1
|
Wang AYL, Chen KH, Lin HC, Loh CYY, Chang YC, Aviña AE, Lee CM, Chu IM, Wei FC. Sustained Release of Tacrolimus Embedded in a Mixed Thermosensitive Hydrogel for Improving Functional Recovery of Injured Peripheral Nerves in Extremities. Pharmaceutics 2023; 15:pharmaceutics15020508. [PMID: 36839830 PMCID: PMC9960741 DOI: 10.3390/pharmaceutics15020508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/28/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Vascularized composite allotransplantation is an emerging strategy for the reconstruction of unique defects such as amputated limbs that cannot be repaired with autologous tissues. In order to ensure the function of transplanted limbs, the functional recovery of the anastomosed peripheral nerves must be confirmed. The immunosuppressive drug, tacrolimus, has been reported to promote nerve recovery in animal models. However, its repeated dosing comes with risks of systemic malignancies and opportunistic infections. Therefore, drug delivery approaches for locally sustained release can be designed to overcome this issue and reduce systemic complications. We developed a mixed thermosensitive hydrogel (poloxamer (PLX)-poly(l-alanine-lysine with Pluronic F-127) for the time-dependent sustained release of tacrolimus in our previous study. In this study, we demonstrated that the hydrogel drug degraded in a sustained manner and locally released tacrolimus in mice over one month without affecting the systemic immunity. The hydrogel drug significantly improved the functional recovery of injured sciatic nerves as assessed using five-toe spread and video gait analysis. Neuroregeneration was validated in hydrogel-drug-treated mice using axonal analysis. The hydrogel drug did not cause adverse effects in the mouse model during long-term follow-up. The local injection of encapsulated-tacrolimus mixed thermosensitive hydrogel accelerated peripheral nerve recovery without systemic adverse effects.
Collapse
Affiliation(s)
- Aline Yen Ling Wang
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Correspondence: (A.Y.L.W.); (F.-C.W.)
| | - Kuan-Hung Chen
- Department of Physical Medicine & Rehabilitation, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Hsiu-Chao Lin
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Charles Yuen Yung Loh
- Department of Plastic Surgery, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 0SP, UK
| | - Yun-Ching Chang
- Department of Health Industry Technology Management, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Ana Elena Aviña
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chin-Ming Lee
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - I-Ming Chu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Fu-Chan Wei
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Plastic Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Correspondence: (A.Y.L.W.); (F.-C.W.)
| |
Collapse
|
2
|
Oishi H, Takemura H, Amano K. Macromolecular tissue volume mapping of lateral geniculate nucleus subdivisions in living human brains. Neuroimage 2023; 265:119777. [PMID: 36462730 DOI: 10.1016/j.neuroimage.2022.119777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
The lateral geniculate nucleus (LGN) is a key thalamic nucleus in the visual system, which has an important function in relaying retinal visual input to the visual cortex. The human LGN is composed mainly of magnocellular (M) and parvocellular (P) subdivisions, each of which has different stimulus selectivity in neural response properties. Previous studies have discussed the potential relationship between LGN subdivisions and visual disorders based on psychophysical data on specific types of visual stimuli. However, these relationships remain speculative because non-invasive measurements of these subdivisions are difficult due to the small size of the LGN. Here we propose a method to identify these subdivisions by combining two structural MR measures: high-resolution proton-density weighted images and macromolecular tissue volume (MTV) maps. We defined the M and P subdivisions based on MTV fraction data and tested the validity of the definition by (1) comparing the data with that from human histological studies, (2) comparing the data with functional magnetic resonance imaging measurements on stimulus selectivity, and (3) analyzing the test-retest reliability. The findings demonstrated that the spatial organization of the M and P subdivisions was consistent across subjects and in line with LGN subdivisions observed in human histological data. Moreover, the difference in stimulus selectivity between the subdivisions identified using MTV was consistent with previous physiology literature. The definition of the subdivisions based on MTV was shown to be robust over measurements taken on different days. These results suggest that MTV mapping is a promising approach for evaluating the tissue properties of LGN subdivisions in living humans. This method potentially will enable neuroscientific and clinical hypotheses about the human LGN subdivisions to be tested.
Collapse
Affiliation(s)
- Hiroki Oishi
- Center for Information and Neural Networks (CiNet), Advanced ICT Research Institute, National Institute of Information and Communications Technology, Suita 565-0871, Japan; Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan; Department of Psychology, University of California, Berkeley, Berkeley, CA 94704, United States.
| | - Hiromasa Takemura
- Center for Information and Neural Networks (CiNet), Advanced ICT Research Institute, National Institute of Information and Communications Technology, Suita 565-0871, Japan; Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan; Division of Sensory and Cognitive Brain Mapping, Department of System Neuroscience, National Institute for Physiological Sciences, Okazaki 444-8585, Japan; Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Hayama 240-0193, Japan.
| | - Kaoru Amano
- Center for Information and Neural Networks (CiNet), Advanced ICT Research Institute, National Institute of Information and Communications Technology, Suita 565-0871, Japan; Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan; Graduate School of Information Science and Technology, The University of Tokyo, Tokyo 113-8656, Japan
| |
Collapse
|
3
|
Krijnen EA, Ngamsombat C, George IC, Yu FF, Fan Q, Tian Q, Huang SY, Klawiter EC. Axonal and myelin changes and their inter-relationship in the optic radiations in people with multiple sclerosis. Mult Scler J Exp Transl Clin 2023; 9:20552173221147620. [PMID: 36814811 PMCID: PMC9940187 DOI: 10.1177/20552173221147620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
Abstract
Background The imaging g-ratio, estimated from axonal volume fraction (AVF) and myelin volume fraction (MVF), is a novel biomarker of microstructural tissue integrity in multiple sclerosis (MS). Objective To assess axonal and myelin changes and their inter-relationship as measured by g-ratio in the optic radiations (OR) in people with MS (pwMS) with and without previous optic neuritis (ON) compared to healthy controls (HC). Methods Thirty pwMS and 17 HCs were scanned on a 3Tesla Connectom scanner. AVF and MVF, derived from a multi-shell diffusion protocol and macromolecular tissue volume, respectively, were measured in normal-appearing white matter (NAWM) and lesions within the OR and used to calculate imaging g-ratio. Results OR AVF and MVF were decreased in pwMS compared to HC, and in OR lesions compared to NAWM, whereas the g-ratio was not different. Compared to pwMS with previous ON, AVF and g-ratio tended to be higher in pwMS without prior ON. AVF and MVF, particularly in NAWM, were positively correlated with retinal thickness, which was more pronounced in pwMS with prior ON. Conclusion Axonal measures reflect microstructural tissue damage in the OR, particularly in the setting of remote ON, and correlate with established metrics of visual health in MS.
Collapse
Affiliation(s)
- Eva A Krijnen
- MS Center Amsterdam, Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Chanon Ngamsombat
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Department of Radiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ilena C George
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Fang F Yu
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qiuyun Fan
- Department of Biomedical Engineering, College of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin, China
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Qiyuan Tian
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Susie Y Huang
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Eric C Klawiter
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Berg RC, Menegaux A, Amthor T, Gilbert G, Mora M, Schlaeger S, Pongratz V, Lauerer M, Sorg C, Doneva M, Vavasour I, Mühlau M, Preibisch C. Comparing myelin-sensitive magnetic resonance imaging measures and resulting g-ratios in healthy and multiple sclerosis brains. Neuroimage 2022; 264:119750. [PMID: 36379421 PMCID: PMC9931395 DOI: 10.1016/j.neuroimage.2022.119750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 11/15/2022] Open
Abstract
The myelin concentration and the degree of myelination of nerve fibers can provide valuable information on the integrity of human brain tissue. Magnetic resonance imaging (MRI) of myelin-sensitive parameters can help to non-invasively evaluate demyelinating diseases such as multiple sclerosis (MS). Several different myelin-sensitive MRI methods have been proposed to determine measures of the degree of myelination, in particular the g-ratio. However, variability in underlying physical principles and different biological models influence measured myelin concentrations, and consequently g-ratio values. We therefore investigated similarities and differences between five different myelin-sensitive MRI measures and their effects on g-ratio mapping in the brains of both MS patients and healthy volunteers. We compared two different estimates of the myelin water fraction (MWF) as well as the inhomogeneous magnetization transfer ratio (ihMTR), magnetization transfer saturation (MTsat), and macromolecular tissue volume (MTV) in 13 patients with MS and 14 healthy controls. In combination with diffusion-weighted imaging, we derived g-ratio parameter maps for each of the five different myelin measures. The g-ratio values calculated from different myelin measures varied strongly, especially in MS lesions. While, compared to normal-appearing white matter, MTsat and one estimate of the MWF resulted in higher g-ratio values within lesions, ihMTR, MTV, and the second MWF estimate resulted in lower lesion g-ratio values. As myelin-sensitive measures provide rough estimates of myelin content rather than absolute myelin concentrations, resulting g-ratio values strongly depend on the utilized myelin measure and model used for g-ratio mapping. When comparing g-ratio values, it is, thus, important to utilize the same MRI methods and models or to consider methodological differences. Particular caution is necessary in pathological tissue such as MS lesions.
Collapse
Affiliation(s)
- Ronja C. Berg
- Technical University of Munich, School of Medicine, Department of Diagnostic and Interventional Neuroradiology, Munich, Germany,Technical University of Munich, School of Medicine, Department of Neurology, Munich, Germany,Corresponding author at: Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Diagnostic and Interventional Neuroradiology, Ismaninger Str. 22, 81675, München, Germany. (R.C. Berg)
| | - Aurore Menegaux
- Technical University of Munich, School of Medicine, Department of Diagnostic and Interventional Neuroradiology, Munich, Germany,Technical University of Munich, School of Medicine, TUM Neuroimaging Center, Munich, Germany
| | | | | | - Maria Mora
- Technical University of Munich, School of Medicine, Department of Diagnostic and Interventional Neuroradiology, Munich, Germany
| | - Sarah Schlaeger
- Technical University of Munich, School of Medicine, Department of Diagnostic and Interventional Neuroradiology, Munich, Germany
| | - Viola Pongratz
- Technical University of Munich, School of Medicine, Department of Neurology, Munich, Germany,Technical University of Munich, School of Medicine, TUM Neuroimaging Center, Munich, Germany
| | - Markus Lauerer
- Technical University of Munich, School of Medicine, Department of Neurology, Munich, Germany,Technical University of Munich, School of Medicine, TUM Neuroimaging Center, Munich, Germany
| | - Christian Sorg
- Technical University of Munich, School of Medicine, Department of Diagnostic and Interventional Neuroradiology, Munich, Germany,Technical University of Munich, School of Medicine, TUM Neuroimaging Center, Munich, Germany,Technical University of Munich, School of Medicine, Department of Psychiatry, Munich, Germany
| | | | - Irene Vavasour
- University of British Columbia, Department of Radiology, Vancouver, BC, Canada
| | - Mark Mühlau
- Technical University of Munich, School of Medicine, Department of Neurology, Munich, Germany,Technical University of Munich, School of Medicine, TUM Neuroimaging Center, Munich, Germany
| | - Christine Preibisch
- Technical University of Munich, School of Medicine, Department of Diagnostic and Interventional Neuroradiology, Munich, Germany,Technical University of Munich, School of Medicine, Department of Neurology, Munich, Germany,Technical University of Munich, School of Medicine, TUM Neuroimaging Center, Munich, Germany
| |
Collapse
|
5
|
Combes AJE, Clarke MA, O'Grady KP, Schilling KG, Smith SA. Advanced spinal cord MRI in multiple sclerosis: Current techniques and future directions. Neuroimage Clin 2022; 36:103244. [PMID: 36306717 PMCID: PMC9668663 DOI: 10.1016/j.nicl.2022.103244] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 09/02/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022]
Abstract
Spinal cord magnetic resonance imaging (MRI) has a central role in multiple sclerosis (MS) clinical practice for diagnosis and disease monitoring. Advanced MRI sequences capable of visualizing and quantifying tissue macro- and microstructure and reflecting different pathological disease processes have been used in MS research; however, the spinal cord remains under-explored, partly due to technical obstacles inherent to imaging this structure. We propose that the study of the spinal cord merits equal ambition in overcoming technical challenges, and that there is much information to be exploited to make valuable contributions to our understanding of MS. We present a narrative review on the latest progress in advanced spinal cord MRI in MS, covering in the first part structural, functional, metabolic and vascular imaging methods. We focus on recent studies of MS and those making significant technical steps, noting the challenges that remain to be addressed and what stands to be gained from such advances. Throughout we also refer to other works that presend more in-depth review on specific themes. In the second part, we present several topics that, in our view, hold particular potential. The need for better imaging of gray matter is discussed. We stress the importance of developing imaging beyond the cervical spinal cord, and explore the use of ultra-high field MRI. Finally, some recommendations are given for future research, from study design to newer developments in analysis, and the need for harmonization of sequences and methods within the field. This review is aimed at researchers and clinicians with an interest in gaining an overview of the current state of advanced MRI research in this field and what is primed to be the future of spinal cord imaging in MS research.
Collapse
Affiliation(s)
- Anna J E Combes
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, 1161 21st Avenue South, Medical Center North, AA-1105, Nashville, TN 37232-2310, United States; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Medical Center North, 1161 21st Ave. South, Nashville, TN 37232, United States.
| | - Margareta A Clarke
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, 1161 21st Avenue South, Medical Center North, AA-1105, Nashville, TN 37232-2310, United States
| | - Kristin P O'Grady
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, 1161 21st Avenue South, Medical Center North, AA-1105, Nashville, TN 37232-2310, United States; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Medical Center North, 1161 21st Ave. South, Nashville, TN 37232, United States; Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, PMB 351826, Nashville, TN 37235-1826, United States
| | - Kurt G Schilling
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, 1161 21st Avenue South, Medical Center North, AA-1105, Nashville, TN 37232-2310, United States; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Medical Center North, 1161 21st Ave. South, Nashville, TN 37232, United States
| | - Seth A Smith
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, 1161 21st Avenue South, Medical Center North, AA-1105, Nashville, TN 37232-2310, United States; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Medical Center North, 1161 21st Ave. South, Nashville, TN 37232, United States; Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, PMB 351826, Nashville, TN 37235-1826, United States
| |
Collapse
|
6
|
Trò R, Roascio M, Tortora D, Severino M, Rossi A, Cohen-Adad J, Fato MM, Arnulfo G. Diffusion Kurtosis Imaging of Neonatal Spinal Cord in Clinical Routine. FRONTIERS IN RADIOLOGY 2022; 2:794981. [PMID: 37492682 PMCID: PMC10365122 DOI: 10.3389/fradi.2022.794981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/20/2022] [Indexed: 07/27/2023]
Abstract
Diffusion kurtosis imaging (DKI) has undisputed advantages over the more classical diffusion magnetic resonance imaging (dMRI) as witnessed by the fast-increasing number of clinical applications and software packages widely adopted in brain imaging. However, in the neonatal setting, DKI is still largely underutilized, in particular in spinal cord (SC) imaging, because of its inherently demanding technological requirements. Due to its extreme sensitivity to non-Gaussian diffusion, DKI proves particularly suitable for detecting complex, subtle, fast microstructural changes occurring in this area at this early and critical stage of development, which are not identifiable with only DTI. Given the multiplicity of congenital anomalies of the spinal canal, their crucial effect on later developmental outcome, and the close interconnection between the SC region and the brain above, managing to apply such a method to the neonatal cohort becomes of utmost importance. This study will (i) mention current methodological challenges associated with the application of advanced dMRI methods, like DKI, in early infancy, (ii) illustrate the first semi-automated pipeline built on Spinal Cord Toolbox for handling the DKI data of neonatal SC, from acquisition setting to estimation of diffusion measures, through accurate adjustment of processing algorithms customized for adult SC, and (iii) present results of its application in a pilot clinical case study. With the proposed pipeline, we preliminarily show that DKI is more sensitive than DTI-related measures to alterations caused by brain white matter injuries in the underlying cervical SC.
Collapse
Affiliation(s)
- Rosella Trò
- Departments of Informatics, Bioengineering, Robotics, and System Engineering, University of Genoa, Genoa, Italy
| | - Monica Roascio
- Departments of Informatics, Bioengineering, Robotics, and System Engineering, University of Genoa, Genoa, Italy
| | | | | | - Andrea Rossi
- Neuroradiology Unit, Istituto Giannina Gaslini, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
- Functional Neuroimaging Unit, CRIUGM, Université de Montréal, Montreal, QC, Canada
- Mila—Quebec AI Institute, Montreal, QC, Canada
| | - Marco Massimo Fato
- Departments of Informatics, Bioengineering, Robotics, and System Engineering, University of Genoa, Genoa, Italy
| | - Gabriele Arnulfo
- Departments of Informatics, Bioengineering, Robotics, and System Engineering, University of Genoa, Genoa, Italy
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
7
|
Hori M, Maekawa T, Kamiya K, Hagiwara A, Goto M, Takemura MY, Fujita S, Andica C, Kamagata K, Cohen-Adad J, Aoki S. Advanced Diffusion MR Imaging for Multiple Sclerosis in the Brain and Spinal Cord. Magn Reson Med Sci 2022; 21:58-70. [PMID: 35173096 PMCID: PMC9199983 DOI: 10.2463/mrms.rev.2021-0091] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Diffusion tensor imaging (DTI) has been established its usefulness in evaluating normal-appearing white matter (NAWM) and other lesions that are difficult to evaluate with routine clinical MRI in the evaluation of the brain and spinal cord lesions in multiple sclerosis (MS), a demyelinating disease. With the recent advances in the software and hardware of MRI systems, increasingly complex and sophisticated MRI and analysis methods, such as q-space imaging, diffusional kurtosis imaging, neurite orientation dispersion and density imaging, white matter tract integrity, and multiple diffusion encoding, referred to as advanced diffusion MRI, have been proposed. These are capable of capturing in vivo microstructural changes in the brain and spinal cord in normal and pathological states in greater detail than DTI. This paper reviews the current status of recent advanced diffusion MRI for assessing MS in vivo as part of an issue celebrating two decades of magnetic resonance in medical sciences (MRMS), an official journal of the Japanese Society of Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Masaaki Hori
- Department of Radiology, Toho University Omori Medical Center.,Department of Radiology, Juntendo University School of Medicine
| | - Tomoko Maekawa
- Department of Radiology, Juntendo University School of Medicine
| | - Kouhei Kamiya
- Department of Radiology, Toho University Omori Medical Center.,Department of Radiology, Juntendo University School of Medicine
| | | | - Masami Goto
- Department of Radiological Technology, Faculty of Health Science, Juntendo University
| | | | - Shohei Fujita
- Department of Radiology, Juntendo University School of Medicine
| | | | - Koji Kamagata
- Department of Radiology, Juntendo University School of Medicine
| | | | - Shigeki Aoki
- Department of Radiology, Juntendo University School of Medicine
| |
Collapse
|
8
|
Ogawa S, Takemura H, Horiguchi H, Miyazaki A, Matsumoto K, Masuda Y, Yoshikawa K, Nakano T. Multi-Contrast Magnetic Resonance Imaging of Visual White Matter Pathways in Patients With Glaucoma. Invest Ophthalmol Vis Sci 2022; 63:29. [PMID: 35201263 PMCID: PMC8883150 DOI: 10.1167/iovs.63.2.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 02/03/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose Glaucoma is a disorder that involves visual field loss caused by retinal ganglion cell damage. Previous diffusion magnetic resonance imaging (dMRI) studies have demonstrated that retinal ganglion cell damage affects tissues in the optic tract (OT) and optic radiation (OR). However, because previous studies have used a simple diffusion tensor model to analyze dMRI data, the microstructural interpretation of white matter tissue changes remains uncertain. In this study, we used a multi-contrast MRI approach to further clarify the type of microstructural damage that occurs in patients with glaucoma. Methods We collected dMRI data from 17 patients with glaucoma and 30 controls using 3-tesla (3T) MRI. Using the dMRI data, we estimated three types of tissue property metrics: intracellular volume fraction (ICVF), orientation dispersion index (ODI), and isotropic volume fraction (IsoV). Quantitative T1 (qT1) data, which may be relatively specific to myelin, were collected from all subjects. Results In the OT, all four metrics showed significant differences between the glaucoma and control groups. In the OR, only the ICVF showed significant between-group differences. ICVF was significantly correlated with qT1 in the OR of the glaucoma group, although qT1 did not show any abnormality at the group level. Conclusions Our results suggest that, at the group level, tissue changes in OR caused by glaucoma might be explained by axonal damage, which is reflected in the intracellular diffusion signals, rather than myelin damage. The significant correlation between ICVF and qT1 suggests that myelin damage might also occur in a smaller number of severe cases.
Collapse
Affiliation(s)
- Shumpei Ogawa
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiromasa Takemura
- Center for Information and Neural Networks (CiNet), Advanced ICT Research Institute, National Institute of Information and Communications Technology, Suita, Japan
| | - Hiroshi Horiguchi
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Kenji Matsumoto
- Brain Science Institute, Tamagawa University, Machida, Japan
| | - Yoichiro Masuda
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| | - Keiji Yoshikawa
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
- Yoshikawa Eye Clinic, Machida, Japan
| | - Tadashi Nakano
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
9
|
Yu FF, Yi Huang S, Kumar A, Witzel T, Liao C, Duval T, Cohen-Adad J, Bilgic B. Rapid simultaneous acquisition of macromolecular tissue volume, susceptibility, and relaxometry maps. Magn Reson Med 2022; 87:781-790. [PMID: 34480768 PMCID: PMC8627440 DOI: 10.1002/mrm.28995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 07/13/2021] [Accepted: 08/10/2021] [Indexed: 02/03/2023]
Abstract
PURPOSE A major obstacle to the clinical implementation of quantitative MR is the lengthy acquisition time required to derive multi-contrast parametric maps. We sought to reduce the acquisition time for QSM and macromolecular tissue volume by acquiring both contrasts simultaneously by leveraging their redundancies. The joint virtual coil concept with GRAPPA (JVC-GRAPPA) was applied to reduce acquisition time further. METHODS Three adult volunteers were imaged on a 3 Tesla scanner using a multi-echo 3D GRE sequence acquired at 3 head orientations. Macromolecular tissue volume, QSM, R2∗ , T1 , and proton density maps were reconstructed. The same sequence (GRAPPA R = 4) was performed in subject 1 with a single head orientation for comparison. Fully sampled data was acquired in subject 2, from which retrospective undersampling was performed (R = 6 GRAPPA and R = 9 JVC-GRAPPA). Prospective undersampling was performed in subject 3 (R = 6 GRAPPA and R = 9 JVC-GRAPPA) using gradient blips to shift k-space sampling in later echoes. RESULTS Subject 1's multi-orientation and single-orientation macromolecular tissue volume maps were not significantly different based on RMSE. For subject 2, the retrospectively undersampled JVC-GRAPPA and GRAPPA generated similar results as fully sampled data. This approach was validated with the prospectively undersampled images in subject 3. Using QSM, R2∗ , and macromolecular tissue volume, the contributions of myelin and iron content to susceptibility were estimated. CONCLUSION We have developed a novel strategy to simultaneously acquire data for the reconstruction of 5 intrinsically coregistered 1-mm isotropic resolution multi-parametric maps, with a scan time of 6 min using JVC-GRAPPA.
Collapse
Affiliation(s)
- Fang Frank Yu
- Radiology, University of Texas Southwestern Medical Center, Dallas, TX, United States,,Corresponding author. Fang Frank Yu, MD, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, Ph: 214-648-7813, Fax: 214-648-3904,
| | - Susie Yi Huang
- Department of Radiology, Harvard Medical School, Boston, MA, United States,Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, United States,Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States
| | - Ashwin Kumar
- Vanderbilt University, Nashville, TN, United States
| | | | - Congyu Liao
- Radiological Sciences Laboratory, Stanford Medicine, Stanford, CA, United States
| | - Tanguy Duval
- Institute of Biomedical Engineering, Ecole Polytechnique de Montreal, Montreal, QC, Canada
| | - Julien Cohen-Adad
- Institute of Biomedical Engineering, Ecole Polytechnique de Montreal, Montreal, QC, Canada
| | - Berkin Bilgic
- Department of Radiology, Harvard Medical School, Boston, MA, United States,Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, United States,Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States
| |
Collapse
|
10
|
Tian Q, Fan Q, Witzel T, Polackal MN, Ohringer NA, Ngamsombat C, Russo AW, Machado N, Brewer K, Wang F, Setsompop K, Polimeni JR, Keil B, Wald LL, Rosen BR, Klawiter EC, Nummenmaa A, Huang SY. Comprehensive diffusion MRI dataset for in vivo human brain microstructure mapping using 300 mT/m gradients. Sci Data 2022; 9:7. [PMID: 35042861 PMCID: PMC8766594 DOI: 10.1038/s41597-021-01092-6] [Citation(s) in RCA: 3] [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] [Received: 02/25/2021] [Accepted: 10/25/2021] [Indexed: 12/27/2022] Open
Abstract
Strong gradient systems can improve the signal-to-noise ratio of diffusion MRI measurements and enable a wider range of acquisition parameters that are beneficial for microstructural imaging. We present a comprehensive diffusion MRI dataset of 26 healthy participants acquired on the MGH-USC 3 T Connectome scanner equipped with 300 mT/m maximum gradient strength and a custom-built 64-channel head coil. For each participant, the one-hour long acquisition systematically sampled the accessible diffusion measurement space, including two diffusion times (19 and 49 ms), eight gradient strengths linearly spaced between 30 mT/m and 290 mT/m for each diffusion time, and 32 or 64 uniformly distributed directions. The diffusion MRI data were preprocessed to correct for gradient nonlinearity, eddy currents, and susceptibility induced distortions. In addition, scan/rescan data from a subset of seven individuals were also acquired and provided. The MGH Connectome Diffusion Microstructure Dataset (CDMD) may serve as a test bed for the development of new data analysis methods, such as fiber orientation estimation, tractography and microstructural modelling.
Collapse
Affiliation(s)
- Qiyuan Tian
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, United States
- Harvard Medical School, Boston, Massachusetts, United States
| | - Qiuyun Fan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, United States
- Harvard Medical School, Boston, Massachusetts, United States
| | - Thomas Witzel
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, United States
- Harvard Medical School, Boston, Massachusetts, United States
| | - Maya N Polackal
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, United States
| | - Ned A Ohringer
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, United States
| | - Chanon Ngamsombat
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, United States
| | - Andrew W Russo
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Natalya Machado
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Kristina Brewer
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Fuyixue Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, United States
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States
| | - Kawin Setsompop
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, United States
- Harvard Medical School, Boston, Massachusetts, United States
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States
| | - Jonathan R Polimeni
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, United States
- Harvard Medical School, Boston, Massachusetts, United States
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States
| | - Boris Keil
- Department of Life Science Engineering, Institute of Medical Physics and Radiation Protection, Giessen, Germany
| | - Lawrence L Wald
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, United States
- Harvard Medical School, Boston, Massachusetts, United States
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States
| | - Bruce R Rosen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, United States
- Harvard Medical School, Boston, Massachusetts, United States
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States
| | - Eric C Klawiter
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, United States
- Harvard Medical School, Boston, Massachusetts, United States
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Aapo Nummenmaa
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, United States
- Harvard Medical School, Boston, Massachusetts, United States
| | - Susie Y Huang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, United States.
- Harvard Medical School, Boston, Massachusetts, United States.
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States.
| |
Collapse
|
11
|
Cottaar M, Wu W, Tendler BC, Nagy Z, Miller K, Jbabdi S. Quantifying myelin in crossing fibers using diffusion-prepared phase imaging: Theory and simulations. Magn Reson Med 2021; 86:2618-2634. [PMID: 34254349 PMCID: PMC8581995 DOI: 10.1002/mrm.28907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE Myelin has long been the target of neuroimaging research. However, most available techniques can only provide a voxel-averaged estimate of myelin content. In the human brain, white matter fiber pathways connecting different brain areas and carrying different functions often cross each other in the same voxel. A measure that can differentiate the degree of myelination of crossing fibers would provide a more specific marker of myelination. THEORY AND METHODS One MRI signal property that is sensitive to myelin is the phase accumulation. This sensitivity is used by measuring the phase accumulation of the signal remaining after diffusion-weighting, which is called diffusion-prepared phase imaging (DIPPI). Including diffusion-weighting before estimating the phase accumulation has two distinct advantages for estimating the degree of myelination: (1) It increases the relative contribution of intra-axonal water, whose phase is related linearly to the thickness of the surrounding myelin (in particular the log g-ratio); and (2) it gives directional information, which can be used to distinguish between crossing fibers. Here the DIPPI sequence is described, an approach is proposed to estimate the log g-ratio, and simulations are used and DIPPI data acquired in an isotropic phantom to quantify other sources of phase accumulation. RESULTS The expected bias is estimated in the log g-ratio for reasonable in vivo acquisition parameters caused by eddy currents (~4%-10%), remaining extra-axonal signal (~15%), and gradients in the bulk off-resonance field (<10% for most of the brain). CONCLUSION This new sequence may provide a g-ratio estimate per fiber population crossing within a voxel.
Collapse
Affiliation(s)
- Michiel Cottaar
- Wellcome Centre for Integrative Neuroimaging—Centre for Functional Magnetic Resonance Imaging of the BrainJohn Radcliffe HospitalUniversity of OxfordOxfordUnited Kingdom
| | - Wenchuan Wu
- Wellcome Centre for Integrative Neuroimaging—Centre for Functional Magnetic Resonance Imaging of the BrainJohn Radcliffe HospitalUniversity of OxfordOxfordUnited Kingdom
| | - Benjamin C. Tendler
- Wellcome Centre for Integrative Neuroimaging—Centre for Functional Magnetic Resonance Imaging of the BrainJohn Radcliffe HospitalUniversity of OxfordOxfordUnited Kingdom
| | - Zoltan Nagy
- Laboratory for Social and Neural Systems ResearchUniversity of ZurichZurichSwitzerland
| | - Karla Miller
- Wellcome Centre for Integrative Neuroimaging—Centre for Functional Magnetic Resonance Imaging of the BrainJohn Radcliffe HospitalUniversity of OxfordOxfordUnited Kingdom
| | - Saad Jbabdi
- Wellcome Centre for Integrative Neuroimaging—Centre for Functional Magnetic Resonance Imaging of the BrainJohn Radcliffe HospitalUniversity of OxfordOxfordUnited Kingdom
| |
Collapse
|
12
|
Zheng M, Liu Z, Mana L, Qin G, Huang S, Gong Z, Tian M, He Y, Wang P. Shenzhiling oral liquid protects the myelin sheath against Alzheimer's disease through the PI3K/Akt-mTOR pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 278:114264. [PMID: 34082015 DOI: 10.1016/j.jep.2021.114264] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 05/22/2021] [Accepted: 05/27/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shenzhiling oral liquid (SZL), a traditional Chinese medicine (TCM) compound, is firstly approved by the Chinese Food and Drug Administration (CFDA) for the treatment of mild to moderate Alzheimer's disease (AD). SZL is composed of ten Chinese herbs, and the precise therapy mechanism of its action to AD is far from fully understood. AIM OF THE STUDY The purpose of this study was to observe whether SZL is an effective therapy for amyloid-beta (Aβ)-induced myelin sheath and oligodendrocytes impairments. Notably, the primary aim was to elucidate whether and through what underlying mechanism SZL protects the myelin sheath through the PI3K/Akt-mTOR signaling pathway in Aβ42-induced OLN-93 oligodendrocytes in vitro. MATERIALS AND METHODS APP/PS1 mice were treated with SZL or donepezil continuously for three months, and Aβ42-induced oligodendrocyte OLN-93 cells mimicking AD pathogenesis of myelin sheath impairments were incubated with SZL-containing serum or with donepezil. LC-MS/MS was used to analysis the active components of SZL and SZL-containing serum. The Y maze test was administered after 3 months of treatment, and the hippocampal tissues of the APP/PS1 mice were then harvested for observation of myelin sheath and oligodendrocyte morphology. Cell viability and toxicity were assessed using CCK-8 and lactate dehydrogenase (LDH) release assays, and flow cytometry was used to measure cell apoptosis. The expression of the myelin proteins MBP, PLP, and MAG and that of Aβ42 and Aβ40 in the hippocampi of APP/PS1 mice were examined after SZL treatment. Simultaneously, the expression of p-PI3K, PI3K, p-Akt, Akt, p-mTOR, and mTOR were also examined. The expression of proteins, including CNPase, Olig2, NKX2.2, MBP, PLP, MAG, MOG, p-PI3K, PI3K, p-Akt, Akt, p-mTOR, and mTOR, was determined by immunofluorescence and Western blot, and the corresponding gene expression was evaluated by qPCR in Aβ42-induced OLN-93 oligodendrocytes. RESULTS LC-MS/MS detected a total of 126 active compounds in SZL-containing serum, including terpenoids, flavones, phenols, phenylpropanoids and phenolic acids. SZL treatment significantly improved memory and cognition in APP/PS1 mice and decreased the G-ratio of myelin sheath, alleviated myelin sheath and oligodendrocyte impairments by decreasing Aβ42 and Aβ40 accumulation and increasing the expression of myelin proteins MBP, PLP, MAG, and PI3K/Akt-mTOR signaling pathway associated protein in the hippocampi of APP/PS1 mice. SZL-containing serum also significantly reversed the OLN-93 cell injury induced by Aβ42 by increasing cell viability and enhanced the expression of MBP, PLP, MAG, and MOG. Meanwhile, SZL-containing serum facilitated the maturation and differentiation of oligodendrocytes in Aβ42-induced OLN-93 cells by heightening the expression of CNPase, Olig2 and NKX2.2. SZL-containing serum treatment also fostered the expression of p-PI3K, PI3K, p-Akt, Akt, p-mTOR, and mTOR, indicating an activating PI3K/Akt-mTOR signaling pathway in OLN-93 cells. Furthermore, the effects of SZL on myelin proteins, p-Akt, and p-mTOR were clearly inhibited by LY294002 and/or rapamycin, antagonists of PI3K and m-TOR, respectively. CONCLUSIONS Our findings indicate that SZL exhibits a neuroprotective effect on the myelin sheath by promoting the expression of myelin proteins during AD, and its mechanism of action is closely related to the activation of the PI3K/Akt-mTOR signaling pathway.
Collapse
Affiliation(s)
- Mingcui Zheng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Zhenhong Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China; Institute for Brain Disorders, Beijing University of Chinese Medicine (BUCM), Beijing, 100029, China.
| | - Lulu Mana
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China; Xinjiang Medical University, Urumqi, 830011, China.
| | - Gaofeng Qin
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Shuaiyang Huang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Zhuoyan Gong
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Meijing Tian
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Yannan He
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Pengwen Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, 100700, China; Key Laboratory of Pharmacology Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
13
|
Cohen-Adad J, Alonso-Ortiz E, Abramovic M, Arneitz C, Atcheson N, Barlow L, Barry RL, Barth M, Battiston M, Büchel C, Budde M, Callot V, Combes AJE, De Leener B, Descoteaux M, de Sousa PL, Dostál M, Doyon J, Dvorak A, Eippert F, Epperson KR, Epperson KS, Freund P, Finsterbusch J, Foias A, Fratini M, Fukunaga I, Wheeler-Kingshott CAMG, Germani G, Gilbert G, Giove F, Gros C, Grussu F, Hagiwara A, Henry PG, Horák T, Hori M, Joers J, Kamiya K, Karbasforoushan H, Keřkovský M, Khatibi A, Kim JW, Kinany N, Kitzler H, Kolind S, Kong Y, Kudlička P, Kuntke P, Kurniawan ND, Kusmia S, Labounek R, Laganà MM, Laule C, Law CS, Lenglet C, Leutritz T, Liu Y, Llufriu S, Mackey S, Martinez-Heras E, Mattera L, Nestrasil I, O'Grady KP, Papinutto N, Papp D, Pareto D, Parrish TB, Pichiecchio A, Prados F, Rovira À, Ruitenberg MJ, Samson RS, Savini G, Seif M, Seifert AC, Smith AK, Smith SA, Smith ZA, Solana E, Suzuki Y, Tackley G, Tinnermann A, Valošek J, Van De Ville D, Yiannakas MC, Weber KA, Weiskopf N, Wise RG, Wyss PO, Xu J. Generic acquisition protocol for quantitative MRI of the spinal cord. Nat Protoc 2021; 16:4611-4632. [PMID: 34400839 PMCID: PMC8811488 DOI: 10.1038/s41596-021-00588-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 06/10/2021] [Indexed: 02/08/2023]
Abstract
Quantitative spinal cord (SC) magnetic resonance imaging (MRI) presents many challenges, including a lack of standardized imaging protocols. Here we present a prospectively harmonized quantitative MRI protocol, which we refer to as the spine generic protocol, for users of 3T MRI systems from the three main manufacturers: GE, Philips and Siemens. The protocol provides guidance for assessing SC macrostructural and microstructural integrity: T1-weighted and T2-weighted imaging for SC cross-sectional area computation, multi-echo gradient echo for gray matter cross-sectional area, and magnetization transfer and diffusion weighted imaging for assessing white matter microstructure. In a companion paper from the same authors, the spine generic protocol was used to acquire data across 42 centers in 260 healthy subjects. The key details of the spine generic protocol are also available in an open-access document that can be found at https://github.com/spine-generic/protocols . The protocol will serve as a starting point for researchers and clinicians implementing new SC imaging initiatives so that, in the future, inclusion of the SC in neuroimaging protocols will be more common. The protocol could be implemented by any trained MR technician or by a researcher/clinician familiar with MRI acquisition.
Collapse
Affiliation(s)
- Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, Quebec, Canada.
- Functional Neuroimaging Unit, CRIUGM, University of Montreal, Montreal, Quebec, Canada.
- Mila-Quebec AI Institute, Montreal, Quebec, Canada.
| | - Eva Alonso-Ortiz
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, Quebec, Canada
| | - Mihael Abramovic
- Department of Radiology, Swiss Paraplegic Centre, Nottwil, Switzerland
| | - Carina Arneitz
- Department of Radiology, Swiss Paraplegic Centre, Nottwil, Switzerland
| | - Nicole Atcheson
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland, Australia
| | - Laura Barlow
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert L Barry
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Harvard-Massachusetts Institute of Technology Health Sciences & Technology, Cambridge, MA, USA
| | - Markus Barth
- School of Information Technology and Electrical Engineering, The University of Queensland, Brisbane, Queensland, Australia
| | - Marco Battiston
- NMR Research Unit, Queen Square MS Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Christian Büchel
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthew Budde
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Virginie Callot
- Aix-Marseille Univ, CNRS, CRMBM, Marseille, France
- APHM, Hopital Universitaire Timone, CEMEREM, Marseille, France
| | - Anna J E Combes
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Benjamin De Leener
- Department of Computer and Software Engineering, Polytechnique Montreal, Montreal, Quebec, Canada
- CHU Sainte-Justine Research Centre, Montreal, Quebec, Canada
| | - Maxime Descoteaux
- Centre de Recherche CHUS, CIMS, Sherbrooke, Quebec, Canada
- Sherbrooke Connectivity Imaging Lab (SCIL), Computer Science department, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | - Marek Dostál
- UHB - University Hospital Brno and Masaryk University, Department of Radiology and Nuclear Medicine, Brno, Czech Republic
| | - Julien Doyon
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Adam Dvorak
- Department of Physics and Astronomy, University of British Columbia, Vancouver, British Columbia, Canada
| | - Falk Eippert
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Karla R Epperson
- Richard M. Lucas Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Kevin S Epperson
- Richard M. Lucas Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Patrick Freund
- Spinal Cord Injury Center Balgrist, University of Zurich, Zurich, Switzerland
| | - Jürgen Finsterbusch
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexandru Foias
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, Quebec, Canada
| | - Michela Fratini
- Institute of Nanotechnology, CNR, Rome, Italy
- IRCCS Santa Lucia Foundation, Rome, Italy
| | - Issei Fukunaga
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Claudia A M Gandini Wheeler-Kingshott
- NMR Research Unit, Queen Square MS Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
- Brain MRI 3T Research Centre, IRCCS Mondino Foundation, Pavia, Italy
| | - Giancarlo Germani
- Brain MRI 3T Research Centre, IRCCS Mondino Foundation, Pavia, Italy
| | | | - Federico Giove
- IRCCS Santa Lucia Foundation, Rome, Italy
- CREF - Museo storico della fisica e Centro studi e ricerche Enrico Fermi, Rome, Italy
| | - Charley Gros
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, Quebec, Canada
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland, Australia
| | - Francesco Grussu
- NMR Research Unit, Queen Square MS Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
- Radiomics Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Akifumi Hagiwara
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Pierre-Gilles Henry
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, USA
| | - Tomáš Horák
- Multimodal and functional imaging laboratory, Central European Institute of Technology (CEITEC), Brno, Czech Republic
| | - Masaaki Hori
- Department of Radiology, Toho University Omori Medical Center, Tokyo, Japan
| | - James Joers
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, USA
| | - Kouhei Kamiya
- Department of Radiology, the University of Tokyo, Tokyo, Japan
| | - Haleh Karbasforoushan
- Interdepartmental Neuroscience Program, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, Stanford, CA, USA
| | - Miloš Keřkovský
- UHB - University Hospital Brno and Masaryk University, Department of Radiology and Nuclear Medicine, Brno, Czech Republic
| | - Ali Khatibi
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Centre of Precision Rehabilitation for Spinal Pain (CPR Spine), School of Sport, Exercise and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Joo-Won Kim
- BioMedical Engineering and Imaging Institute (BMEII), Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nawal Kinany
- Institute of Bioengineering/Center for Neuroprosthetics, Ecole Polytechnique Fédérale de Lausanne, Geneva, Switzerland
- Department of Radiology and Medical Informatics, University of Geneva, Geneva, Switzerland
| | - Hagen Kitzler
- Institute of Diagnostic and Interventional Neuroradiology, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Shannon Kolind
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Physics and Astronomy, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medicine (Neurology), University of British Columbia, Vancouver, British Columbia, Canada
| | - Yazhuo Kong
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
- Wellcome Centre For Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Petr Kudlička
- Multimodal and functional imaging laboratory, Central European Institute of Technology (CEITEC), Brno, Czech Republic
| | - Paul Kuntke
- Institute of Diagnostic and Interventional Neuroradiology, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Nyoman D Kurniawan
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland, Australia
| | - Slawomir Kusmia
- CUBRIC, Cardiff University, Wales, UK
- Centre for Medical Image Computing (CMIC), Medical Physics and Biomedical Engineering Department, University College London, London, UK
- Epilepsy Society MRI Unit, Chalfont St Peter, UK
| | - René Labounek
- Division of Clinical Behavioral Neuroscience, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Departments of Neurology and Biomedical Engineering, University Hospital Olomouc, Olomouc, Czech Republic
| | | | - Cornelia Laule
- Departments of Radiology, Pathology & Laboratory Medicine, Physics & Astronomy; International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - Christine S Law
- Division of Pain Medicine, Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Christophe Lenglet
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, USA
| | - Tobias Leutritz
- Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Yaou Liu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Tiantan Image Research Center, China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Sara Llufriu
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain
| | - Sean Mackey
- Division of Pain Medicine, Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Eloy Martinez-Heras
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain
| | - Loan Mattera
- Fondation Campus Biotech Genève, Geneva, Switzerland
| | - Igor Nestrasil
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, USA
- Division of Clinical Behavioral Neuroscience, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Kristin P O'Grady
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nico Papinutto
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Daniel Papp
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, Quebec, Canada
- Wellcome Centre For Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Deborah Pareto
- Neuroradiology Section, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Todd B Parrish
- Interdepartmental Neuroscience Program, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Anna Pichiecchio
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
- Brain MRI 3T Research Centre, IRCCS Mondino Foundation, Pavia, Italy
| | - Ferran Prados
- NMR Research Unit, Queen Square MS Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
- Centre for Medical Image Computing (CMIC), Medical Physics and Biomedical Engineering Department, University College London, London, UK
- E-health Centre, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Àlex Rovira
- Neuroradiology Section, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Rebecca S Samson
- NMR Research Unit, Queen Square MS Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Giovanni Savini
- Brain MRI 3T Research Centre, IRCCS Mondino Foundation, Pavia, Italy
| | - Maryam Seif
- Spinal Cord Injury Center Balgrist, University of Zurich, Zurich, Switzerland
- Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Alan C Seifert
- BioMedical Engineering and Imaging Institute (BMEII), Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alex K Smith
- Wellcome Centre For Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Seth A Smith
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zachary A Smith
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Elisabeth Solana
- Center of Neuroimmunology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clinic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Universitat de Barcelona, Barcelona, Spain
| | - Yuichi Suzuki
- Department of Radiology, the University of Tokyo, Tokyo, Japan
| | | | - Alexandra Tinnermann
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Valošek
- Department of Neurology, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
| | - Dimitri Van De Ville
- Institute of Bioengineering/Center for Neuroprosthetics, Ecole Polytechnique Fédérale de Lausanne, Geneva, Switzerland
- Department of Radiology and Medical Informatics, University of Geneva, Geneva, Switzerland
| | - Marios C Yiannakas
- NMR Research Unit, Queen Square MS Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Kenneth A Weber
- Division of Pain Medicine, Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nikolaus Weiskopf
- Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Felix Bloch Institute for Solid State Physics, Faculty of Physics and Earth Sciences, Leipzig University, Leipzig, Germany
| | - Richard G Wise
- CUBRIC, Cardiff University, Wales, UK
- Institute for Advanced Biomedical Technologies, Department of Neuroscience, Imaging and Clinical Sciences, "G. D'Annunzio University" of Chieti-Pescara, Chieti, Italy
| | - Patrik O Wyss
- Department of Radiology, Swiss Paraplegic Centre, Nottwil, Switzerland
| | - Junqian Xu
- BioMedical Engineering and Imaging Institute (BMEII), Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
14
|
Mancini M, Tian Q, Fan Q, Cercignani M, Huang SY. Dissecting whole-brain conduction delays through MRI microstructural measures. Brain Struct Funct 2021; 226:2651-2663. [PMID: 34390416 PMCID: PMC8448685 DOI: 10.1007/s00429-021-02358-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/28/2021] [Indexed: 01/24/2023]
Abstract
Network models based on structural connectivity have been increasingly used as the blueprint for large-scale simulations of the human brain. As the nodes of this network are distributed through the cortex and interconnected by white matter pathways with different characteristics, modeling the associated conduction delays becomes important. The goal of this study is to estimate and characterize these delays directly from the brain structure. To achieve this, we leveraged microstructural measures from a combination of advanced magnetic resonance imaging acquisitions and computed the main determinants of conduction velocity, namely axonal diameter and myelin content. Using the model proposed by Rushton, we used these measures to calculate the conduction velocity and estimated the associated delays using tractography. We observed that both the axonal diameter and conduction velocity distributions presented a rather constant trend across different connection lengths, with resulting delays that scale linearly with the connection length. Relying on insights from graph theory and Kuramoto simulations, our results support the approximation of constant conduction velocity but also show path- and region-specific differences.
Collapse
Affiliation(s)
- Matteo Mancini
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK. .,Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff University, Cardiff, UK. .,NeuroPoly Lab, Polytechnique Montréal, Montréal, Canada.
| | - Qiyuan Tian
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Qiuyun Fan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Mara Cercignani
- Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff University, Cardiff, UK
| | - Susie Y Huang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Harvard Medical School, Boston, MA, USA.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
15
|
Cohen-Adad J. Quantitative magnetic resonance imaging of spinal cord microstructure in adults with cerebral palsy. Dev Med Child Neurol 2021; 63:896. [PMID: 33990941 DOI: 10.1111/dmcn.14927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Julien Cohen-Adad
- Polytechnique Montreal - NeuroPoly Lab, Institute of Biomedical Engineering, Montreal, Quebec, Canada.,Functional Neuroimaging Unit, CRIUGM, University of Montreal, Montreal, Quebec, Canada.,Mila - Quebec AI Institute, Montreal, Quebec, Canada
| |
Collapse
|
16
|
Buyanova IS, Arsalidou M. Cerebral White Matter Myelination and Relations to Age, Gender, and Cognition: A Selective Review. Front Hum Neurosci 2021; 15:662031. [PMID: 34295229 PMCID: PMC8290169 DOI: 10.3389/fnhum.2021.662031] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 06/02/2021] [Indexed: 12/22/2022] Open
Abstract
White matter makes up about fifty percent of the human brain. Maturation of white matter accompanies biological development and undergoes the most dramatic changes during childhood and adolescence. Despite the advances in neuroimaging techniques, controversy concerning spatial, and temporal patterns of myelination, as well as the degree to which the microstructural characteristics of white matter can vary in a healthy brain as a function of age, gender and cognitive abilities still exists. In a selective review we describe methods of assessing myelination and evaluate effects of age and gender in nine major fiber tracts, highlighting their role in higher-order cognitive functions. Our findings suggests that myelination indices vary by age, fiber tract, and hemisphere. Effects of gender were also identified, although some attribute differences to methodological factors or social and learning opportunities. Findings point to further directions of research that will improve our understanding of the complex myelination-behavior relation across development that may have implications for educational and clinical practice.
Collapse
Affiliation(s)
- Irina S. Buyanova
- Neuropsy Lab, HSE University, Moscow, Russia
- Center for Language and Brain, HSE University, Moscow, Russia
| | - Marie Arsalidou
- Neuropsy Lab, HSE University, Moscow, Russia
- Cognitive Centre, Sirius University of Science and Technology, Sochi, Russia
- Department of Psychology, York University, Toronto, ON, Canada
| |
Collapse
|
17
|
Savini G, Asteggiano C, Paoletti M, Parravicini S, Pezzotti E, Solazzo F, Muzic SI, Santini F, Deligianni X, Gardani A, Germani G, Farina LM, Bergsland N, Gandini Wheeler-Kingshott CAM, Berardinelli A, Bastianello S, Pichiecchio A. Pilot Study on Quantitative Cervical Cord and Muscular MRI in Spinal Muscular Atrophy: Promising Biomarkers of Disease Evolution and Treatment? Front Neurol 2021; 12:613834. [PMID: 33854470 PMCID: PMC8039452 DOI: 10.3389/fneur.2021.613834] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Introduction: Nusinersen is a recent promising therapy approved for the treatment of spinal muscular atrophy (SMA), a rare disease characterized by the degeneration of alpha motor neurons (αMN) in the spinal cord (SC) leading to progressive muscle atrophy and dysfunction. Muscle and cervical SC quantitative magnetic resonance imaging (qMRI) has never been used to monitor drug treatment in SMA. The aim of this pilot study is to investigate whether qMRI can provide useful biomarkers for monitoring treatment efficacy in SMA. Methods: Three adult SMA 3a patients under treatment with nusinersen underwent longitudinal clinical and qMRI examinations every 4 months from baseline to 21-month follow-up. The qMRI protocol aimed to quantify thigh muscle fat fraction (FF) and water-T2 (w-T2) and to characterize SC volumes and microstructure. Eleven healthy controls underwent the same SC protocol (single time point). We evaluated clinical and imaging outcomes of SMA patients longitudinally and compared SC data between groups transversally. Results: Patient motor function was stable, with only Patient 2 showing moderate improvements. Average muscle FF was already high at baseline (50%) and progressed over time (57%). w-T2 was also slightly higher than previously published data at baseline and slightly decreased over time. Cross-sectional area of the whole SC, gray matter (GM), and ventral horns (VHs) of Patients 1 and 3 were reduced compared to controls and remained stable over time, while GM and VHs areas of Patient 2 slightly increased. We found altered diffusion and magnetization transfer parameters in SC structures of SMA patients compared to controls, thus suggesting changes in tissue microstructure and myelin content. Conclusion: In this pilot study, we found a progression of FF in thigh muscles of SMA 3a patients during nusinersen therapy and a concurrent slight reduction of w-T2 over time. The SC qMRI analysis confirmed previous imaging and histopathological studies suggesting degeneration of αMN of the VHs, resulting in GM atrophy and demyelination. Our longitudinal data suggest that qMRI could represent a feasible technique for capturing microstructural changes induced by SMA in vivo and a candidate methodology for monitoring the effects of treatment, once replicated on a larger cohort.
Collapse
Affiliation(s)
- Giovanni Savini
- Advanced Imaging and Radiomics Center, Neuroradiology Department, IRCCS Mondino Foundation, Pavia, Italy
| | - Carlo Asteggiano
- Advanced Imaging and Radiomics Center, Neuroradiology Department, IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Matteo Paoletti
- Advanced Imaging and Radiomics Center, Neuroradiology Department, IRCCS Mondino Foundation, Pavia, Italy
| | - Stefano Parravicini
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Elena Pezzotti
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Francesca Solazzo
- Advanced Imaging and Radiomics Center, Neuroradiology Department, IRCCS Mondino Foundation, Pavia, Italy
| | - Shaun I. Muzic
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Francesco Santini
- Department of Radiology, Division of Radiological Physics, University Hospital Basel, Basel, Switzerland
- Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland
| | - Xeni Deligianni
- Department of Radiology, Division of Radiological Physics, University Hospital Basel, Basel, Switzerland
- Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland
| | - Alice Gardani
- Child Neuropsychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Giancarlo Germani
- Advanced Imaging and Radiomics Center, Neuroradiology Department, IRCCS Mondino Foundation, Pavia, Italy
| | - Lisa M. Farina
- Advanced Imaging and Radiomics Center, Neuroradiology Department, IRCCS Mondino Foundation, Pavia, Italy
| | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
- IRCCS, Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | - Claudia A. M. Gandini Wheeler-Kingshott
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College London, Russell Square, London, United Kingdom
- Brain Connectivity Research Unit, IRCCS Mondino Foundation, Pavia, Italy
| | | | - Stefano Bastianello
- Advanced Imaging and Radiomics Center, Neuroradiology Department, IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Anna Pichiecchio
- Advanced Imaging and Radiomics Center, Neuroradiology Department, IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
18
|
Mohammadi S, Callaghan MF. Towards in vivo g-ratio mapping using MRI: Unifying myelin and diffusion imaging. J Neurosci Methods 2021; 348:108990. [PMID: 33129894 PMCID: PMC7840525 DOI: 10.1016/j.jneumeth.2020.108990] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/21/2020] [Accepted: 10/20/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND The g-ratio, quantifying the comparative thickness of the myelin sheath encasing an axon, is a geometrical invariant that has high functional relevance because of its importance in determining neuronal conduction velocity. Advances in MRI data acquisition and signal modelling have put in vivo mapping of the g-ratio, across the entire white matter, within our reach. This capacity would greatly increase our knowledge of the nervous system: how it functions, and how it is impacted by disease. NEW METHOD This is the second review on the topic of g-ratio mapping using MRI. RESULTS This review summarizes the most recent developments in the field, while also providing methodological background pertinent to aggregate g-ratio weighted mapping, and discussing pitfalls associated with these approaches. COMPARISON WITH EXISTING METHODS Using simulations based on recently published data, this review reveals caveats to the state-of-the-art calibration methods that have been used for in vivo g-ratio mapping. It highlights the need to estimate both the slope and offset of the relationship between these MRI-based markers and the true myelin volume fraction if we are really to achieve the goal of precise, high sensitivity g-ratio mapping in vivo. Other challenges discussed in this review further evidence the need for gold standard measurements of human brain tissue from ex vivo histology. CONCLUSIONS We conclude that the quest to find the most appropriate MRI biomarkers to enable in vivo g-ratio mapping is ongoing, with the full potential of many novel techniques yet to be investigated.
Collapse
Affiliation(s)
- Siawoosh Mohammadi
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| | - Martina F Callaghan
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, University College London, UK
| |
Collapse
|
19
|
Jelescu IO, Palombo M, Bagnato F, Schilling KG. Challenges for biophysical modeling of microstructure. J Neurosci Methods 2020; 344:108861. [PMID: 32692999 PMCID: PMC10163379 DOI: 10.1016/j.jneumeth.2020.108861] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
The biophysical modeling efforts in diffusion MRI have grown considerably over the past 25 years. In this review, we dwell on the various challenges along the journey of bringing a biophysical model from initial design to clinical implementation, identifying both hurdles that have been already overcome and outstanding issues. First, we describe the critical initial task of selecting which features of tissue microstructure can be estimated using a model and which acquisition protocol needs to be implemented to make the estimation possible. The model performance should necessarily be tested in realistic numerical simulations and in experimental data - adapting the fitting strategy accordingly, and parameter estimates should be validated against complementary techniques, when/if available. Secondly, the model performance and validity should be explored in pathological conditions, and, if appropriate, dedicated models for pathology should be developed. We build on examples from tumors, ischemia and demyelinating diseases. We then discuss the challenges associated with clinical translation and added value. Finally, we single out four major unresolved challenges that are related to: the availability of a microstructural ground truth, the validation of model parameters which cannot be accessed with complementary techniques, the development of a generalized standard model for any brain region and pathology, and the seamless communication between different parties involved in the development and application of biophysical models of diffusion.
Collapse
|
20
|
Grussu F, Battiston M, Veraart J, Schneider T, Cohen-Adad J, Shepherd TM, Alexander DC, Fieremans E, Novikov DS, Gandini Wheeler-Kingshott CAM. Multi-parametric quantitative in vivo spinal cord MRI with unified signal readout and image denoising. Neuroimage 2020; 217:116884. [PMID: 32360689 PMCID: PMC7378937 DOI: 10.1016/j.neuroimage.2020.116884] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/18/2020] [Accepted: 04/23/2020] [Indexed: 12/11/2022] Open
Abstract
Multi-parametric quantitative MRI (qMRI) of the spinal cord is a promising non-invasive tool to probe early microstructural damage in neurological disorders. It is usually performed in vivo by combining acquisitions with multiple signal readouts, which exhibit different thermal noise levels, geometrical distortions and susceptibility to physiological noise. This ultimately hinders joint multi-contrast modelling and makes the geometric correspondence of parametric maps challenging. We propose an approach to overcome these limitations, by implementing state-of-the-art microstructural MRI of the spinal cord with a unified signal readout in vivo (i.e. with matched spatial encoding parameters across a range of imaging contrasts). We base our acquisition on single-shot echo planar imaging with reduced field-of-view, and obtain data from two different vendors (vendor 1: Philips Achieva; vendor 2: Siemens Prisma). Importantly, the unified acquisition allows us to compare signal and noise across contrasts, thus enabling overall quality enhancement via multi-contrast image denoising methods. As a proof-of-concept, here we provide a demonstration with one such method, known as Marchenko-Pastur (MP) Principal Component Analysis (PCA) denoising. MP-PCA is a singular value (SV) decomposition truncation approach that relies on redundant acquisitions, i.e. such that the number of measurements is large compared to the number of components that are maintained in the truncated SV decomposition. Here we used in vivo and synthetic data to test whether a unified readout enables more efficient MP-PCA denoising of less redundant acquisitions, since these can be denoised jointly with more redundant ones. We demonstrate that a unified readout provides robust multi-parametric maps, including diffusion and kurtosis tensors from diffusion MRI, myelin metrics from two-pool magnetisation transfer, and T1 and T2 from relaxometry. Moreover, we show that MP-PCA improves the quality of our multi-contrast acquisitions, since it reduces the coefficient of variation (i.e. variability) by up to 17% for mean kurtosis, 8% for bound pool fraction (myelin-sensitive), and 13% for T1, while enabling more efficient denoising of modalities limited in redundancy (e.g. relaxometry). In conclusion, multi-parametric spinal cord qMRI with unified readout is feasible and provides robust microstructural metrics with matched resolution and distortions, whose quality benefits from multi-contrast denoising methods such as MP-PCA.
Collapse
Affiliation(s)
- Francesco Grussu
- Queen Square MS Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK; Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK.
| | - Marco Battiston
- Queen Square MS Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Jelle Veraart
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, USA
| | | | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, Canada; Functional Neuroimaging Unit, CRIUGM, Université de Montréal, Montreal, Canada
| | - Timothy M Shepherd
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, USA
| | - Daniel C Alexander
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - Els Fieremans
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, USA
| | - Dmitry S Novikov
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, USA
| | - Claudia A M Gandini Wheeler-Kingshott
- Queen Square MS Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK; Brain MRI 3T Research Centre, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
21
|
Capllonch-Juan M, Sepulveda F. Modelling the effects of ephaptic coupling on selectivity and response patterns during artificial stimulation of peripheral nerves. PLoS Comput Biol 2020; 16:e1007826. [PMID: 32479499 PMCID: PMC7263584 DOI: 10.1371/journal.pcbi.1007826] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 03/27/2020] [Indexed: 11/18/2022] Open
Abstract
Artificial electrical stimulation of peripheral nerves for sensory feedback restoration can greatly benefit from computational models for simulation-based neural implant design in order to reduce the trial-and-error approach usually taken, thus potentially significantly reducing research and development costs and time. To this end, we built a computational model of a peripheral nerve trunk in which the interstitial space between the fibers and the tissues was modelled using a resistor network, thus enabling distance-dependent ephaptic coupling between myelinated axons and between fascicles as well. We used the model to simulate a) the stimulation of a nerve trunk model with a cuff electrode, and b) the propagation of action potentials along the axons. Results were used to investigate the effect of ephaptic interactions on recruitment and selectivity stemming from artificial (i.e., neural implant) stimulation and on the relative timing between action potentials during propagation. Ephaptic coupling was found to increase the number of fibers that are activated by artificial stimulation, thus reducing the artificial currents required for axonal recruitment, and it was found to reduce and shift the range of optimal stimulation amplitudes for maximum inter-fascicular selectivity. During propagation, while fibers of similar diameters tended to lock their action potentials and reduce their conduction velocities, as expected from previous knowledge on bundles of identical axons, the presence of many other fibers of different diameters was found to make their interactions weaker and unstable.
Collapse
|
22
|
Inter-individual Differences in Occipital Alpha Oscillations Correlate with White Matter Tissue Properties of the Optic Radiation. eNeuro 2020; 7:ENEURO.0224-19.2020. [PMID: 32156741 PMCID: PMC7189484 DOI: 10.1523/eneuro.0224-19.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/31/2022] Open
Abstract
Neural oscillations at ∼10 Hz, called alpha oscillations, are one of the most prominent components of neural oscillations in the human brain. In recent years, characteristics (power/frequency/phase) of occipital alpha oscillations have been correlated with various perceptual phenomena. However, the relationship between inter-individual differences in alpha oscillatory characteristics and the properties of the underlying brain structures, such as white matter pathways, is unclear. A possibility is that intrinsic occipital alpha oscillations are mediated by thalamocortical interaction; we hypothesized that the most promising candidate for characterizing the intrinsic alpha oscillation is optic radiation (OR), which is the geniculo-cortical pathway carrying signals between the lateral geniculate nucleus (LGN) and primary visual cortex (V1). We used resting-state magnetoencephalography (MEG) and diffusion-weighted/quantitative magnetic resonance imaging (MRI) (dMRI/qMRI) to correlate the frequency and power of occipital alpha oscillations with the tissue properties of the OR by focusing on the different characteristics across individuals. We found that the peak alpha frequency (PAF) negatively correlated with intracellular volume fraction (ICVF), reflecting diffusion properties in intracellular (axonal) space, whereas the peak alpha power was not correlated with any tissue properties measurements. No significant correlation was found between OR and beta frequency/amplitude or between other white matter tract connecting parietal and inferotemporal cortex and alpha frequency/amplitude. These results support the hypothesis that an interaction between thalamic nuclei and early visual areas is essential for the occipital alpha oscillatory rhythm.
Collapse
|
23
|
Anterior fissure, central canal, posterior septum and more: New insights into the cervical spinal cord gray and white matter regional organization using T1 mapping at 7T. Neuroimage 2020; 205:116275. [DOI: 10.1016/j.neuroimage.2019.116275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/16/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022] Open
|
24
|
Freund P, Seif M, Weiskopf N, Friston K, Fehlings MG, Thompson AJ, Curt A. MRI in traumatic spinal cord injury: from clinical assessment to neuroimaging biomarkers. Lancet Neurol 2019; 18:1123-1135. [DOI: 10.1016/s1474-4422(19)30138-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 03/22/2019] [Accepted: 03/28/2019] [Indexed: 01/18/2023]
|
25
|
Drakesmith M, Harms R, Rudrapatna SU, Parker GD, Evans CJ, Jones DK. Estimating axon conduction velocity in vivo from microstructural MRI. Neuroimage 2019; 203:116186. [PMID: 31542512 PMCID: PMC6854468 DOI: 10.1016/j.neuroimage.2019.116186] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 11/19/2022] Open
Abstract
The conduction velocity (CV) of action potentials along axons is a key neurophysiological property central to neural communication. The ability to estimate CV in humans in vivo from non-invasive MRI methods would therefore represent a significant advance in neuroscience. However, there are two major challenges that this paper aims to address: (1) Much of the complexity of the neurophysiology of action potentials cannot be captured with currently available MRI techniques. Therefore, we seek to establish the variability in CV that can be captured when predicting CV purely from parameters that have been reported to be estimatable from MRI: inner axon diameter (AD) and g-ratio. (2) errors inherent in existing MRI-based biophysical models of tissue will propagate through to estimates of CV, the extent to which is currently unknown. Issue (1) is investigated by performing a sensitivity analysis on a comprehensive model of axon electrophysiology and determining the relative sensitivity to various morphological and electrical parameters. The investigations suggest that 85% of the variance in CV is accounted for by variation in AD and g-ratio. The observed dependency of CV on AD and g-ratio is well characterised by the previously reported model by Rushton. Issue (2) is investigated through simulation of diffusion and relaxometry MRI data for a range of axon morphologies, applying models of restricted diffusion and relaxation processes to derive estimates of axon volume fraction (AVF), AD and g-ratio and estimating CV from the derived parameters. The results show that errors in the AVF have the biggest detrimental impact on estimates of CV, particularly for sparse fibre populations (AVF<0.3). For our equipment set-up and acquisition protocol, CV estimates are most accurate (below 5% error) where AVF is above 0.3, g-ratio is between 0.6 and 0.85 and AD is high (above 4μm). CV estimates are robust to errors in g-ratio estimation but are highly sensitive to errors in AD estimation, particularly where ADs are small. We additionally show CV estimates in human corpus callosum in a small number of subjects. In conclusion, we demonstrate accurate CV estimates are possible in regions of the brain where AD is sufficiently large. Problems with estimating ADs for smaller axons presents a problem for estimating CV across the whole CNS and should be the focus of further study.
Collapse
Affiliation(s)
- Mark Drakesmith
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, United Kingdom; Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom.
| | - Robbert Harms
- Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, the Netherlands
| | - Suryanarayana Umesh Rudrapatna
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, United Kingdom; Phillips Inovation Campus, Bangalore, India
| | - Greg D Parker
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, United Kingdom; Experimental MRI Centre (EMRIC), School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - C John Evans
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, United Kingdom; Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Derek K Jones
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, United Kingdom; Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom; Mary McKillop Institute for Health Research, Faculty of Health Sciences, Australian Catholic University, Melbourne, Victoria, 3065, Australia
| |
Collapse
|
26
|
Anaby D, Morozov D, Seroussi I, Hametner S, Sochen N, Cohen Y. Single- and double-Diffusion encoding MRI for studying ex vivo apparent axon diameter distribution in spinal cord white matter. NMR IN BIOMEDICINE 2019; 32:e4170. [PMID: 31573745 DOI: 10.1002/nbm.4170] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 07/28/2019] [Accepted: 07/31/2019] [Indexed: 06/10/2023]
Abstract
Mapping average axon diameter (AAD) and axon diameter distribution (ADD) in neuronal tissues non-invasively is a challenging task that may have a tremendous effect on our understanding of the normal and diseased central nervous system (CNS). Water diffusion is used to probe microstructure in neuronal tissues, however, the different water populations and barriers that are present in these tissues turn this into a complex task. Therefore, it is not surprising that recently we have witnessed a burst in the development of new approaches and models that attempt to obtain, non-invasively, detailed microstructural information in the CNS. In this work, we aim at challenging and comparing the microstructural information obtained from single diffusion encoding (SDE) with double diffusion encoding (DDE) MRI. We first applied SDE and DDE MR spectroscopy (MRS) on microcapillary phantoms and then applied SDE and DDE MRI on an ex vivo porcine spinal cord (SC), using similar experimental conditions. The obtained diffusion MRI data were fitted by the same theoretical model, assuming that the signal in every voxel can be approximated as the superposition of a Gaussian-diffusing component and a series of restricted components having infinite cylindrical geometries. The diffusion MRI results were then compared with histological findings. We found a good agreement between the fittings and the experimental data in white matter (WM) voxels of the SC in both diffusion MRI methods. The microstructural information and apparent AADs extracted from SDE MRI were found to be similar or somewhat larger than those extracted from DDE MRI especially when the diffusion time was set to 40 ms. The apparent ADDs extracted from SDE and DDE MRI show reasonable agreement but somewhat weaker correspondence was observed between the diffusion MRI results and histology. The apparent subtle differences between the microstructural information obtained from SDE and DDE MRI are briefly discussed.
Collapse
Affiliation(s)
- Debbie Anaby
- School of Chemistry, The Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Diagnostic Imaging, Sheba Medical Center, Tel HaShomer, Israel
| | - Darya Morozov
- School of Chemistry, The Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Inbar Seroussi
- School of Mathematical Sciences, The Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Simon Hametner
- Neuroimmunology Department, Center of Brain Research, Medical University of Vienna, Vienna, Austria
| | - Nir Sochen
- School of Mathematical Sciences, The Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Yoram Cohen
- School of Chemistry, The Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
27
|
Yu F, Fan Q, Tian Q, Ngamsombat C, Machado N, Bireley JD, Russo AW, Nummenmaa A, Witzel T, Wald LL, Klawiter EC, Huang SY. Imaging G-Ratio in Multiple Sclerosis Using High-Gradient Diffusion MRI and Macromolecular Tissue Volume. AJNR Am J Neuroradiol 2019; 40:1871-1877. [PMID: 31694819 DOI: 10.3174/ajnr.a6283] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Remyelination represents an area of great therapeutic interest in multiple sclerosis but currently lacks a robust imaging marker. The purpose of this study was to use high-gradient diffusion MRI and macromolecular tissue volume imaging to obtain estimates of axonal volume fraction, myelin volume fraction, and the imaging g-ratio in patients with MS and healthy controls and to explore their relationship to neurologic disability in MS. MATERIALS AND METHODS Thirty individuals with MS (23 relapsing-remitting MS, 7 progressive MS) and 19 age-matched healthy controls were scanned on a 3T MRI scanner equipped with 300 mT/m maximum gradient strength using a comprehensive multishell diffusion MRI protocol. Macromolecular tissue volume imaging was performed to quantify the myelin volume fraction. Diffusion data were fitted to a 3-compartment model of white matter using a spheric mean approach to yield estimates of axonal volume fraction. The imaging g-ratio was calculated from the ratio of myelin volume fraction and axonal volume fraction. Imaging metrics were compared between groups using 2-sided t tests with a Bonferroni correction. RESULTS The mean g-ratio was significantly elevated in lesions compared with normal-appearing WM (0.74 vs 0.67, P < .001). Axonal volume fraction (0.17 vs 0.23, P < .001) and myelin volume fraction (0.17 vs 0.25, P < .001) were significantly lower in lesions than normal-appearing WM. Myelin volume fraction was lower in normal-appearing WM compared with that in healthy controls (0.25 vs 0.27, P = .009). Disability, as measured by the Expanded Disability Status Scale, was significantly associated with myelin volume fraction (β = -40.5, P = .001) and axonal volume fraction (β = -41.0, P = .016) in normal-appearing WM. CONCLUSIONS The imaging g-ratio may serve as a biomarker for the relative degree of axonal and myelin loss in MS.
Collapse
Affiliation(s)
- F Yu
- From the Division of Neuroradiology (F.Y.), Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Q Fan
- Athinoula A. Martinos Center for Biomedical Imaging (Q.F., Q.T., C.N., A.N., T.W., L.L.W., S.Y.H.), Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Q Tian
- Athinoula A. Martinos Center for Biomedical Imaging (Q.F., Q.T., C.N., A.N., T.W., L.L.W., S.Y.H.), Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts
| | - C Ngamsombat
- Athinoula A. Martinos Center for Biomedical Imaging (Q.F., Q.T., C.N., A.N., T.W., L.L.W., S.Y.H.), Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts
| | - N Machado
- Department of Neurology (N.M., J.D.B., A.W.R., E.C.K., S.Y.H.)
| | - J D Bireley
- Department of Neurology (N.M., J.D.B., A.W.R., E.C.K., S.Y.H.)
| | - A W Russo
- Department of Neurology (N.M., J.D.B., A.W.R., E.C.K., S.Y.H.)
| | - A Nummenmaa
- Athinoula A. Martinos Center for Biomedical Imaging (Q.F., Q.T., C.N., A.N., T.W., L.L.W., S.Y.H.), Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts
| | - T Witzel
- Athinoula A. Martinos Center for Biomedical Imaging (Q.F., Q.T., C.N., A.N., T.W., L.L.W., S.Y.H.), Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts
| | - L L Wald
- Athinoula A. Martinos Center for Biomedical Imaging (Q.F., Q.T., C.N., A.N., T.W., L.L.W., S.Y.H.), Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts.,Harvard-MIT Division of Health Sciences and Technology (L.L.W., S.Y.H.), Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - E C Klawiter
- Department of Neurology (N.M., J.D.B., A.W.R., E.C.K., S.Y.H.)
| | - S Y Huang
- Athinoula A. Martinos Center for Biomedical Imaging (Q.F., Q.T., C.N., A.N., T.W., L.L.W., S.Y.H.), Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts.,Department of Neurology (N.M., J.D.B., A.W.R., E.C.K., S.Y.H.).,Division of Neuroradiology (S.Y.H.), Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts.,Harvard-MIT Division of Health Sciences and Technology (L.L.W., S.Y.H.), Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
28
|
Rasoanandrianina H, Massire A, Taso M, Guye M, Ranjeva JP, Kober T, Callot V. Regional T 1 mapping of the whole cervical spinal cord using an optimized MP2RAGE sequence. NMR IN BIOMEDICINE 2019; 32:e4142. [PMID: 31393649 DOI: 10.1002/nbm.4142] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 05/20/2019] [Accepted: 06/18/2019] [Indexed: 06/10/2023]
Abstract
The recently-proposed MP2RAGE sequence was purposely optimized for cervical spinal cord imaging at 3T. Sequence parameters were chosen to optimize gray/white matter T1 contrast with sub-millimetric resolution and scan-time < 10 min while preserving reliable T1 determination with minimal B1+ variation effects within a range of values compatible with pathologies and surrounding structures. Results showed good agreements with IR-based measurements, high MP2RAGE-based T1 reproducibility and preliminary evidences of age- and tract-related T1 variations in the healthy spinal cord.
Collapse
Affiliation(s)
- Henitsoa Rasoanandrianina
- Aix-Marseille University, CNRS, CRMBM, Marseille, France
- APHM, Hôpital Universitaire Timone, CEMEREM, Marseille, France
- Aix-Marseille University, IFSTTAR, LBA UMR_T24, Marseille, France
- iLab-Spine International Associated Laboratory, Marseille, France-, Montreal, Canada
| | - Aurélien Massire
- Aix-Marseille University, CNRS, CRMBM, Marseille, France
- APHM, Hôpital Universitaire Timone, CEMEREM, Marseille, France
- iLab-Spine International Associated Laboratory, Marseille, France-, Montreal, Canada
| | - Manuel Taso
- Aix-Marseille University, CNRS, CRMBM, Marseille, France
- APHM, Hôpital Universitaire Timone, CEMEREM, Marseille, France
- iLab-Spine International Associated Laboratory, Marseille, France-, Montreal, Canada
- Division of MRI Research, Department of Radiology, Beth Israel Deaconess Medical Center & Harvard Medical School, Boston, Massachusetts, USA
| | - Maxime Guye
- Aix-Marseille University, CNRS, CRMBM, Marseille, France
- APHM, Hôpital Universitaire Timone, CEMEREM, Marseille, France
| | - Jean-Philippe Ranjeva
- Aix-Marseille University, CNRS, CRMBM, Marseille, France
- APHM, Hôpital Universitaire Timone, CEMEREM, Marseille, France
- iLab-Spine International Associated Laboratory, Marseille, France-, Montreal, Canada
| | - Tobias Kober
- Advanced Clinical Imaging Technology, Siemens Healthcare, Lausanne, Switzerland
- Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Signal Processing Laboratory (LTS 5), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Virginie Callot
- Aix-Marseille University, CNRS, CRMBM, Marseille, France
- APHM, Hôpital Universitaire Timone, CEMEREM, Marseille, France
- iLab-Spine International Associated Laboratory, Marseille, France-, Montreal, Canada
| |
Collapse
|
29
|
Cadotte DW, Akbar MA, Fehlings MG, Stroman PW, Cohen-Adad J. What Has Been Learned from Magnetic Resonance Imaging Examination of the Injured Human Spinal Cord: A Canadian Perspective. J Neurotrauma 2019; 35:1942-1957. [PMID: 30074873 DOI: 10.1089/neu.2018.5903] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Magnetic resonance imaging (MRI) has transformed the way surgeons and researchers study and treat spinal cord injury. In this narrative review, we explore the historical context of imaging the human spinal cord and describe how MRI has evolved from providing the first visualization of the human spinal cord in the 1980s to a remarkable set of imaging tools today. The article focuses in particular on the role of Canadian researchers to this field. We begin by outlining the clinical context of traumatic injury to the human spinal cord and describe why current MRI standards fall short when it comes to treating this disabling condition. Parts 2 and 3 of this work explore an exciting and dramatic shift in the use of MRI technology to aid in our understanding and treatment of traumatic injury to the spinal cord. We explore the use of functional imaging (part 2) and structural imaging (part 3) and explore how these techniques have evolved, how they are used, and the challenges that we face for continued refinement and application to patients who live with the neurological and functional deficits caused by injury to the delicate spinal cord.
Collapse
Affiliation(s)
- David W Cadotte
- 1 University of Calgary Spine Program, Division of Neurosurgery, Department of Clinical Neurosciences, University of Calgary , Foothills Medical Centre, Calgary, Alberta, Canada
| | - M Ali Akbar
- 2 Department of Surgery, Division of Neurosurgery and Spinal Program, Toronto Western Hospital, University of Toronto , Toronto, Ontario, Canada
| | - Michael G Fehlings
- 2 Department of Surgery, Division of Neurosurgery and Spinal Program, Toronto Western Hospital, University of Toronto , Toronto, Ontario, Canada
| | - Patrick W Stroman
- 3 Centre for Neuroscience Studies, Queens University , Kingston, Ontario, Canada
| | - Julien Cohen-Adad
- 4 NeuroPoly Lab, Institute of Biomedical Engineering , Polytechnique Montreal, Montreal, Quebéc, Canada .,5 Functional Neuroimaging Unit, CRIUGM, Université de Montréal , Montreal, Quebéc, Canada
| |
Collapse
|
30
|
Wang AYL, Loh CYY, Shen HH, Hsieh SY, Wang IK, Chuang SH, Wei FC. Topical Application of Human Wharton's Jelly Mesenchymal Stem Cells Accelerates Mouse Sciatic Nerve Recovery and is Associated with Upregulated Neurotrophic Factor Expression. Cell Transplant 2019; 28:1560-1572. [PMID: 31565957 PMCID: PMC6923547 DOI: 10.1177/0963689719880543] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Peripheral nerve regeneration following injury is often slow and impaired, which results in weakened and denervated muscle with subsequent atrophy. Human Wharton's jelly mesenchymal stem cells (hWJ-MSC) have potential regenerative properties which, however, remain unknown in mouse nerve recovery. This study investigated the effect of the topical application of hWJ-MSC onto repairing transected sciatic nerves in a mouse model. Human adipocyte-derived stem cells (hADSC) were used as a positive control. The sciatic nerve of BALB/c mice was transected at a fixed point and repaired under the microscope using 10-0 sutures. hWJ-MSC and hADSC were applied to the site of repair and mice were followed up for 1 year. The hWJ-MSC group had significantly better functional recovery of five-toe spread and gait angles compared with the negative control and hADSC groups. hWJ-MSC improved sciatic nerve regeneration in a dose-dependent fashion. The hWJ-MSC group had a better quality of regenerated nerve with an increased number of myelinated axons throughout. hWJ-MSC appear to be safe in mice after 1 year of follow-up. hWJ-MSC also expressed higher levels of neurotrophic factor-3, brain-derived neurotrophic factor, and glial-derived neurotrophic factor than hADSC. hWJ-MSC may promote better nerve recovery than hADSC because of this upregulation of neurotrophic factors.
Collapse
Affiliation(s)
- Aline Yen Ling Wang
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | | | - Hsin-Hsin Shen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Sing-Ying Hsieh
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Ing-Kae Wang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Sheng-Hao Chuang
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Fu-Chan Wei
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Plastic Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
31
|
Feng Y, Feng F, Zheng C, Zhou Z, Jiang M, Liu Z, Xie F, Sun X, Wu Z. Tanshinone IIA attenuates demyelination and promotes remyelination in A. cantonensis-infected BALB/c mice. Int J Biol Sci 2019; 15:2211-2223. [PMID: 31592236 PMCID: PMC6775289 DOI: 10.7150/ijbs.35266] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/09/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Angiostrongylus cantonensis infection can cause demyelination in the central nervous system, and there is no effective treatment. METHODS We used dexamethasone, Tanshinone IIA (TSIIA) and Cryptotanshinone(Two traditional Chinese medicine monomers) in combination with albendazole (AB, a standard anti-helminthic compound) to observe their therapeutic effect on demyelination in A. cantonensis-infected mice. Luxol fast blue staining and electron microscope of myelin sheath, Oligodendrocyte (OL) number and myelin basic protein (MBP) expression in brain was detected in above groups. RESULTS TSIIA+AB facilitated OL proliferation and significantly increased both myelin sheath thickness and the population of small-diameter axons. In addition, TSIIA treatment inhibited the expression of inflammation-related factors (interleukin [IL]-6, IL-1β, tumor necrosis factor [TNF]-α, inducible nitric oxide synthase [iNOS]) rather than inhibiting eosinophil infiltration in brain. TSIIA also decreased microglial activation and shifted their phenotype from M1 to M2. CONCLUSIONS Taken together, these results provide evidence that TSIIA combined with AB may be an effective treatment for demyelination caused by A. cantonensis infection and other demyelinating diseases.
Collapse
Affiliation(s)
- Ying Feng
- Medical School of South China University of Technology, Guangzhou, China
| | - Feng Feng
- The Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Cunjing Zheng
- Histology and Embryology Department of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zongpu Zhou
- Medical School of South China University of Technology, Guangzhou, China
| | - Meihua Jiang
- Anatomy Department of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhen Liu
- Guangzhou First People's Hospital, Guangzhou, China
| | - Fukang Xie
- Histology and Embryology Department of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xi Sun
- Parasitology Department of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China
| | - Zhongdao Wu
- Parasitology Department of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China
| |
Collapse
|
32
|
Liu H, Ljungberg E, Dvorak AV, Lee LE, Yik JT, MacMillan EL, Barlow L, Li DKB, Traboulsee A, Kolind SH, Kramer JLK, Laule C. Myelin Water Fraction and Intra/Extracellular Water Geometric Mean T 2 Normative Atlases for the Cervical Spinal Cord from 3T MRI. J Neuroimaging 2019; 30:50-57. [PMID: 31407400 DOI: 10.1111/jon.12659] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/19/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Acquiring and interpreting quantitative myelin-specific MRI data at an individual level is challenging because of technical difficulties and natural myelin variation in the population. To overcome these challenges, we used multiecho T2 myelin water imaging (MWI) to create T2 metric healthy population atlases that depict the mean and variation of myelin water fraction (MWF), and intra- and extracellular water mobility as described by geometric mean T2 (IEGMT2 ). METHODS Cervical cord MWI was performed at 3T on 20 healthy individuals (10M/10F, mean age: 36 years) and 3 relapsing remitting multiple sclerosis (RRMS) participants (1M/2F, age: 39/42/37 years). Anatomical data were collected for the purpose of image segmentation and registration. Atlases were created by coregistering and averaging T2 metrics from all controls. Voxel-wise z-score maps from 3 RRMS participants were produced to demonstrate the preliminary utility of the MWF and IEGMT2 atlases. RESULTS The average MWF atlas provides a representation of myelin in the spinal cord consistent with well-known spinal cord anatomical characteristics. The IEGMT2 atlas also depicted structural variations in the spinal cord. Z-score analysis illustrated distinct abnormalities in MWF and IEGMT2 in the 3 RRMS cases. CONCLUSIONS Our findings highlight the potential for using a quantitative T2 relaxation metric atlas to visualize and detect pathology in spinal cord. Our MWF and IEGMT2 atlases (URL: https://sourceforge.net/projects/mwi-spinal-cord-atlases/) can serve as normative references in the cervical spinal cord for other studies.
Collapse
Affiliation(s)
- Hanwen Liu
- Department of Physics & Astronomy, University of British Columbia, Vancouver, Canada.,International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Emil Ljungberg
- Department of Neuroimaging, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK
| | - Adam V Dvorak
- Department of Physics & Astronomy, University of British Columbia, Vancouver, Canada.,International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Lisa Eunyoung Lee
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Jackie T Yik
- Department of Physics & Astronomy, University of British Columbia, Vancouver, Canada.,International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Erin L MacMillan
- Philips, Markham, Canada.,School of Mechatronic Systems Engineering, Simon Fraser University, Canada.,Department of Radiology, University of British Columbia, Vancouver, Canada
| | | | - David K B Li
- Department of Medicine, University of British Columbia, Vancouver, Canada.,Department of Radiology, University of British Columbia, Vancouver, Canada
| | - Anthony Traboulsee
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Shannon H Kolind
- Department of Physics & Astronomy, University of British Columbia, Vancouver, Canada.,International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada.,Department of Medicine, University of British Columbia, Vancouver, Canada.,Department of Radiology, University of British Columbia, Vancouver, Canada
| | - John L K Kramer
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada.,School of Kinesiology, University of British Columbia, Vancouver, Canada
| | - Cornelia Laule
- Department of Physics & Astronomy, University of British Columbia, Vancouver, Canada.,International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada.,Department of Radiology, University of British Columbia, Vancouver, Canada.,Pathology & Laboratory Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
33
|
Schilling KG, By S, Feiler HR, Box BA, O'Grady KP, Witt A, Landman BA, Smith SA. Diffusion MRI microstructural models in the cervical spinal cord - Application, normative values, and correlations with histological analysis. Neuroimage 2019; 201:116026. [PMID: 31326569 DOI: 10.1016/j.neuroimage.2019.116026] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 12/14/2022] Open
Abstract
Multi-compartment tissue modeling using diffusion magnetic resonance imaging has proven valuable in the brain, offering novel indices sensitive to the tissue microstructural environment in vivo on clinical MRI scanners. However, application, characterization, and validation of these models in the spinal cord remain relatively under-studied. In this study, we apply a diffusion "signal" model (diffusion tensor imaging, DTI) and two commonly implemented "microstructural" models (neurite orientation dispersion and density imaging, NODDI; spherical mean technique, SMT) in the human cervical spinal cord of twenty-one healthy controls. We first provide normative values of DTI, SMT, and NODDI indices in a number of white matter ascending and descending pathways, as well as various gray matter regions. We then aim to validate the sensitivity and specificity of these diffusion-derived contrasts by relating these measures to indices of the tissue microenvironment provided by a histological template. We find that DTI indices are sensitive to a number of microstructural features, but lack specificity. The microstructural models also show sensitivity to a number of microstructure features; however, they do not capture the specific microstructural features explicitly modelled. Although often regarded as a simple extension of the brain in the central nervous system, it may be necessary to re-envision, or specifically adapt, diffusion microstructural models for application to the human spinal cord with clinically feasible acquisitions - specifically, adjusting, adapting, and re-validating the modeling as it relates to both theory (i.e. relevant biology, assumptions, and signal regimes) and parameter estimation (for example challenges of acquisition, artifacts, and processing).
Collapse
Affiliation(s)
- Kurt G Schilling
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Samantha By
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Haley R Feiler
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bailey A Box
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kristin P O'Grady
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Atlee Witt
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bennett A Landman
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Department of Electrical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Seth A Smith
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
34
|
Tournier JD. Diffusion MRI in the brain - Theory and concepts. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2019; 112-113:1-16. [PMID: 31481155 DOI: 10.1016/j.pnmrs.2019.03.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/05/2019] [Accepted: 03/07/2019] [Indexed: 06/10/2023]
Abstract
Over the past two decades, diffusion MRI has become an essential tool in neuroimaging investigations. This is due to its sensitivity to the motion of water molecules as they diffuse through the microstructural environment, allowing diffusion MRI to be used as a 'probe' of tissue microstructure. Furthermore, this sensitivity is strongly direction-dependent, notably in brain white matter, due to the alignment of structures that restrict or hinder the motion of water molecules, notably axonal membranes. This provides a means of inferring the orientation of fibres in vivo, and by use of appropriate fibre-tracking algorithms, of delineating the path of white matter tracts in the brain. The ability to perform so-called tractography in humans in vivo non-invasively is unique to diffusion MRI, and is now used in applications such as neurosurgery planning and more broadly within investigations of brain connectomics. This review describes the theory and concepts of diffusion MRI and describes its most important areas of application in the brain, with a strong focus on tractography.
Collapse
Affiliation(s)
- J-Donald Tournier
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, UK; Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, UK.
| |
Collapse
|
35
|
Slater DA, Melie‐Garcia L, Preisig M, Kherif F, Lutti A, Draganski B. Evolution of white matter tract microstructure across the life span. Hum Brain Mapp 2019; 40:2252-2268. [PMID: 30673158 PMCID: PMC6865588 DOI: 10.1002/hbm.24522] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 12/28/2018] [Accepted: 01/02/2019] [Indexed: 01/13/2023] Open
Abstract
The human brain undergoes dramatic structural change over the life span. In a large imaging cohort of 801 individuals aged 7-84 years, we applied quantitative relaxometry and diffusion microstructure imaging in combination with diffusion tractography to investigate tissue property dynamics across the human life span. Significant nonlinear aging effects were consistently observed across tracts and tissue measures. The age at which white matter (WM) fascicles attain peak maturation varies substantially across tissue measurements and tracts. These observations of heterochronicity and spatial heterogeneity of tract maturation highlight the importance of using multiple tissue measurements to investigate each region of the WM. Our data further provide additional quantitative evidence in support of the last-in-first-out retrogenesis hypothesis of aging, demonstrating a strong correlational relationship between peak maturational timing and the extent of quadratic measurement differences across the life span for the most myelin sensitive measures. These findings present an important baseline from which to assess divergence from normative aging trends in developmental and degenerative disorders, and to further investigate the mechanisms connecting WM microstructure to cognition.
Collapse
Affiliation(s)
- David A. Slater
- Laboratory of Research in Neuroimaging (LREN) – Department of Clinical Neurosciences – CHUVUniversity of LausanneLausanneSwitzerland
| | - Lester Melie‐Garcia
- Laboratory of Research in Neuroimaging (LREN) – Department of Clinical Neurosciences – CHUVUniversity of LausanneLausanneSwitzerland
| | - Martin Preisig
- Department of Psychiatry – CHUVUniversity of LausanneLausanneSwitzerland
| | - Ferath Kherif
- Laboratory of Research in Neuroimaging (LREN) – Department of Clinical Neurosciences – CHUVUniversity of LausanneLausanneSwitzerland
| | - Antoine Lutti
- Laboratory of Research in Neuroimaging (LREN) – Department of Clinical Neurosciences – CHUVUniversity of LausanneLausanneSwitzerland
| | - Bogdan Draganski
- Laboratory of Research in Neuroimaging (LREN) – Department of Clinical Neurosciences – CHUVUniversity of LausanneLausanneSwitzerland
- Department of Clinical NeurosciencesMax‐Planck‐Institute for Human Cognitive and Brain SciencesLeipzigGermany
| |
Collapse
|
36
|
Takemura H, Ogawa S, Mezer AA, Horiguchi H, Miyazaki A, Matsumoto K, Shikishima K, Nakano T, Masuda Y. Diffusivity and quantitative T1 profile of human visual white matter tracts after retinal ganglion cell damage. NEUROIMAGE-CLINICAL 2019; 23:101826. [PMID: 31026624 PMCID: PMC6482365 DOI: 10.1016/j.nicl.2019.101826] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/27/2019] [Accepted: 04/13/2019] [Indexed: 02/04/2023]
Abstract
In patients with retinal ganglion cell diseases, recent diffusion tensor imaging (DTI) studies have revealed structural abnormalities in visual white matter tracts such as the optic tract, and optic radiation. However, the microstructural origin of these diffusivity changes is unknown as DTI metrics involve multiple biological factors and do not correlate directly with specific microstructural properties. In contrast, recent quantitative T1 (qT1) mapping methods provide tissue property measurements relatively specific to myelin volume fractions in white matter. This study aims to improve our understanding of microstructural changes in visual white matter tracts following retinal ganglion cell damage in Leber's hereditary optic neuropathy (LHON) patients by combining DTI and qT1 measurements. We collected these measurements from seven LHON patients and twenty age-matched control subjects. For all individuals, we identified the optic tract and the optic radiation using probabilistic tractography, and evaluated diffusivity and qT1 profiles along them. Both diffusivity and qT1 measurements in the optic tract differed significantly between LHON patients and controls. In the optic radiation, these changes were observed in diffusivity but were not evident in qT1 measurements. This suggests that myelin loss may not explain trans-synaptic diffusivity changes in the optic radiation as a consequence of retinal ganglion cell disease. Retinal ganglion cell damage affects diffusivity and T1 along visual pathways. DTI metric identified white matter change in both optic tract and optic radiation. T1 measurement in optic radiation did not exhibit abnormality, unlike DTI metric. Myelin loss may not be a major cause of diffusivity change along optic radiation.
Collapse
Affiliation(s)
- Hiromasa Takemura
- Center for Information and Neural Networks (CiNet), National Institute of Information and Communications Technology, and Osaka University, Suita, Japan; Graduate School of Frontier Biosciences, Osaka University, Suita, Japan.
| | - Shumpei Ogawa
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan; Department of Ophthalmology, Atsugi city hospital, Atsugi, Japan.
| | - Aviv A Mezer
- The Edmond and Lily Safra Center for Brain Science, The Hebrew University of Jerusalem, Israel
| | - Hiroshi Horiguchi
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Kenji Matsumoto
- Brain Science Institute, Tamagawa University, Machida, Japan
| | - Keigo Shikishima
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| | - Tadashi Nakano
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoichiro Masuda
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
37
|
Schmierer K, Miquel ME. Magnetic resonance imaging correlates of neuro-axonal pathology in the MS spinal cord. Brain Pathol 2019; 28:765-772. [PMID: 30375114 DOI: 10.1111/bpa.12648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 07/18/2018] [Indexed: 12/21/2022] Open
Abstract
In people with multiple sclerosis (MS), the spinal cord is the structure most commonly affected by clinically detectable pathology at presentation, and a key part of the central nervous system involved in chronic disease deterioration. Indices, such as the spinal cord cross-sectional area at the level C2 have been developed as tools to predict future disability, and-by inference-axonal loss. However, this and other histo-pathological correlates of spinal cord magnetic resonance imaging (MRI) changes in MS remain incompletely understood. In recent years, there has been a surge of interest in developing quantitative MRI tools to measure specific tissue features, including axonal density, myelin content, neurite density, and orientation, among others, with an emphasis on the spinal cord. Quantitative MRI techniques including T1 and T2 , magnetization transfer and a number of diffusion-derived indices have all been applied to MS spinal cord. Particularly diffusion-based MRI techniques combined with microscopic resolution achievable using high magnetic field scanners enable a new level of anatomical detail and quantification of indices that are clinically meaningful.
Collapse
Affiliation(s)
- Klaus Schmierer
- Queen Mary University of London, Barts and The London School of Medicine & Dentistry, Blizard Institute (Neuroscience), London, UK.,Barts Health NHS Trust, Clinical Board Medicine (Neuroscience), The Royal London Hospital, London, UK
| | - Marc E Miquel
- Barts Health NHS Trust, Clinical Physics, London, UK
| |
Collapse
|
38
|
Magnetic resonance imaging in immune-mediated myelopathies. J Neurol 2019; 267:1233-1244. [PMID: 30694379 DOI: 10.1007/s00415-019-09206-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 01/15/2019] [Accepted: 01/18/2019] [Indexed: 10/27/2022]
Abstract
Immune-mediated myelopathies are a heterogeneous group of inflammatory spinal cord disorders including autoimmune disorders with known antibodies, e.g. aquaporin-4 IgG channelopathy or anti-myelin oligodendrocyte glycoprotein-associated myelitis, myelopathies in the context of multiple sclerosis and systemic autoimmune disorders with myelopathy, as well as post-infectious and paraneoplastic myelopathies. Although magnetic resonance imaging of the spinal cord is still challenging due to the small dimension of the cord cross-section and frequent movement and susceptibility artifacts, recent methodological advances have led to improved diagnostic evaluation and characterization of immune-mediated myelopathies. Topography, length and width of the lesion, gadolinium enhancement pattern, and changes in morphology over time help in narrowing the broad differential diagnosis. In this review, we give an overview of recent advances in magnetic resonance imaging of immune-mediated myelopathies and its role in the differential diagnosis and monitoring of this heterogeneous group of disorders.
Collapse
|
39
|
Jones DK, Alexander DC, Bowtell R, Cercignani M, Dell'Acqua F, McHugh DJ, Miller KL, Palombo M, Parker GJM, Rudrapatna US, Tax CMW. Microstructural imaging of the human brain with a 'super-scanner': 10 key advantages of ultra-strong gradients for diffusion MRI. Neuroimage 2018; 182:8-38. [PMID: 29793061 DOI: 10.1016/j.neuroimage.2018.05.047] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 12/13/2022] Open
Abstract
The key component of a microstructural diffusion MRI 'super-scanner' is a dedicated high-strength gradient system that enables stronger diffusion weightings per unit time compared to conventional gradient designs. This can, in turn, drastically shorten the time needed for diffusion encoding, increase the signal-to-noise ratio, and facilitate measurements at shorter diffusion times. This review, written from the perspective of the UK National Facility for In Vivo MR Imaging of Human Tissue Microstructure, an initiative to establish a shared 300 mT/m-gradient facility amongst the microstructural imaging community, describes ten advantages of ultra-strong gradients for microstructural imaging. Specifically, we will discuss how the increase of the accessible measurement space compared to a lower-gradient systems (in terms of Δ, b-value, and TE) can accelerate developments in the areas of 1) axon diameter distribution mapping; 2) microstructural parameter estimation; 3) mapping micro-vs macroscopic anisotropy features with gradient waveforms beyond a single pair of pulsed-gradients; 4) multi-contrast experiments, e.g. diffusion-relaxometry; 5) tractography and high-resolution imaging in vivo and 6) post mortem; 7) diffusion-weighted spectroscopy of metabolites other than water; 8) tumour characterisation; 9) functional diffusion MRI; and 10) quality enhancement of images acquired on lower-gradient systems. We finally discuss practical barriers in the use of ultra-strong gradients, and provide an outlook on the next generation of 'super-scanners'.
Collapse
Affiliation(s)
- D K Jones
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK; School of Psychology, Faculty of Health Sciences, Australian Catholic University, Melbourne, Victoria, 3065, Australia.
| | - D C Alexander
- Centre for Medical Image Computing (CMIC), Department of Computer Science, UCL (University College London), Gower Street, London, UK; Clinical Imaging Research Centre, National University of Singapore, Singapore
| | - R Bowtell
- Sir Peter Mansfield Magnetic Resonance Centre, School of Physics and Astronomy, University of Nottingham, University Park, Nottingham, UK
| | - M Cercignani
- Department of Psychiatry, Brighton and Sussex Medical School, Brighton, UK
| | - F Dell'Acqua
- Natbrainlab, Department of Neuroimaging, King's College London, London, UK
| | - D J McHugh
- Division of Informatics, Imaging and Data Sciences, The University of Manchester, Manchester, UK; CRUK and EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge and Manchester, UK
| | - K L Miller
- Oxford Centre for Functional MRI of the Brain, University of Oxford, Oxford, UK
| | - M Palombo
- Centre for Medical Image Computing (CMIC), Department of Computer Science, UCL (University College London), Gower Street, London, UK
| | - G J M Parker
- Division of Informatics, Imaging and Data Sciences, The University of Manchester, Manchester, UK; CRUK and EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge and Manchester, UK; Bioxydyn Ltd., Manchester, UK
| | - U S Rudrapatna
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK
| | - C M W Tax
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK
| |
Collapse
|
40
|
Microstructural properties of the vertical occipital fasciculus explain the variability in human stereoacuity. Proc Natl Acad Sci U S A 2018; 115:12289-12294. [PMID: 30429321 PMCID: PMC6275509 DOI: 10.1073/pnas.1804741115] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Seeing in the three-dimensional world—stereopsis—is an innate human ability, but it varies substantially among individuals. The neurobiological basis of this variability is not understood. We combined diffusion and quantitative MRI imaging with a psychophysical measurements, and found that variability in stereoacuity is associated with microstructural differences in the right vertical occipital fasciculus, a white matter tract connecting dorsal and ventral visual cortex. This result suggests that the microstructure of the pathways that support information transmission across dorsal and ventral visual areas plays an important role human stereopsis. Stereopsis is a fundamental visual function that has been studied extensively. However, it is not clear why depth discrimination (stereoacuity) varies more significantly among people than other modalities. Previous studies have reported the involvement of both dorsal and ventral visual areas in stereopsis, implying that not only neural computations in cortical areas but also the anatomical properties of white matter tracts connecting those areas can impact stereopsis. Here, we studied how human stereoacuity relates to white matter properties by combining psychophysics, diffusion MRI (dMRI), and quantitative MRI (qMRI). We performed a psychophysical experiment to measure stereoacuity and, in the same participants, we analyzed the microstructural properties of visual white matter tracts on the basis of two independent measurements, dMRI (fractional anisotropy, FA) and qMRI (macromolecular tissue volume; MTV). Microstructural properties along the right vertical occipital fasciculus (VOF), a major tract connecting dorsal and ventral visual areas, were highly correlated with measures of stereoacuity. This result was consistent for both FA and MTV, suggesting that the behavioral–structural relationship reflects differences in neural tissue density, rather than differences in the morphological configuration of fibers. fMRI confirmed that binocular disparity stimuli activated the dorsal and ventral visual regions near VOF endpoints. No other occipital tracts explained the variance in stereoacuity. In addition, the VOF properties were not associated with differences in performance on a different psychophysical task (contrast detection). These series of experiments suggest that stereoscopic depth discrimination performance is, at least in part, constrained by dorso-ventral communication through the VOF.
Collapse
|
41
|
Duval T, Saliani A, Nami H, Nanci A, Stikov N, Leblond H, Cohen-Adad J. Axons morphometry in the human spinal cord. Neuroimage 2018; 185:119-128. [PMID: 30326296 DOI: 10.1016/j.neuroimage.2018.10.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 10/05/2018] [Accepted: 10/10/2018] [Indexed: 12/15/2022] Open
Abstract
Due to the technical challenges of large-scale microscopy and analysis, to date only limited knowledge has been made available about axon morphometry (diameter, shape, myelin thickness, volume fraction), thereby limiting our understanding of neuronal microstructure and slowing down research on neurodegenerative pathologies. This study addresses this knowledge gap by establishing a state-of-the-art acquisition and analysis framework for mapping axon morphometry, and providing the first comprehensive mapping of axon morphometry in the human spinal cord. We dissected, fixed and stained a human spinal cord with osmium tetroxide, and used a scanning electron microscope to image the entirety of 23 axial slices, covering C1 to L5 spinal levels. An automatic method based on deep learning was then used to segment each axon and myelin sheath to produce maps of axon morphometry. These maps were then registered to a standard spinal cord magnetic resonance imaging (MRI) template. Between 500,000 (lumbar) and 1 million (cervical) myelinated axons were segmented at each level of this human spinal cord. Morphometric features show a large disparity between tracts, but high right-left symmetry. Our results suggest a modality-based organization of the dorsal column in the human, as it has been observed in the rat. The generated axon morphometry template is publicly available at https://osf.io/8k7jr/ and could be used as a reference for quantitative MRI studies. The proposed framework for axon morphometry mapping could be extended to other parts of the central or peripheral nervous system that exhibit coherently-oriented axons.
Collapse
Affiliation(s)
- Tanguy Duval
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
| | - Ariane Saliani
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
| | - Harris Nami
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
| | - Antonio Nanci
- Laboratory for the Study of Calcified Tissues and Biomaterials, Faculty of Dental Medicine, University of Montreal, Montreal, QC, Canada
| | - Nikola Stikov
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada; Montreal Heart Institute, Montreal, QC, Canada
| | - Hugues Leblond
- Anatomy department, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada; Functional Neuroimaging Unit, CRIUGM, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
42
|
Grussu F, Ianuş A, Tur C, Prados F, Schneider T, Kaden E, Ourselin S, Drobnjak I, Zhang H, Alexander DC, Gandini Wheeler-Kingshott CAM. Relevance of time-dependence for clinically viable diffusion imaging of the spinal cord. Magn Reson Med 2018; 81:1247-1264. [PMID: 30229564 PMCID: PMC6586052 DOI: 10.1002/mrm.27463] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 12/17/2022]
Abstract
Purpose Time‐dependence is a key feature of the diffusion‐weighted (DW) signal, knowledge of which informs biophysical modelling. Here, we study time‐dependence in the human spinal cord, as its axonal structure is specific and different from the brain. Methods We run Monte Carlo simulations using a synthetic model of spinal cord white matter (WM) (large axons), and of brain WM (smaller axons). Furthermore, we study clinically feasible multi‐shell DW scans of the cervical spinal cord (b = 0; b = 711 s mm−2; b = 2855 s mm−2), obtained using three diffusion times (Δ of 29, 52 and 76 ms) from three volunteers. Results Both intra‐/extra‐axonal perpendicular diffusivities and kurtosis excess show time‐dependence in our synthetic spinal cord model. This time‐dependence is reflected mostly in the intra‐axonal perpendicular DW signal, which also exhibits strong decay, unlike our brain model. Time‐dependence of the total DW signal appears detectable in the presence of noise in our synthetic spinal cord model, but not in the brain. In WM in vivo, we observe time‐dependent macroscopic and microscopic diffusivities and diffusion kurtosis, NODDI and two‐compartment SMT metrics. Accounting for large axon calibers improves fitting of multi‐compartment models to a minor extent. Conclusions Time‐dependence of clinically viable DW MRI metrics can be detected in vivo in spinal cord WM, thus providing new opportunities for the non‐invasive estimation of microstructural properties. The time‐dependence of the perpendicular DW signal may feature strong intra‐axonal contributions due to large spinal axon caliber. Hence, a popular model known as “stick” (zero‐radius cylinder) may be sub‐optimal to describe signals from the largest spinal axons.
Collapse
Affiliation(s)
- Francesco Grussu
- Queen Square MS Centre, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom.,Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Andrada Ianuş
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom.,Champalimaud Centre for the Unknown, Champalimaud Foundation, Lisbon, Portugal
| | - Carmen Tur
- Queen Square MS Centre, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Ferran Prados
- Queen Square MS Centre, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom.,Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | | | - Enrico Kaden
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Sébastien Ourselin
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Ivana Drobnjak
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Hui Zhang
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Daniel C Alexander
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom.,Clinical Imaging Research Centre, National University of Singapore, Singapore, Singapore
| | - Claudia A M Gandini Wheeler-Kingshott
- Queen Square MS Centre, UCL Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom.,Brain MRI 3T Research Centre, C. Mondino National Neurological Institute, Pavia, Italy.,Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
43
|
Duval T, Smith V, Stikov N, Klawiter EC, Cohen-Adad J. Scan-rescan of axcaliber, macromolecular tissue volume, and g-ratio in the spinal cord. Magn Reson Med 2018; 79:2759-2765. [PMID: 28994487 PMCID: PMC5821542 DOI: 10.1002/mrm.26945] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 09/01/2017] [Accepted: 09/03/2017] [Indexed: 01/25/2023]
Abstract
PURPOSE Recent MRI techniques have been introduced that can extract microstructural information in the white matter, such as the density or macromolecular content. Translating quantitative MRI to the clinic raises many challenges in terms of acquisition strategy, modeling of the MRI signal, artifact corrections, and metric extraction (template registration and partial volume effects). In this work, we investigated the scan-rescan repeatability of several quantitative MRI techniques in the human spinal cord. METHODS AxCaliber metrics, macromolecular tissue volume, and the fiber g-ratio were estimated in the spinal cord of eight healthy subjects, scanned and rescanned the same day in two different sessions. RESULTS Scan-rescan repeatability deviation was 3% for all metrics, in average in the white matter of all subjects. Intraclass correlation coefficient was up to 0.9. A three-way analysis of variance showed significant effects of white matter pathway, laterality, and subject. CONCLUSION The present study suggests that quantitative MRI gives stable measurements of white matter microstructure in the spinal cord of healthy subjects. Our findings remain to be evaluated in diseased populations. Magn Reson Med 79:2759-2765, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Tanguy Duval
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
| | - Victoria Smith
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Nikola Stikov
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
- Montreal Heart Institute, Montreal, QC, Canada
| | - Eric C. Klawiter
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
- Functional Neuroimaging Unit, CRIUGM, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
44
|
Cohen-Adad J. Microstructural imaging in the spinal cord and validation strategies. Neuroimage 2018; 182:169-183. [PMID: 29635029 DOI: 10.1016/j.neuroimage.2018.04.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 03/02/2018] [Accepted: 04/06/2018] [Indexed: 12/13/2022] Open
Abstract
In vivo histology using magnetic resonance imaging (MRI) is a newly emerging research field that aims to non-invasively characterize tissue microstructure. The implications of in vivo histology are many, from discovering novel biomarkers to studying human development, to providing tools for disease diagnosis and monitoring the effects of novel treatments on tissue. This review focuses on quantitative MRI (qMRI) techniques that are used to map spinal cord microstructure. Opening with a rationale for non-invasive imaging of the spinal cord, this article continues with a brief overview of the existing MRI techniques for axon and myelin imaging, followed by the specific challenges and potential solutions for acquiring and processing such data. The final part of this review focuses on histological validation, with suggested tissue preparation, acquisition and processing protocols for large-scale microscopy.
Collapse
Affiliation(s)
- J Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada; Functional Neuroimaging Unit, CRIUGM, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
45
|
Hori M, Hagiwara A, Fukunaga I, Ueda R, Kamiya K, Suzuki Y, Liu W, Murata K, Takamura T, Hamasaki N, Irie R, Kamagata K, Kumamaru KK, Suzuki M, Aoki S. Application of Quantitative Microstructural MR Imaging with Atlas-based Analysis for the Spinal Cord in Cervical Spondylotic Myelopathy. Sci Rep 2018; 8:5213. [PMID: 29581458 PMCID: PMC5979956 DOI: 10.1038/s41598-018-23527-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 03/15/2018] [Indexed: 12/14/2022] Open
Abstract
Mapping of MR fiber g-ratio, which is the ratio of the diameter of the axon to the diameter of the neuronal fiber, is introduced in this article. We investigated the MR fiber g-ratio, the axon volume fraction (AVF) and the myelin volume fraction (MVF) to evaluate microstructural changes in the spinal cord in patients with cervical spondylotic myelopathy (CSM) in vivo, using atlas-based analysis. We used diffusion MRI data acquired with a new simultaneous multi-slice accelerated readout-segmented echo planar imaging sequence for diffusion analysis for AVF calculation and magnetization transfer saturation imaging for MVF calculation. The AVFs of fasciculus gracilis in the affected side spinal cord, fasciculus cuneatus and lateral corticospinal tracts (LSCT) in the affected and unaffected side spinal cord were significantly lower (P = 0.019, 0.001, 0019, 0.000, and 0.002, respectively) than those of normal controls. No difference was found in the MVFs. The fiber g-ratio of LSCT was significantly lower (P = 0.040) in the affected side spinal cords than in the normal controls. The pathological microstructural changes in the spinal cord in patients with CSM, presumably partial axonal degenerations with preserved myelin. This technique has the potential to be a clinical biomarker in patients with CSM in vivo.
Collapse
Affiliation(s)
- Masaaki Hori
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan.
| | - Akifumi Hagiwara
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan.,Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Issei Fukunaga
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ryo Ueda
- Health Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Kouhei Kamiya
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuichi Suzuki
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Wei Liu
- Siemens Shenzhen Magnetic Resonance Ltd, Shenzhen, China
| | | | - Tomohiro Takamura
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Nozomi Hamasaki
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ryusuke Irie
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan.,Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Koji Kamagata
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | | | - Michimasa Suzuki
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Shigeki Aoki
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
46
|
Duyn JH. Studying brain microstructure with magnetic susceptibility contrast at high-field. Neuroimage 2018; 168:152-161. [PMID: 28242317 PMCID: PMC5569005 DOI: 10.1016/j.neuroimage.2017.02.046] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 02/03/2017] [Accepted: 02/16/2017] [Indexed: 12/14/2022] Open
Abstract
A rapidly developing application of high field MRI is the study of brain anatomy and function with contrast based on the magnetic susceptibility of tissues. To study the subtle variations in susceptibility contrast between tissues and with changes in brain activity, dedicated scan techniques such as susceptibility-weighted MRI and blood-oxygen level dependent functional MRI have been developed. Particularly strong susceptibility contrast has been observed with systems that operate at 7T and above, and their recent widespread use has led to an improved understanding of contributing sources and mechanisms. To interpret magnetic susceptibility contrast, analysis approaches have been developed with the goal of extracting measures that report on local tissue magnetic susceptibility, a physical quantity that, under certain conditions, allows estimation of blood oxygenation, local tissue iron content, and quantification of its changes with disease. Interestingly, high field studies have also brought to light that not only the makeup of tissues affects MRI susceptibility contrast, but that also a tissue's sub-voxel structure at scales all the way down to the molecular level plays an important role as well. In this review, various ways will be discussed by which sub-voxel structure can affect the MRI signal in general, and magnetic susceptibility in particular, sometimes in a complex fashion. In the light of this complexity, it appears likely that accurate, brain-wide quantification of iron will require the combination of multiple contrasts that may include diffusion and magnetization transfer information with susceptibility-weighted contrast. On the other hand, this complexity also offers opportunities to use magnetic susceptibility contrast to inform about specific microstructural aspects of brain tissue. Details and several examples will be presented in this review.
Collapse
Affiliation(s)
- Jeff H Duyn
- Advanced MRI Section, Laboratory of Functional and Molecular Imaging, National Institutes of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
47
|
Kafali SG, Çukur T, Saritas EU. Phase-correcting non-local means filtering for diffusion-weighted imaging of the spinal cord. Magn Reson Med 2018; 80:1020-1035. [PMID: 29427379 DOI: 10.1002/mrm.27105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/26/2017] [Accepted: 01/02/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Sevgi Gokce Kafali
- Department of Electrical and Electronics Engineering, Bilkent University, Ankara, Turkey.,National Magnetic Resonance Research Center (UMRAM), Bilkent University, Ankara, Turkey
| | - Tolga Çukur
- Department of Electrical and Electronics Engineering, Bilkent University, Ankara, Turkey.,National Magnetic Resonance Research Center (UMRAM), Bilkent University, Ankara, Turkey.,Neuroscience Program, Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey
| | - Emine Ulku Saritas
- Department of Electrical and Electronics Engineering, Bilkent University, Ankara, Turkey.,National Magnetic Resonance Research Center (UMRAM), Bilkent University, Ankara, Turkey.,Neuroscience Program, Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey
| |
Collapse
|
48
|
Lévy S, Guertin MC, Khatibi A, Mezer A, Martinu K, Chen JI, Stikov N, Rainville P, Cohen-Adad J. Test-retest reliability of myelin imaging in the human spinal cord: Measurement errors versus region- and aging-induced variations. PLoS One 2018; 13:e0189944. [PMID: 29293550 PMCID: PMC5749716 DOI: 10.1371/journal.pone.0189944] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 12/05/2017] [Indexed: 01/06/2023] Open
Abstract
PURPOSE To implement a statistical framework for assessing the precision of several quantitative MRI metrics sensitive to myelin in the human spinal cord: T1, Magnetization Transfer Ratio (MTR), saturation imposed by an off-resonance pulse (MTsat) and Macromolecular Tissue Volume (MTV). METHODS Thirty-three healthy subjects within two age groups (young, elderly) were scanned at 3T. Among them, 16 underwent the protocol twice to assess repeatability. Statistical reliability indexes such as the Minimal Detectable Change (MDC) were compared across metrics quantified within different cervical levels and white matter (WM) sub-regions. The differences between pathways and age groups were quantified and interpreted in context of the test-retest repeatability of the measurements. RESULTS The MDC was respectively 105.7ms, 2.77%, 0.37% and 4.08% for T1, MTR, MTsat and MTV when quantified over all WM, while the standard-deviation across subjects was 70.5ms, 1.34%, 0.20% and 2.44%. Even though particular WM regions did exhibit significant differences, these differences were on the same order as test-retest errors. No significant difference was found between age groups for all metrics. CONCLUSION While T1-based metrics (T1 and MTV) exhibited better reliability than MT-based measurements (MTR and MTsat), the observed differences between subjects or WM regions were comparable to (and often smaller than) the MDC. This makes it difficult to determine if observed changes are due to variations in myelin content, or simply due to measurement error. Measurement error remains a challenge in spinal cord myelin imaging, but this study provides statistical guidelines to standardize the field and make it possible to conduct large-scale multi-center studies.
Collapse
Affiliation(s)
- Simon Lévy
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal (CRIUGM), Montréal, QC, Canada
| | - Marie-Claude Guertin
- Montreal Health Innovations Coordinating Center (MHICC), Montreal Heart Institute, Montreal, QC, Canada
| | - Ali Khatibi
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal (CRIUGM), Montréal, QC, Canada
- Psychology Department, Bilkent University, Ankara, Turkey
- Interdisciplinary program in Neuroscience, Bilkent University, Ankara, Turkey
- National Magnetic Resonance Research Center (UMRAM), Bilkent University, Ankara, Turkey
| | - Aviv Mezer
- The Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Kristina Martinu
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal (CRIUGM), Montréal, QC, Canada
| | - Jen-I Chen
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal (CRIUGM), Montréal, QC, Canada
- Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montreal, QC, Canada
| | - Nikola Stikov
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
- Montreal Heart Institute, Montreal, QC, Canada
| | - Pierre Rainville
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal (CRIUGM), Montréal, QC, Canada
- Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montreal, QC, Canada
| | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
- Functional Neuroimaging Unit, CRIUGM, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
49
|
Ellerbrock I, Mohammadi S. Four in vivo g-ratio-weighted imaging methods: Comparability and repeatability at the group level. Hum Brain Mapp 2018; 39:24-41. [PMID: 29091341 PMCID: PMC6866374 DOI: 10.1002/hbm.23858] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 10/11/2017] [Accepted: 10/16/2017] [Indexed: 12/18/2022] Open
Abstract
A recent method, denoted in vivo g-ratio-weighted imaging, has related the microscopic g-ratio, only accessible by ex vivo histology, to noninvasive MRI markers for the fiber volume fraction (FVF) and myelin volume fraction (MVF). Different MRI markers have been proposed for g-ratio weighted imaging, leaving open the question which combination of imaging markers is optimal. To address this question, the repeatability and comparability of four g-ratio methods based on different combinations of, respectively, two imaging markers for FVF (tract-fiber density, TFD, and neurite orientation dispersion and density imaging, NODDI) and two imaging markers for MVF (magnetization transfer saturation rate, MT, and, from proton density maps, macromolecular tissue volume, MTV) were tested in a scan-rescan experiment in two groups. Moreover, it was tested how the repeatability and comparability were affected by two key processing steps, namely the masking of unreliable voxels (e.g., due to partial volume effects) at the group level and the calibration value used to link MRI markers to MVF (and FVF). Our data showed that repeatability and comparability depend largely on the marker for the FVF (NODDI outperformed TFD), and that they were improved by masking. Overall, the g-ratio method based on NODDI and MT showed the highest repeatability (90%) and lowest variability between groups (3.5%). Finally, our results indicate that the calibration procedure is crucial, for example, calibration to a lower g-ratio value (g = 0.6) than the commonly used one (g = 0.7) can change not only repeatability and comparability but also the reported dependency on the FVF imaging marker. Hum Brain Mapp 39:24-41, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Isabel Ellerbrock
- Department of Systems NeuroscienceUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Siawoosh Mohammadi
- Department of Systems NeuroscienceUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
50
|
PAM50: Unbiased multimodal template of the brainstem and spinal cord aligned with the ICBM152 space. Neuroimage 2018; 165:170-179. [DOI: 10.1016/j.neuroimage.2017.10.041] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/16/2017] [Accepted: 10/20/2017] [Indexed: 11/17/2022] Open
|