germany
- Institute of NeurogeneticsUniversity of Lübeck Lübeck Germany
| | - Marcus Heldmann
- Department of NeurologyUniversity Medical Center Schleswig‐Holstein, Campus Lübeck Lübeck Germany
| | - Cid C. Diesta
- Asian Hospital and Medical Center, Filinvest Corporate City, Alabang Muntinlupa City the Philippines
| | - Aloysius Domingo
- Institute of NeurogeneticsUniversity of Lübeck Lübeck Germany
- Department of NeurologyMassachusetts General Hospital Boston MA
| | - Martin Göttlich
- Department of NeurologyUniversity Medical Center Schleswig‐Holstein, Campus Lübeck Lübeck Germany
| | - Anne J. Blood
- Mood and Motor Control LaboratoryMassachusetts General Hospital Charlestown MA
- Laboratory of Neuroimaging and GeneticsMassachusetts General Hospital Charlestown MA
- Department of Neurology and PsychiatryMassachusetts General Hospital Boston MA
- Martinos Center for Biomedical Imaging, Department of RadiologyMassachusetts General Hospital Charlestown MA
- Division of Child NeurologyBoston Children's Hospital Boston MA
| | - Raymond L. Rosales
- Department of Neurology and Psychiatry, Faculty of Medicine and SurgeryUniversity of Santo Tomas Manila the Philippines
| | - Roland D. G. Jamora
- Department of Neurosciences, College of Medicine–Philippine General HospitalUniversity of the Philippines Manila Manila the Philippines
| | - Thomas F. Münte
- Department of NeurologyUniversity Medical Center Schleswig‐Holstein, Campus Lübeck Lübeck Germany
| | - Christine Klein
- Institute of NeurogeneticsUniversity of Lübeck Lübeck Germany
| | - Norbert Brüggemann
- Department of NeurologyUniversity Medical Center Schleswig‐Holstein, Campus Lübeck Lübeck Germany
- Institute of NeurogeneticsUniversity of Lübeck Lübeck Germany
| |
Collapse
|
74
|
Bollmann S, Rasmussen KGB, Kristensen M, Blendal RG, Østergaard LR, Plocharski M, O'Brien K, Langkammer C, Janke A, Barth M. DeepQSM - using deep learning to solve the dipole inversion for quantitative susceptibility mapping. Neuroimage 2019; 195:373-383. [PMID: 30935908 DOI: 10.1016/j.neuroimage.2019.03.060] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 03/03/2019] [Accepted: 03/26/2019] [Indexed: 12/21/2022] Open
Abstract
Quantitative susceptibility mapping (QSM) is based on magnetic resonance imaging (MRI) phase measurements and has gained broad interest because it yields relevant information on biological tissue properties, predominantly myelin, iron and calcium in vivo. Thereby, QSM can also reveal pathological changes of these key components in widespread diseases such as Parkinson's disease, Multiple Sclerosis, or hepatic iron overload. While the ill-posed field-to-source-inversion problem underlying QSM is conventionally assessed by the means of regularization techniques, we trained a fully convolutional deep neural network - DeepQSM - to directly invert the magnetic dipole kernel convolution. DeepQSM learned the physical forward problem using purely synthetic data and is capable of solving the ill-posed field-to-source inversion on in vivo MRI phase data. The magnetic susceptibility maps reconstructed by DeepQSM enable identification of deep brain substructures and provide information on their respective magnetic tissue properties. In summary, DeepQSM can invert the magnetic dipole kernel convolution and delivers robust solutions to this ill-posed problem.
Collapse
Affiliation(s)
- Steffen Bollmann
- Centre for Advanced Imaging, The University of Queensland, Building 57 of University Dr, St Lucia, QLD, 4072, Brisbane, Australia.
| | | | - Mads Kristensen
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 7, 9000, Aalborg, Denmark
| | - Rasmus Guldhammer Blendal
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 7, 9000, Aalborg, Denmark
| | - Lasse Riis Østergaard
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 7, 9000, Aalborg, Denmark
| | - Maciej Plocharski
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 7, 9000, Aalborg, Denmark
| | - Kieran O'Brien
- Centre for Advanced Imaging, The University of Queensland, Building 57 of University Dr, St Lucia, QLD, 4072, Brisbane, Australia; Siemens Healthcare Pty Ltd, Brisbane, Australia
| | - Christian Langkammer
- Department of Neurology, Medical University of Graz, Auenbruggerplatz 22, 8036, Graz, Austria
| | - Andrew Janke
- Centre for Advanced Imaging, The University of Queensland, Building 57 of University Dr, St Lucia, QLD, 4072, Brisbane, Australia
| | - Markus Barth
- Centre for Advanced Imaging, The University of Queensland, Building 57 of University Dr, St Lucia, QLD, 4072, Brisbane, Australia
| |
Collapse
|
75
|
Detection of dentate nuclei abnormality in a patient with dentatorubral-pallidoluysian atrophy using the quantitative susceptibility mapping. J Neurol Sci 2019; 403:97-98. [PMID: 31247449 DOI: 10.1016/j.jns.2019.06.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/08/2019] [Accepted: 06/17/2019] [Indexed: 11/22/2022]
|
76
|
Lancione M, Donatelli G, Cecchi P, Cosottini M, Tosetti M, Costagli M. Echo-time dependency of quantitative susceptibility mapping reproducibility at different magnetic field strengths. Neuroimage 2019; 197:557-564. [PMID: 31075389 DOI: 10.1016/j.neuroimage.2019.05.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/10/2019] [Accepted: 05/02/2019] [Indexed: 12/13/2022] Open
Abstract
Quantitative Susceptibility Mapping (QSM) provides a way of measuring iron concentration and myelination non-invasively and has the potential of becoming a tool of paramount importance in the study of a host of different pathologies. However, several experimental factors and the physical properties of magnetic susceptibility (χ) can impair the reliability of QSM, and it is therefore essential to assess QSM reproducibility for repeated acquisitions and different field strength. In particular, it has recently been demonstrated that QSM measurements strongly depend on echo time (TE): the same tissue, measured on the same scanner, exhibits different apparent frequency shifts depending on the TE used. This study aims to assess the influence of TE on intra-scanner and inter-scanner reproducibility of QSM, by using MRI systems operating at 3T and 7T. To maximize intra-scanner reproducibility it is necessary to match the TEs of the acquisition protocol, but the application of this rule leads to inconsistent QSM values across scanners operating at different static magnetic field. This study however demonstrates that, provided a careful choice of acquisition parameters, and in particular by using TEs at 3T that are approximately 2.6 times longer than those at 7T, highly reproducible whole-brain χ maps can be achieved also across different scanners, which renders QSM a suitable technique for longitudinal follow-up in clinical settings and in multi-center studies.
Collapse
Affiliation(s)
| | - Graziella Donatelli
- IMAGO7 Foundation, Pisa, Italy; Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Paolo Cecchi
- Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | | | - Michela Tosetti
- IMAGO7 Foundation, Pisa, Italy; IRCCS Stella Maris, Pisa, Italy.
| | - Mauro Costagli
- IMAGO7 Foundation, Pisa, Italy; IRCCS Stella Maris, Pisa, Italy
| |
Collapse
|
77
|
Li X, Chen L, Kutten K, Ceritoglu C, Li Y, Kang N, Hsu JT, Qiao Y, Wei H, Liu C, Miller MI, Mori S, Yousem DM, van Zijl PCM, Faria AV. Multi-atlas tool for automated segmentation of brain gray matter nuclei and quantification of their magnetic susceptibility. Neuroimage 2019; 191:337-349. [PMID: 30738207 PMCID: PMC6464637 DOI: 10.1016/j.neuroimage.2019.02.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 02/03/2019] [Accepted: 02/06/2019] [Indexed: 01/09/2023] Open
Abstract
Quantification of tissue magnetic susceptibility using MRI offers a non-invasive measure of important tissue components in the brain, such as iron and myelin, potentially providing valuable information about normal and pathological conditions during aging. Despite many advances made in recent years on imaging techniques of quantitative susceptibility mapping (QSM), accurate and robust automated segmentation tools for QSM images that can help generate universal and sharable susceptibility measures in a biologically meaningful set of structures are still not widely available. In the present study, we developed an automated process to segment brain nuclei and quantify tissue susceptibility in these regions based on a susceptibility multi-atlas library, consisting of 10 atlases with T1-weighted images, gradient echo (GRE) magnitude images and QSM images of brains with different anatomic patterns. For each atlas in this library, 10 regions of interest in iron-rich deep gray matter structures that are better defined by QSM contrast were manually labeled, including caudate, putamen, globus pallidus internal/external, thalamus, pulvinar, subthalamic nucleus, substantia nigra, red nucleus and dentate nucleus in both left and right hemispheres. We then tested different pipelines using different combinations of contrast channels to bring the set of labels from the multi-atlases to each target brain and compared them with the gold standard manual delineation. The results showed that the segmentation accuracy using dual contrasts QSM/T1 pipeline outperformed other dual-contrast or single-contrast pipelines. The dice values of 0.77 ± 0.09 using the QSM/T1 multi-atlas pipeline rivaled with the segmentation reliability obtained from multiple evaluators with dice values of 0.79 ± 0.07 and gave comparable or superior performance in segmenting subcortical nuclei in comparison with standard FSL FIRST or recent multi-atlas package of volBrain. The segmentation performance of the QSM/T1 multi-atlas was further tested on QSM images acquired using different acquisition protocols and platforms and showed good reliability and reproducibility with average dice of 0.79 ± 0.08 to manual labels and 0.89 ± 0.04 in an inter-protocol manner. The extracted quantitative magnetic susceptibility values in the deep gray matter nuclei also correlated well between different protocols with inter-protocol correlation constants all larger than 0.97. Such reliability and performance was ultimately validated in an external dataset acquired at another study site with consistent susceptibility measures obtained using the QSM/T1 multi-atlas approach in comparison to those using manual delineation. In summary, we designed a susceptibility multi-atlas tool for automated and reliable segmentation of QSM images and for quantification of magnetic susceptibilities. It is publicly available through our cloud-based platform (www.mricloud.org). Further improvement on the performance of this multi-atlas tool is expected by increasing the number of atlases in the future.
Collapse
Affiliation(s)
- Xu Li
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA.
| | - Lin Chen
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA; Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | - Kwame Kutten
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD, USA
| | - Can Ceritoglu
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Yue Li
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ningdong Kang
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John T Hsu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ye Qiao
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hongjiang Wei
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, USA
| | - Chunlei Liu
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, USA
| | - Michael I Miller
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Susumu Mori
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David M Yousem
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter C M van Zijl
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Andreia V Faria
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
78
|
Chen L, Hua J, Ross CA, Cai S, van Zijl PC, Li X. Altered brain iron content and deposition rate in Huntington's disease as indicated by quantitative susceptibility MRI. J Neurosci Res 2019; 97:467-479. [PMID: 30489648 PMCID: PMC6367012 DOI: 10.1002/jnr.24358] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/08/2018] [Accepted: 11/08/2018] [Indexed: 12/14/2022]
Abstract
Altered brain iron content in the striatum of premanifest and manifest Huntington's disease (HD) has been reported. However, its natural history remains unclear. This study aims to investigate altered brain iron content in premanifest and early HD, and the iron deposition rate in these patients through a longitudinal one-year follow-up test, with quantitative magnetic susceptibility as an iron imaging marker. Twenty-four gene mutation carriers divided into three groups (further-from-onset, closer-to-onset and early HD) and 16 age-matched healthy controls were recruited at baseline, and of these, 14 carriers and 7 controls completed the one-year follow-up. Quantitative magnetic susceptibility and effective transverse relaxation rate ( R 2 ∗ ) were measured at 7.0 Tesla and correlated with atrophy and available clinical and cognitive measurements. Higher susceptibility values indicating higher iron content in the striatum and globus pallidus were only observed in closer-to-onset (N = 6, p < 0.05 in caudate and p < 0.01 in putamen) and early HD (N = 9, p < 0.05 in caudate and globus pallidus and p < 0.01 in putamen). Similar results were found by R 2 ∗ measurement. Such increases directly correlated with HD CAG-age product score and brain atrophy, but not with motor or cognitive scores. More importantly, a significantly higher iron deposition rate (11.9%/years in caudate and 6.1%/years in globus pallidus) was firstly observed in closer-to-onset premanifest HD and early HD as compared to the controls. These results suggest that monitoring brain iron may provide further insights into the pathophysiology of HD disease progression, and may provide a biomarker for clinical trials.
Collapse
Affiliation(s)
- Lin Chen
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
- Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Jun Hua
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
- Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Christopher A. Ross
- Department of Psychiatry, Division of Neurobiology, and Departments of Neurology, Neuroscience and Pharmacology, Johns Hopkins University, Baltimore, Maryland, United States
| | - Shuhui Cai
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | - Peter C.M. van Zijl
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
- Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Xu Li
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States
- Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
79
|
Mi Y, Gao X, Xu H, Cui Y, Zhang Y, Gou X. The Emerging Roles of Ferroptosis in Huntington's Disease. Neuromolecular Med 2019; 21:110-119. [PMID: 30600476 DOI: 10.1007/s12017-018-8518-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/08/2018] [Indexed: 12/11/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant and fatal neurodegenerative disorder, which is caused by an abnormal CAG repeat in the huntingtin gene. Despite its well-defined genetic origin, the molecular mechanisms of neuronal death are unclear yet, thus there are no effective strategies to block or postpone the process of HD. Ferroptosis, a recently identified iron-dependent cell death, attracts considerable attention due to its putative involvement in neurodegenerative diseases. Accumulative data suggest that ferroptosis is very likely to participate in HD, and inhibition of the molecules and signaling pathways involved in ferroptosis can significantly eliminate the symptoms and pathology of HD. This review first describes evidence for the close relevance of ferroptosis and HD in patients and mouse models, then summarizes advances for the mechanisms of ferroptosis involved in HD, finally outlines some therapeutic strategies targeted ferroptosis. Comprehensive understanding of the emerging roles of ferroptosis in the occurrence of HD will help us to explore effective therapies for slowing the progression of this disease.
Collapse
Affiliation(s)
- Yajing Mi
- Shaanxi Key Laboratory of Brain Disorders, and Department of Basic Medicine, Xi'an Medical University, Xi'an, 710021, China
| | - Xingchun Gao
- Shaanxi Key Laboratory of Brain Disorders, and Department of Basic Medicine, Xi'an Medical University, Xi'an, 710021, China
| | - Hao Xu
- Shaanxi Key Laboratory of Brain Disorders, and Department of Basic Medicine, Xi'an Medical University, Xi'an, 710021, China
| | - Yuanyuan Cui
- Shaanxi Key Laboratory of Brain Disorders, and Department of Basic Medicine, Xi'an Medical University, Xi'an, 710021, China
| | - Yuelin Zhang
- Shaanxi Key Laboratory of Brain Disorders, and Department of Basic Medicine, Xi'an Medical University, Xi'an, 710021, China.
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders, and Department of Basic Medicine, Xi'an Medical University, Xi'an, 710021, China.
| |
Collapse
|
80
|
Kwakye GF, Jiménez JA, Thomas MG, Kingsley BA, McIIvin M, Saito MA, Korley EM. Heterozygous huntingtin promotes cadmium neurotoxicity and neurodegeneration in striatal cells via altered metal transport and protein kinase C delta dependent oxidative stress and apoptosis signaling mechanisms. Neurotoxicology 2019; 70:48-61. [DOI: 10.1016/j.neuro.2018.10.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 10/22/2018] [Accepted: 10/31/2018] [Indexed: 12/20/2022]
|
81
|
Ladd ME, Bachert P, Meyerspeer M, Moser E, Nagel AM, Norris DG, Schmitter S, Speck O, Straub S, Zaiss M. Pros and cons of ultra-high-field MRI/MRS for human application. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2018; 109:1-50. [PMID: 30527132 DOI: 10.1016/j.pnmrs.2018.06.001] [Citation(s) in RCA: 321] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 06/06/2018] [Accepted: 06/07/2018] [Indexed: 05/08/2023]
Abstract
Magnetic resonance imaging and spectroscopic techniques are widely used in humans both for clinical diagnostic applications and in basic research areas such as cognitive neuroimaging. In recent years, new human MR systems have become available operating at static magnetic fields of 7 T or higher (≥300 MHz proton frequency). Imaging human-sized objects at such high frequencies presents several challenges including non-uniform radiofrequency fields, enhanced susceptibility artifacts, and higher radiofrequency energy deposition in the tissue. On the other side of the scale are gains in signal-to-noise or contrast-to-noise ratio that allow finer structures to be visualized and smaller physiological effects to be detected. This review presents an overview of some of the latest methodological developments in human ultra-high field MRI/MRS as well as associated clinical and scientific applications. Emphasis is given to techniques that particularly benefit from the changing physical characteristics at high magnetic fields, including susceptibility-weighted imaging and phase-contrast techniques, imaging with X-nuclei, MR spectroscopy, CEST imaging, as well as functional MRI. In addition, more general methodological developments such as parallel transmission and motion correction will be discussed that are required to leverage the full potential of higher magnetic fields, and an overview of relevant physiological considerations of human high magnetic field exposure is provided.
Collapse
Affiliation(s)
- Mark E Ladd
- Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine, University of Heidelberg, Heidelberg, Germany; Faculty of Physics and Astronomy, University of Heidelberg, Heidelberg, Germany; Erwin L. Hahn Institute for MRI, University of Duisburg-Essen, Essen, Germany.
| | - Peter Bachert
- Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Physics and Astronomy, University of Heidelberg, Heidelberg, Germany.
| | - Martin Meyerspeer
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria; MR Center of Excellence, Medical University of Vienna, Vienna, Austria.
| | - Ewald Moser
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria; MR Center of Excellence, Medical University of Vienna, Vienna, Austria.
| | - Armin M Nagel
- Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - David G Norris
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, Netherlands; Erwin L. Hahn Institute for MRI, University of Duisburg-Essen, Essen, Germany.
| | - Sebastian Schmitter
- Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Germany.
| | - Oliver Speck
- Department of Biomedical Magnetic Resonance, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; German Center for Neurodegenerative Diseases, Magdeburg, Germany; Center for Behavioural Brain Sciences, Magdeburg, Germany; Leibniz Institute for Neurobiology, Magdeburg, Germany.
| | - Sina Straub
- Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Moritz Zaiss
- High-Field Magnetic Resonance Center, Max-Planck-Institute for Biological Cybernetics, Tübingen, Germany.
| |
Collapse
|
82
|
Acosta-Cabronero J, Milovic C, Mattern H, Tejos C, Speck O, Callaghan MF. A robust multi-scale approach to quantitative susceptibility mapping. Neuroimage 2018; 183:7-24. [PMID: 30075277 PMCID: PMC6215336 DOI: 10.1016/j.neuroimage.2018.07.065] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/29/2018] [Accepted: 07/29/2018] [Indexed: 12/11/2022] Open
Abstract
Quantitative Susceptibility Mapping (QSM), best known as a surrogate for tissue iron content, is becoming a highly relevant MRI contrast for monitoring cellular and vascular status in aging, addiction, traumatic brain injury and, in general, a wide range of neurological disorders. In this study we present a new Bayesian QSM algorithm, named Multi-Scale Dipole Inversion (MSDI), which builds on the nonlinear Morphology-Enabled Dipole Inversion (nMEDI) framework, incorporating three additional features: (i) improved implementation of Laplace's equation to reduce the influence of background fields through variable harmonic filtering and subsequent deconvolution, (ii) improved error control through dynamic phase-reliability compensation across spatial scales, and (iii) scalewise use of the morphological prior. More generally, this new pre-conditioned QSM formalism aims to reduce the impact of dipole-incompatible fields and measurement errors such as flow effects, poor signal-to-noise ratio or other data inconsistencies that can lead to streaking and shadowing artefacts. In terms of performance, MSDI is the first algorithm to rank in the top-10 for all metrics evaluated in the 2016 QSM Reconstruction Challenge. It also demonstrated lower variance than nMEDI and more stable behaviour in scan-rescan reproducibility experiments for different MRI acquisitions at 3 and 7 Tesla. In the present work, we also explored new forms of susceptibility MRI contrast making explicit use of the differential information across spatial scales. Specifically, we show MSDI-derived examples of: (i) enhanced anatomical detail with susceptibility inversions from short-range dipole fields (hereby referred to as High-Pass Susceptibility Mapping or HPSM), (ii) high specificity to venous-blood susceptibilities for highly regularised HPSM (making a case for MSDI-based Venography or VenoMSDI), (iii) improved tissue specificity (and possibly statistical conditioning) for Macroscopic-Vessel Suppressed Susceptibility Mapping (MVSSM), and (iv) high spatial specificity and definition for HPSM-based Susceptibility-Weighted Imaging (HPSM-SWI) and related intensity projections.
Collapse
Affiliation(s)
- Julio Acosta-Cabronero
- Wellcome Centre for Human Neuroimaging, UCL Institute of Neurology, University College London, London, United Kingdom; German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
| | - Carlos Milovic
- Department of Electrical Engineering, Pontificia Universidad Catolica de Chile, Santiago, Chile; Biomedical Imaging Center, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Hendrik Mattern
- Department of Biomedical Magnetic Resonance, Institute of Experimental Physics, Otto von Guericke University, Magdeburg, Germany
| | - Cristian Tejos
- Department of Electrical Engineering, Pontificia Universidad Catolica de Chile, Santiago, Chile; Biomedical Imaging Center, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Oliver Speck
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany; Department of Biomedical Magnetic Resonance, Institute of Experimental Physics, Otto von Guericke University, Magdeburg, Germany; Center for Behavioural Brain Sciences, Magdeburg, Germany; Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Martina F Callaghan
- Wellcome Centre for Human Neuroimaging, UCL Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
83
|
Khattab TA, Fouda MMG, Allam AA, Othman SI, Bin‐Jumah M, Al‐Harbi HM, Rehan M. Selective Colorimetric Detection of Fe (III) Using Metallochromic Tannin‐Impregnated Silica Strips. ChemistrySelect 2018. [DOI: 10.1002/slct.201802506] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Tawfik A. Khattab
- Dyeing, Printing and Auxiliaries Department, Textile Industries Research DivisionNational Research Centre 33 El-Buhouth Street, Dokki Cairo 12622 Egypt
| | - Moustafa M. G. Fouda
- Pretreatrment and Finishing of Cellulosic Based Textiles Department Textile Industries Research DivisionNational Research Centre 33 El-Buhouth Street, Dokki Cairo 12622 Egypt
| | - Ahmed A. Allam
- Department of Zoology, Faculty of ScienceBeni-suef University Beni-suef 65211 Egypt
| | - Sarah I. Othman
- College of sciencePrincess Nourah bint Abdulrahman UniversityDepartment of biology Saudi Arabia
| | - May Bin‐Jumah
- College of sciencePrincess Nourah bint Abdulrahman UniversityDepartment of biology Saudi Arabia
| | - Hanan M. Al‐Harbi
- College of sciencePrincess Nourah bint Abdulrahman UniversityDepartment of biology Saudi Arabia
| | - Mohamed Rehan
- Pretreatrment and Finishing of Cellulosic Based Textiles Department Textile Industries Research DivisionNational Research Centre 33 El-Buhouth Street, Dokki Cairo 12622 Egypt
| |
Collapse
|
84
|
Mattern H, Sciarra A, Lüsebrink F, Acosta-Cabronero J, Speck O. Prospective motion correction improves high-resolution quantitative susceptibility mapping at 7T. Magn Reson Med 2018; 81:1605-1619. [PMID: 30298692 DOI: 10.1002/mrm.27509] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/12/2018] [Accepted: 08/06/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE Recent literature has shown the potential of high-resolution quantitative susceptibility mapping (QSM) with ultra-high field MRI for imaging the anatomy, the vasculature, and investigating their magnetostatic properties. Higher spatial resolutions, however, translate to longer scans resulting, therefore, in higher vulnerability to, and likelihood of, subject movement. We propose a gradient-recalled echo sequence with prospective motion correction (PMC) to address such limitation. METHODS Data from 4 subjects were acquired at 7T. The effect of small and large motion on QSM with and without PMC was assessed qualitatively and quantitatively. Full brain QSM and QSM-based venograms with up to 0.33 mm isotropic voxel size were reconstructed. RESULTS With PMC, motion artifacts in QSM and QSM-based venograms were largely eliminated, enabling-in both large- and small-amplitude motion regimes-accurate depiction of the cortex, vasculature, and other small anatomical structures that are often blurred as a result of head movement or indiscernible at lower image resolutions. Quantitative analyses demonstrated that uncorrected motion could bias regional susceptibility distributions, a trend that was greatly reduced with PMC. CONCLUSION Qualitatively, PMC prevented image degradation because of motion artifacts, providing highly detailed QSM images and venograms. Quantitatively, PMC increased the reproducibility of susceptibility measures.
Collapse
Affiliation(s)
- Hendrik Mattern
- Department of Biomedical Magnetic Resonance, Institute for Physics, Otto-von-Guericke-University, Magdeburg, Germany
| | - Alessandro Sciarra
- Department of Biomedical Magnetic Resonance, Institute for Physics, Otto-von-Guericke-University, Magdeburg, Germany
| | - Falk Lüsebrink
- Department of Biomedical Magnetic Resonance, Institute for Physics, Otto-von-Guericke-University, Magdeburg, Germany
| | - Julio Acosta-Cabronero
- Wellcome Centre for Human Neuroimaging, Institute of Neurology, University College London, London, United Kingdom.,German Center for Neurodegenerative Diseases, Magdeburg, Germany
| | - Oliver Speck
- Department of Biomedical Magnetic Resonance, Institute for Physics, Otto-von-Guericke-University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany.,Leibniz Institute for Neurobiology, Magdeburg, Germany
| |
Collapse
|
85
|
Structural Magnetic Resonance Imaging in Huntington's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 142:335-380. [PMID: 30409258 DOI: 10.1016/bs.irn.2018.09.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder, caused by expansion of the CAG repeat in the huntingtin gene. HD is characterized clinically by progressive motor, cognitive and neuropsychiatric symptoms. There are currently no disease modifying treatments available for HD, and there is a great need for biomarkers to monitor disease progression and identify new targets for therapeutic intervention. Neuroimaging techniques provide a powerful tool for assessing disease pathology and progression in premanifest stages, before the onset of overt motor symptoms. Structural magnetic resonance imaging (MRI) is non-invasive imaging techniques which have been employed to study structural and microstructural changes in premanifest and manifest HD gene carriers. This chapter described structural imaging techniques and analysis methods employed across HD MRI studies. Current evidence for structural MRI abnormalities in HD, and associations between atrophy, structural white matter changes, iron deposition and clinical performance are discussed; together with the use of structural MRI measures as a diagnostic tool, to assess longitudinal changes, and as potential biomarkers and endpoints for clinical trials.
Collapse
|
86
|
Niu L, Ye C, Sun Y, Peng T, Yang S, Wang W, Li H. Mutant huntingtin induces iron overload via up-regulating IRP1 in Huntington's disease. Cell Biosci 2018; 8:41. [PMID: 30002810 PMCID: PMC6033216 DOI: 10.1186/s13578-018-0239-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023] Open
Abstract
Background Iron accumulation in basal ganglia accompanies neuronal loss in Huntington’s disease (HD) patients and mouse disease models. Disruption of HD brain iron homeostasis occurs before the onset of clinical signs. Therefore, investigating the mechanism of iron accumulation is essential to understanding its role in disease pathogenesis. Methods N171-82Q HD transgenic mice brain iron was detected by using Diaminobenzidine-enhanced Perls’ stain. Iron homeostatic proteins including iron response protein 1 (IRP1), transferrin (Tf), ferritin and transferrin receptor (TfR) were determined by using western blotting and immunohistochemistry, and their relative expression levels of RNA were measured by RT-PCR in both N171-82Q HD transgenic mice and HEK293 cells expressing N-terminal of huntingtin. Results Iron was increased in striatum and cortex of N171-82Q HD transgenic mice. Analysis of iron homeostatic proteins revealed increased expression of IRP1, Tf, ferritin and TfR in N171-82Q mice striatum and cortex. The same results were obtained in HEK293 cells expressing N-terminal of mutant huntingtin containing 160 CAG repeats. Conclusion We conclude that mutant huntingtin may cause abnormal iron homeostatic pathways by increasing IRP1 expression in Huntington’s disease, suggesting potential therapeutic target.
Collapse
Affiliation(s)
- Li Niu
- 1Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030 People's Republic of China
| | - Cuifang Ye
- 1Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030 People's Republic of China.,2Institute for Brain Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 People's Republic of China
| | - Yun Sun
- 1Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030 People's Republic of China
| | - Ting Peng
- 1Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030 People's Republic of China.,2Institute for Brain Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 People's Republic of China.,3Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030 People's Republic of China
| | - Shiming Yang
- 1Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030 People's Republic of China
| | - Weixi Wang
- 1Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030 People's Republic of China
| | - He Li
- 1Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030 People's Republic of China.,2Institute for Brain Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 People's Republic of China.,3Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030 People's Republic of China
| |
Collapse
|
87
|
Keuken MC, Isaacs BR, Trampel R, van der Zwaag W, Forstmann BU. Visualizing the Human Subcortex Using Ultra-high Field Magnetic Resonance Imaging. Brain Topogr 2018; 31:513-545. [PMID: 29497874 PMCID: PMC5999196 DOI: 10.1007/s10548-018-0638-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/28/2018] [Indexed: 12/15/2022]
Abstract
With the recent increased availability of ultra-high field (UHF) magnetic resonance imaging (MRI), substantial progress has been made in visualizing the human brain, which can now be done in extraordinary detail. This review provides an extensive overview of the use of UHF MRI in visualizing the human subcortex for both healthy and patient populations. The high inter-subject variability in size and location of subcortical structures limits the usability of atlases in the midbrain. Fortunately, the combined results of this review indicate that a large number of subcortical areas can be visualized in individual space using UHF MRI. Current limitations and potential solutions of UHF MRI for visualizing the subcortex are also discussed.
Collapse
Affiliation(s)
- M C Keuken
- Integrative Model-Based Cognitive Neuroscience Research Unit, University of Amsterdam, Postbus 15926, 1001NK, Amsterdam, The Netherlands.
- Cognitive Psychology Unit, Institute of Psychology and Leiden Institute for Brain and Cognition, Leiden University, Leiden, The Netherlands.
| | - B R Isaacs
- Integrative Model-Based Cognitive Neuroscience Research Unit, University of Amsterdam, Postbus 15926, 1001NK, Amsterdam, The Netherlands
- Maastricht University Medical Center, Maastricht, The Netherlands
| | - R Trampel
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | | | - B U Forstmann
- Integrative Model-Based Cognitive Neuroscience Research Unit, University of Amsterdam, Postbus 15926, 1001NK, Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| |
Collapse
|
88
|
Agrawal S, Fox J, Thyagarajan B, Fox JH. Brain mitochondrial iron accumulates in Huntington's disease, mediates mitochondrial dysfunction, and can be removed pharmacologically. Free Radic Biol Med 2018; 120:317-329. [PMID: 29625173 PMCID: PMC5940499 DOI: 10.1016/j.freeradbiomed.2018.04.002] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/01/2018] [Accepted: 04/02/2018] [Indexed: 01/18/2023]
Abstract
Mitochondrial bioenergetic dysfunction is involved in neurodegeneration in Huntington's disease (HD). Iron is critical for normal mitochondrial bioenergetics but can also contribute to pathogenic oxidation. The accumulation of iron in the brain occurs in mouse models and in human HD. Yet the role of mitochondria-related iron dysregulation as a contributor to bioenergetic pathophysiology in HD is unclear. We demonstrate here that human HD and mouse model HD (12-week R6/2 and 12-month YAC128) brains accumulated mitochondrial iron and showed increased expression of iron uptake protein mitoferrin 2 and decreased iron-sulfur cluster synthesis protein frataxin. Mitochondria-enriched fractions from mouse HD brains had deficits in membrane potential and oxygen uptake and increased lipid peroxidation. In addition, the membrane-permeable iron-selective chelator deferiprone (1 μM) rescued these effects ex-vivo, whereas hydrophilic iron and copper chelators did not. A 10-day oral deferiprone treatment in 9-week R6/2 HD mice indicated that deferiprone removed mitochondrial iron, restored mitochondrial potentials, decreased lipid peroxidation, and improved motor endurance. Neonatal iron supplementation potentiates neurodegeneration in mouse models of HD by unknown mechanisms. We found that neonatal iron supplementation increased brain mitochondrial iron accumulation and potentiated markers of mitochondrial dysfunction in HD mice. Therefore, bi-directional manipulation of mitochondrial iron can potentiate and protect against markers of mouse HD. Our findings thus demonstrate the significance of iron as a mediator of mitochondrial dysfunction and injury in mouse models of human HD and suggest that targeting the iron-mitochondrial pathway may be protective.
Collapse
Affiliation(s)
- Sonal Agrawal
- Department of Veterinary Sciences, University of Wyoming, Laramie, WY 82070, United States
| | - Julia Fox
- Department of Veterinary Sciences, University of Wyoming, Laramie, WY 82070, United States
| | | | - Jonathan H Fox
- Department of Veterinary Sciences, University of Wyoming, Laramie, WY 82070, United States.
| |
Collapse
|
89
|
Halefoglu AM, Yousem DM. Susceptibility weighted imaging: Clinical applications and future directions. World J Radiol 2018; 10:30-45. [PMID: 29849962 PMCID: PMC5971274 DOI: 10.4329/wjr.v10.i4.30] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 04/08/2018] [Accepted: 04/20/2018] [Indexed: 02/06/2023] Open
Abstract
Susceptibility weighted imaging (SWI) is a recently developed magnetic resonance imaging (MRI) technique that is increasingly being used to narrow the differential diagnosis of many neurologic disorders. It exploits the magnetic susceptibility differences of various compounds including deoxygenated blood, blood products, iron and calcium, thus enabling a new source of contrast in MR. In this review, we illustrate its basic clinical applications in neuroimaging. SWI is based on a fully velocity-compensated, high-resolution, three dimensional gradient-echo sequence using magnitude and phase images either separately or in combination with each other, in order to characterize brain tissue. SWI is particularly useful in the setting of trauma and acute neurologic presentations suggestive of stroke, but can also characterize occult low-flow vascular malformations, cerebral microbleeds, intracranial calcifications, neurodegenerative diseases and brain tumors. Furthermore, advanced MRI post-processing technique with quantitative susceptibility mapping, enables detailed anatomical differentiation based on quantification of brain iron from SWI raw data.
Collapse
Affiliation(s)
- Ahmet Mesrur Halefoglu
- Department of Radiology, Sisli Hamidiye Etfal Training and Research Hospital, University of Health Sciences, Istanbul 34371, Turkey
| | - David Mark Yousem
- Division of Neuroradiology, Department of Radiology, Johns Hopkins Medical Institution, Baltimore, MI 21287, United States
| |
Collapse
|
90
|
关 基, 冯 衍. [Quantitative magnetic resonance imaging of brain iron deposition: comparison between quantitative susceptibility mapping and transverse relaxation rate (R2*) mapping]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:305-311. [PMID: 29643036 PMCID: PMC6744171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Indexed: 10/15/2023]
Abstract
OBJECTIVE To evaluate the accuracy and sensitivity of quantitative susceptibility mapping (QSM) and transverse relaxation rate (R2*) mapping in the measurement of brain iron deposition. METHODS Super paramagnetic iron oxide (SPIO) phantoms and mouse models of Parkinson's disease (PD) related to iron deposition in the substantia nigra (SN) underwent 7.0 T magnetic resonance (MR) scans (Bruker, 70/16) with a multi-echo 3D gradient echo sequence, and the acquired data were processed to obtain QSM and R2*. Linear regression analysis was performed for susceptibility and R2* in the SPIO phantoms containing 5 SPIO concentrations (30, 15, 7.5, 3.75 and 1.875 µg/mL) to evaluate the accuracy of QSM and R2* in quantitative iron analysis. The sensitivities of QSM and R2* mapping in quantitative detection of brain iron deposition were assessed using mouse models of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahy-dropyridine (MPTP) in comparison with the control mice. RESULTS In SPIO phantoms, QSM provided a higher accuracy than R2* mapping and their goodness-of-fit coefficients (R2) were 0.98 and 0.89, respectively. In the mouse models of PD and control mice, the susceptibility of the SN was significantly higher in the PD models (5.19∓1.58 vs 2.98∓0.88, n=5; P<0.05), while the R2* values were similar between the two groups (20.22∓0.94 vs 19.74∓1.75; P=0.60). CONCLUSION QSM allows more accurate and sensitive detection of brain iron deposition than R2*, and the susceptibility derived by QSM can be a potentially useful biomarker for studying PD.
Collapse
Affiliation(s)
- 基景 关
- />南方医科大学生物医学工程学院,广东 广州 510515School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - 衍秋 冯
- />南方医科大学生物医学工程学院,广东 广州 510515School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
91
|
关 基, 冯 衍. [Quantitative magnetic resonance imaging of brain iron deposition: comparison between quantitative susceptibility mapping and transverse relaxation rate (R2*) mapping]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:305-311. [PMID: 29643036 PMCID: PMC6744171 DOI: 10.3969/j.issn.1673-4254.2018.03.10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVE To evaluate the accuracy and sensitivity of quantitative susceptibility mapping (QSM) and transverse relaxation rate (R2*) mapping in the measurement of brain iron deposition. METHODS Super paramagnetic iron oxide (SPIO) phantoms and mouse models of Parkinson's disease (PD) related to iron deposition in the substantia nigra (SN) underwent 7.0 T magnetic resonance (MR) scans (Bruker, 70/16) with a multi-echo 3D gradient echo sequence, and the acquired data were processed to obtain QSM and R2*. Linear regression analysis was performed for susceptibility and R2* in the SPIO phantoms containing 5 SPIO concentrations (30, 15, 7.5, 3.75 and 1.875 µg/mL) to evaluate the accuracy of QSM and R2* in quantitative iron analysis. The sensitivities of QSM and R2* mapping in quantitative detection of brain iron deposition were assessed using mouse models of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahy-dropyridine (MPTP) in comparison with the control mice. RESULTS In SPIO phantoms, QSM provided a higher accuracy than R2* mapping and their goodness-of-fit coefficients (R2) were 0.98 and 0.89, respectively. In the mouse models of PD and control mice, the susceptibility of the SN was significantly higher in the PD models (5.19∓1.58 vs 2.98∓0.88, n=5; P<0.05), while the R2* values were similar between the two groups (20.22∓0.94 vs 19.74∓1.75; P=0.60). CONCLUSION QSM allows more accurate and sensitive detection of brain iron deposition than R2*, and the susceptibility derived by QSM can be a potentially useful biomarker for studying PD.
Collapse
Affiliation(s)
- 基景 关
- />南方医科大学生物医学工程学院,广东 广州 510515School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - 衍秋 冯
- />南方医科大学生物医学工程学院,广东 广州 510515School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
92
|
van Bergen JMG, Li X, Quevenco FC, Gietl AF, Treyer V, Meyer R, Buck A, Kaufmann PA, Nitsch RM, van Zijl PCM, Hock C, Unschuld PG. Simultaneous quantitative susceptibility mapping and Flutemetamol-PET suggests local correlation of iron and β-amyloid as an indicator of cognitive performance at high age. Neuroimage 2018; 174:308-316. [PMID: 29548847 DOI: 10.1016/j.neuroimage.2018.03.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 02/17/2018] [Accepted: 03/10/2018] [Indexed: 12/11/2022] Open
Abstract
The accumulation of β-amyloid plaques is a hallmark of Alzheimer's disease (AD), and recently published data suggest that increased brain iron burden may reflect pathologies that synergistically contribute to the development of cognitive dysfunction. While preclinical disease stages are considered most promising for therapeutic intervention, the link between emerging AD-pathology and earliest clinical symptoms remains largely unclear. In the current study we therefore investigated local correlations between iron and β-amyloid plaques, and their possible association with cognitive performance in healthy older adults. 116 older adults (mean age 75 ± 7.4 years) received neuropsychological testing to calculate a composite cognitive score of performance in episodic memory, executive functioning, attention, language and communication. All participants were scanned on a combined PET-MRI instrument and were administered T1-sequences for anatomical mapping, quantitative susceptibility mapping (QSM) for assessing iron, and 18F-Flutemetamol-PET for estimating β-amyloid plaque load. Biological parametric mapping (BPM) was used to generate masks indicating voxels with significant (p < 0.05) correlation between susceptibility and 18F-Flutemetamol-SUVR. We found a bilateral pattern of clusters characterized by a statistical relationship between magnetic susceptibility and 18F-Flutemetamol-SUVR, indicating local correlations between iron and β-amyloid plaque deposition. For two bilateral clusters, located in the frontal and temporal cortex, significant relationships (p<0.05) between local β-amyloid and the composite cognitive performance score could be observed. No relationship between whole-cortex β-amyloid plaque load and cognitive performance was observable. Our data suggest that the local correlation of β-amyloid plaque load and iron deposition may provide relevant information regarding cognitive performance of healthy older adults. Further studies are needed to clarify pathological correlates of the local interaction of β-amyloid, iron and other causes of altered magnetic susceptibility.
Collapse
Affiliation(s)
- J M G van Bergen
- Institute for Regenerative Medicine, University of Zurich, Switzerland.
| | - X Li
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - F C Quevenco
- Institute for Regenerative Medicine, University of Zurich, Switzerland
| | - A F Gietl
- Institute for Regenerative Medicine, University of Zurich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Switzerland
| | - V Treyer
- Institute for Regenerative Medicine, University of Zurich, Switzerland; Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - R Meyer
- Institute for Regenerative Medicine, University of Zurich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Switzerland
| | - A Buck
- Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - P A Kaufmann
- Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - R M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Switzerland
| | - P C M van Zijl
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - C Hock
- Institute for Regenerative Medicine, University of Zurich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Switzerland
| | - P G Unschuld
- Institute for Regenerative Medicine, University of Zurich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Switzerland
| |
Collapse
|
93
|
Kee Y, Liu Z, Zhou L, Dimov A, Cho J, de Rochefort L, Seo JK, Wang Y. Quantitative Susceptibility Mapping (QSM) Algorithms: Mathematical Rationale and Computational Implementations. IEEE Trans Biomed Eng 2018; 64:2531-2545. [PMID: 28885147 DOI: 10.1109/tbme.2017.2749298] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Quantitative susceptibility mapping (QSM) solves the magnetic field-to-magnetization (tissue susceptibility) inverse problem under conditions of noisy and incomplete field data acquired using magnetic resonance imaging. Therefore, sophisticated algorithms are necessary to treat the ill-posed nature of the problem and are reviewed here. The forward problem is typically presented as an integral form, where the field is the convolution of the dipole kernel and tissue susceptibility distribution. This integral form can be equivalently written as a partial differential equation (PDE). Algorithmic challenges are to reduce streaking and shadow artifacts characterized by the fundamental solution of the PDE. Bayesian maximum a posteriori estimation can be employed to solve the inverse problem, where morphological and relevant biomedical knowledge (specific to the imaging situation) are used as priors. As the cost functions in Bayesian QSM framework are typically convex, solutions can be robustly computed using a gradient-based optimization algorithm. Moreover, one can not only accelerate Bayesian QSM, but also increase its effectiveness at reducing shadows using prior knowledge based preconditioners. Improving the efficiency of QSM is under active development, and a rigorous analysis of preconditioning needs to be carried out for further investigation.Quantitative susceptibility mapping (QSM) solves the magnetic field-to-magnetization (tissue susceptibility) inverse problem under conditions of noisy and incomplete field data acquired using magnetic resonance imaging. Therefore, sophisticated algorithms are necessary to treat the ill-posed nature of the problem and are reviewed here. The forward problem is typically presented as an integral form, where the field is the convolution of the dipole kernel and tissue susceptibility distribution. This integral form can be equivalently written as a partial differential equation (PDE). Algorithmic challenges are to reduce streaking and shadow artifacts characterized by the fundamental solution of the PDE. Bayesian maximum a posteriori estimation can be employed to solve the inverse problem, where morphological and relevant biomedical knowledge (specific to the imaging situation) are used as priors. As the cost functions in Bayesian QSM framework are typically convex, solutions can be robustly computed using a gradient-based optimization algorithm. Moreover, one can not only accelerate Bayesian QSM, but also increase its effectiveness at reducing shadows using prior knowledge based preconditioners. Improving the efficiency of QSM is under active development, and a rigorous analysis of preconditioning needs to be carried out for further investigation.
Collapse
Affiliation(s)
- Youngwook Kee
- Department of Radiology, Weill Cornell Medical College, New York, USA
| | - Zhe Liu
- Department of Biomedical Engineering, Cornell University, Ithaca, USA
| | - Liangdong Zhou
- Department of Radiology, Weill Cornell Medical College, New York, USA
| | - Alexey Dimov
- Department of Biomedical Engineering, Cornell University, Ithaca, USA
| | - Junghun Cho
- Department of Biomedical Engineering, Cornell University, Ithaca, USA
| | - Ludovic de Rochefort
- Center for Magnetic Resonance in Biology and Medicine, UMR CNRS 7339, Aix-Marseille University, 13284 Marseille, France
| | - Jin Keun Seo
- Department of Computational Science and Engineering, Yonsei University, Seoul, South Korea
| | - Yi Wang
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
94
|
He K, Qi F, Guo C, Zhan S, Xu H, Liu J, Yang X. Movement deficits and neuronal loss in basal ganglia in TRPC1 deficient mice. Oncotarget 2018; 7:69337-69346. [PMID: 27738307 PMCID: PMC5342481 DOI: 10.18632/oncotarget.12567] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/03/2016] [Indexed: 11/25/2022] Open
Abstract
Transient receptor potential cation (TRPC) channel proteins are abundantly expressed in brain. However, the functions of these TRPC proteins such as TRPC1 are largely unclear. In this study, we reported that TRPC1 deficiency caused movement disorder as measured by swimming test, modified open field test and sunflower seeds eating test. Immunofluorescent staining showed significant loss of both NeuN-positive cells and tyrosine hydroxylase (TH) -positive cells in the caudate putamen (CPu), the external globus pallidus (GPe), and the substantia nigra pars reticulata (SNr) in 5-month-old TRPC1 knockout mice (TRPC1-/-) compared to the wild type (WT) mice. TUNEL staining further revealed that TUNEL-positive cells were significantly increased in the CPu, GPe, and SNr of TRPC1-/- mice. Taken together, these data suggests that TRPC1 is involved in the control of motor function by inhibiting the apoptosis of neuronal cells of basal ganglia.
Collapse
Affiliation(s)
- Kaiwu He
- College of Pharmacy, Jinan University, Guangzhou, China.,Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Fei Qi
- Department of Respiratory Medicine, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Chunni Guo
- Department of Neurology, ShanghaiFirst People's HospitalAffiliated toShanghai Jiaotong University, Shanghai, China
| | - Shuqin Zhan
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Hua Xu
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Jianjun Liu
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| |
Collapse
|
95
|
Gillen KM, Mubarak M, Nguyen TD, Pitt D. Significance and In Vivo Detection of Iron-Laden Microglia in White Matter Multiple Sclerosis Lesions. Front Immunol 2018. [PMID: 29515576 PMCID: PMC5826076 DOI: 10.3389/fimmu.2018.00255] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Microglia are resident immune cells that fulfill protective and homeostatic functions in the central nervous system (CNS) but may also promote neurotoxicity in the aged brain and in chronic disease. In multiple sclerosis (MS), an autoimmune demyelinating disease of the CNS, microglia and macrophages contribute to the development of white matter lesions through myelin phagocytosis, and possibly to disease progression through diffuse activation throughout myelinated white matter. In this review, we discuss an additional compartment of myeloid cell activation in MS, i.e., the rim and normal adjacent white matter of chronic active lesions. In chronic active lesions, microglia and macrophages may contain high amounts of iron, express markers of proinflammatory polarization, are activated for an extended period of time (years), and drive chronic tissue damage. Iron-positive myeloid cells can be visualized and quantified with quantitative susceptibility mapping (QSM), a magnetic resonance imaging technique. Thus, QSM has potential as an in vivo biomarker for chronic inflammatory activity in established white matter MS lesions. Reducing chronic inflammation associated with iron accumulation using existing or novel MS therapies may impact disease severity and progression.
Collapse
Affiliation(s)
- Kelly M Gillen
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Mayyan Mubarak
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| | - Thanh D Nguyen
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - David Pitt
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
96
|
Vela D. Hepcidin, an emerging and important player in brain iron homeostasis. J Transl Med 2018; 16:25. [PMID: 29415739 PMCID: PMC5803919 DOI: 10.1186/s12967-018-1399-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/31/2018] [Indexed: 02/08/2023] Open
Abstract
Hepcidin is emerging as a new important factor in brain iron homeostasis. Studies suggest that there are two sources of hepcidin in the brain; one is local and the other comes from the circulation. Little is known about the molecular mediators of local hepcidin expression, but inflammation and iron-load have been shown to induce hepcidin expression in the brain. The most important source of hepcidin in the brain are glial cells. Role of hepcidin in brain functions has been observed during neuronal iron-load and brain hemorrhage, where secretion of abundant hepcidin is related with the severity of brain damage. This damage can be reversed by blocking systemic and local hepcidin secretion. Studies have yet to unveil its role in other brain conditions, but the rationale exists, since these conditions are characterized by overexpression of the factors that stimulate brain hepcidin expression, such as inflammation, hypoxia and iron-overload.
Collapse
Affiliation(s)
- Driton Vela
- Department of Physiology, Faculty of Medicine, University of Prishtina, Martyr's Boulevard n.n., 10000, Prishtina, Kosova.
| |
Collapse
|
97
|
van Bergen JMG, Li X, Quevenco FC, Gietl AF, Treyer V, Leh SE, Meyer R, Buck A, Kaufmann PA, Nitsch RM, van Zijl PCM, Hock C, Unschuld PG. Low cortical iron and high entorhinal cortex volume promote cognitive functioning in the oldest-old. Neurobiol Aging 2017; 64:68-75. [PMID: 29351872 DOI: 10.1016/j.neurobiolaging.2017.12.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 11/09/2017] [Accepted: 12/13/2017] [Indexed: 10/18/2022]
Abstract
The aging brain is characterized by an increased presence of neurodegenerative and vascular pathologies. However, there is substantial variation regarding the relationship between an individual's pathological burden and resulting cognitive impairment. To identify correlates of preserved cognitive functioning at highest age, the relationship between β-amyloid plaque load, presence of small vessel cerebrovascular disease (SVCD), iron-burden, and brain atrophy was investigated. Eighty cognitively unimpaired participants (44 oldest-old, aged 85-96 years; 36 younger-old, aged 55-80 years) were scanned by integrated positron emission tomography-magnetic resonance imaging for assessing beta regional amyloid plaque load (18F-flutemetamol), white matter hyperintensities as an indicator of SVCD (fluid-attenuated inversion recovery-magnetic resonance imaging), and iron load (quantitative susceptibility mapping). For the oldest-old group, lower cortical volume, increased β-amyloid plaque load, prevalence of SVCD, and lower cognitive performance in the normal range were found. However, compared to normal-old, cortical iron burden was lower in the oldest-old. Moreover, only in the oldest-old, entorhinal cortex volume positively correlated with β-amyloid plaque load. Our data thus indicate that the co-occurrence of aging-associated neuropathologies with reduced quantitative susceptibility mapping measures of cortical iron load constitutes a lower vulnerability to cognitive loss.
Collapse
Affiliation(s)
- Jiri M G van Bergen
- Institute for Regenerative Medicine (IREM), University of Zürich, Zürich, Switzerland.
| | - Xu Li
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Frances C Quevenco
- Institute for Regenerative Medicine (IREM), University of Zürich, Zürich, Switzerland
| | - Anton F Gietl
- Institute for Regenerative Medicine (IREM), University of Zürich, Zürich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zürich, Zürich, Switzerland
| | - Valerie Treyer
- Institute for Regenerative Medicine (IREM), University of Zürich, Zürich, Switzerland; Department of Nuclear Medicine, University Hospital Zürich and University of Zürich, Zürich, Switzerland
| | - Sandra E Leh
- Hospital for Psychogeriatric Medicine, University of Zürich, Zürich, Switzerland
| | - Rafael Meyer
- Institute for Regenerative Medicine (IREM), University of Zürich, Zürich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zürich, Zürich, Switzerland
| | - Alfred Buck
- Department of Nuclear Medicine, University Hospital Zürich and University of Zürich, Zürich, Switzerland
| | - Philipp A Kaufmann
- Department of Nuclear Medicine, University Hospital Zürich and University of Zürich, Zürich, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine (IREM), University of Zürich, Zürich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zürich, Zürich, Switzerland
| | - Peter C M van Zijl
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Christoph Hock
- Institute for Regenerative Medicine (IREM), University of Zürich, Zürich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zürich, Zürich, Switzerland
| | - Paul G Unschuld
- Institute for Regenerative Medicine (IREM), University of Zürich, Zürich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zürich, Zürich, Switzerland
| |
Collapse
|
98
|
Wang Y, Spincemaille P, Liu Z, Dimov A, Deh K, Li J, Zhang Y, Yao Y, Gillen KM, Wilman AH, Gupta A, Tsiouris AJ, Kovanlikaya I, Chiang GCY, Weinsaft JW, Tanenbaum L, Chen W, Zhu W, Chang S, Lou M, Kopell BH, Kaplitt MG, Devos D, Hirai T, Huang X, Korogi Y, Shtilbans A, Jahng GH, Pelletier D, Gauthier SA, Pitt D, Bush AI, Brittenham GM, Prince MR. Clinical quantitative susceptibility mapping (QSM): Biometal imaging and its emerging roles in patient care. J Magn Reson Imaging 2017; 46:951-971. [PMID: 28295954 PMCID: PMC5592126 DOI: 10.1002/jmri.25693] [Citation(s) in RCA: 206] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/10/2017] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Quantitative susceptibility mapping (QSM) has enabled magnetic resonance imaging (MRI) of tissue magnetic susceptibility to advance from simple qualitative detection of hypointense blooming artifacts to precise quantitative measurement of spatial biodistributions. QSM technology may be regarded to be sufficiently developed and validated to warrant wide dissemination for clinical applications of imaging isotropic susceptibility, which is dominated by metals in tissue, including iron and calcium. These biometals are highly regulated as vital participants in normal cellular biochemistry, and their dysregulations are manifested in a variety of pathologic processes. Therefore, QSM can be used to assess important tissue functions and disease. To facilitate QSM clinical translation, this review aims to organize pertinent information for implementing a robust automated QSM technique in routine MRI practice and to summarize available knowledge on diseases for which QSM can be used to improve patient care. In brief, QSM can be generated with postprocessing whenever gradient echo MRI is performed. QSM can be useful for diseases that involve neurodegeneration, inflammation, hemorrhage, abnormal oxygen consumption, substantial alterations in highly paramagnetic cellular iron, bone mineralization, or pathologic calcification; and for all disorders in which MRI diagnosis or surveillance requires contrast agent injection. Clinicians may consider integrating QSM into their routine imaging practices by including gradient echo sequences in all relevant MRI protocols. LEVEL OF EVIDENCE 1 Technical Efficacy: Stage 5 J. Magn. Reson. Imaging 2017;46:951-971.
Collapse
Affiliation(s)
- Yi Wang
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
- Department of Biomedical Engineering, Ithaca, NY, USA
| | | | - Zhe Liu
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
- Department of Biomedical Engineering, Ithaca, NY, USA
| | - Alexey Dimov
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
- Department of Biomedical Engineering, Ithaca, NY, USA
| | - Kofi Deh
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | - Jianqi Li
- Department of Physics, East China Normal University, Shanghai, China
| | - Yan Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Yihao Yao
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Kelly M. Gillen
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | - Alan H. Wilman
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Ajay Gupta
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | | | - Ilhami Kovanlikaya
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | | | - Jonathan W. Weinsaft
- Division of Cardiology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | - Weiwei Chen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Shixin Chang
- Department of Radiology, Yueyang Hospital of Integrated Traditional Chinese & Western Medicine, Shanghai, China
| | - Min Lou
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Brian H. Kopell
- Department of Neurosurgery, Mount Sinai Hospital, New York, NY, USA
| | - Michael G. Kaplitt
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY, USA
| | - David Devos
- Department of Medical Pharmacology, University of Lille, Lille, France
- Department of Neurology and Movement Disorders, University of Lille, Lille, France
- Department of Toxicology, Public Health and Environment, University of Lille, Lille, France
- INSERM U1171, University of Lille, Lille, France
| | - Toshinori Hirai
- Department of Radiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Xuemei Huang
- Department of Neurology, Penn State University-Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Pharmacology, Penn State University-Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Neurosurgery, Penn State University-Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Radiology, Penn State University-Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Yukunori Korogi
- Department of Radiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Alexander Shtilbans
- Department of Neurology, Hospital for Special Surgery, New York, NY, USA
- Parkinson's Disease and Movement Disorder Institute, Weill Cornell Medical College, New York, NY, USA
| | - Geon-Ho Jahng
- Department of Radiology, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, South Korea
| | - Daniel Pelletier
- Department of Neurology, Department of Neurology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Susan A. Gauthier
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY, USA
| | - David Pitt
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
| | - Ashley I. Bush
- Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, AUS
| | - Gary M. Brittenham
- Department of Pediatrics, Columbia University, Children's Hospital of New York, New York, NY, USA
| | - Martin R. Prince
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
99
|
Fang J, Bao L, Li X, van Zijl PC, Chen Z. Background field removal using a region adaptive kernel for quantitative susceptibility mapping of human brain. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2017; 281:130-140. [PMID: 28595120 PMCID: PMC5846686 DOI: 10.1016/j.jmr.2017.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 05/07/2017] [Accepted: 05/09/2017] [Indexed: 06/07/2023]
Abstract
Background field removal is an important MR phase preprocessing step for quantitative susceptibility mapping (QSM). It separates the local field induced by tissue magnetic susceptibility sources from the background field generated by sources outside a region of interest, e.g. brain, such as air-tissue interface. In the vicinity of air-tissue boundary, e.g. skull and paranasal sinuses, where large susceptibility variations exist, present background field removal methods are usually insufficient and these regions often need to be excluded by brain mask erosion at the expense of losing information of local field and thus susceptibility measures in these regions. In this paper, we propose an extension to the variable-kernel sophisticated harmonic artifact reduction for phase data (V-SHARP) background field removal method using a region adaptive kernel (R-SHARP), in which a scalable spherical Gaussian kernel (SGK) is employed with its kernel radius and weights adjustable according to an energy "functional" reflecting the magnitude of field variation. Such an energy functional is defined in terms of a contour and two fitting functions incorporating regularization terms, from which a curve evolution model in level set formation is derived for energy minimization. We utilize it to detect regions of with a large field gradient caused by strong susceptibility variation. In such regions, the SGK will have a small radius and high weight at the sphere center in a manner adaptive to the voxel energy of the field perturbation. Using the proposed method, the background field generated from external sources can be effectively removed to get a more accurate estimation of the local field and thus of the QSM dipole inversion to map local tissue susceptibility sources. Numerical simulation, phantom and in vivo human brain data demonstrate improved performance of R-SHARP compared to V-SHARP and RESHARP (regularization enabled SHARP) methods, even when the whole paranasal sinus regions are preserved in the brain mask. Shadow artifacts due to strong susceptibility variations in the derived QSM maps could also be largely eliminated using the R-SHARP method, leading to more accurate QSM reconstruction.
Collapse
Affiliation(s)
- Jinsheng Fang
- Department of Electronic Science, Xiamen University, Xiamen 361000, China
| | - Lijun Bao
- Department of Electronic Science, Xiamen University, Xiamen 361000, China
| | - Xu Li
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Peter C.M. van Zijl
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Zhong Chen
- Department of Electronic Science, Xiamen University, Xiamen 361000, China
| |
Collapse
|
100
|
McKiernan EF, O'Brien JT. 7T MRI for neurodegenerative dementias in vivo: a systematic review of the literature. J Neurol Neurosurg Psychiatry 2017; 88:564-574. [PMID: 28259856 DOI: 10.1136/jnnp-2016-315022] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/29/2016] [Accepted: 01/09/2017] [Indexed: 01/12/2023]
Abstract
The spatial resolution of 7T MRI approaches the scale of pathologies of interest in degenerative brain diseases, such as amyloid plaques and changes in cortical layers and subcortical nuclei. It may reveal new information about neurodegenerative dementias, although challenges may include increased artefact production and more adverse effects. We performed a systematic review of papers investigating Alzheimer's disease (AD), Lewy body dementia (LBD), frontotemporal dementia (FTD) and Huntington's disease (HD) in vivo using 7T MRI. Of 19 studies identified, 15 investigated AD (the majority of which examined hippocampal subfield changes), and 4 investigated HD. Ultrahigh resolution revealed changes not visible using lower field strengths, such as hippocampal subfield atrophy in mild cognitive impairment. Increased sensitivity to susceptibility-enhanced iron imaging, facilitating amyloid and microbleed examination; for example, higher microbleed prevalence was found in AD than previously recognised. Theoretical difficulties regarding image acquisition and scan tolerance were not reported as problematic. Study limitations included small subject groups, a lack of studies investigating LBD and FTD and an absence of longitudinal data. In vivo 7T MRI may illuminate disease processes and reveal new biomarkers and therapeutic targets. Evidence from AD and HD studies suggest that other neurodegenerative dementias would also benefit from imaging at ultrahigh resolution.
Collapse
Affiliation(s)
| | - John Tiernan O'Brien
- Department of Psychiatry, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| |
Collapse
|