リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Basic Study of Synthetic MRI and Its Application to Multiple Sclerosis」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Basic Study of Synthetic MRI and Its Application to Multiple Sclerosis

萩原, 彰文 東京大学 DOI:10.15083/0002002352

2021.10.13

概要

In clinical practice, T1-weighted, T2-weighted, fluid-attenuated inversion recovery, and other contrast-weighted MRI images are assessed on the basis of relative signal differences. The signal intensity depends on sequence parameters and scanner settings, but also on B0 and B1 inhomogeneity, coil sensitivity profiles and radio frequency amplification settings, making quantitative comparisons difficult. Tissue relaxometry is a more direct approach to obtaining scanner-independent values. Absolute quantification of tissue properties by relaxometry has been reported in research settings for characterization of disease, assessment of disease activity, and monitoring of treatment effect. A number of methods have been proposed for simultaneous relaxometry of T1 and T2, but due to the additional scanning time required, these methods had not been widely introduced into clinical practice. Recently, QRAPMASTER (quantification of relaxation times and proton density by multiecho acquisition of a saturation-recovery using turbo spin-echo readout) pulse sequence for rapid simultaneous measurement of T1 and T2 relaxation times (and their inverses R1 and R2 relaxation rates) and proton density (PD), with correction of B1 field inhomogeneity, was proposed for full head coverage within approximately 6 minutes. These quantitative values allow post- acquisition generation of any contrast-weighted image via synthetic MRI, obviating the need for additional conventional T1-weighted and T2-weighted imaging required in routine clinical settings. The acquired maps are inherently aligned, thus avoiding potential errors due to image coregistration for multi-parametric quantification of a certain area. Myelin estimation can also be performed based on the acquired T1, T2, and PD values. The entire technique is referred to as synthetic MRI, or SyMRI. Herein, we conducted three consecutive studies to evaluate the quantitative values acquired by the QRAPMASTER pulse sequence for synthetic magnetic resonance imaging (MRI) and its application to multiple sclerosis (MS).

According to the Quantitative Imaging Biomarkers Alliance of the Radiological Society of North America, three metrology criteria are critical to the performance of a quantitative imaging biomarker: accuracy, repeatability, and reproducibility. Previous studies evaluated T1, T2, and PD values acquired with the QRAPMASTER sequence on a 1.5T scanner, by assessing accuracy, repeatability, and reproducibility using different head coils. However, to our knowledge, no study has compared quantitative values acquired with the MDME sequence on different scanners. The aim of the first study was to evaluate the linearity, bias, intrascanner repeatability, and interscanner reproducibility of quantitative values derived from the QRAPMASTER sequence for rapid simultaneous relaxometry. The NIST/ISMRM (National Institute of Standards and Technology/International Society for Magnetic Resonance in Medicine) phantom, containing spheres with standardized T1 and T2 relaxation times and proton density (PD), and 10 healthy volunteers, were scanned 10 times on different days and 2 times during the same session, using the QRAPMASTER sequence, on three 3 T scanners from different vendors. For healthy volunteers, brain volumetry and myelin estimation were performed based on the measured T1, T2, and PD. The measured phantom values were compared with reference values; volunteer values were compared with their averages across 3 scanners. The linearity of both phantom and volunteer measurements in T1, T2, and PD values was very strong (R2= 0.973–1.000, 0.979–1.000, and 0.982–0.999, respectively) The highest intrascanner coefficients of variation (CVs) for T1, T2, and PD were 2.07%, 7.60%, and 12.86% for phantom data, and 1.33%, 0.89%, and 0.77% for volunteer data, respectively. The highest interscanner CVs of T1, T2, and PD were 10.86%, 15.27%, and 9.95% for phantom data, and 3.15%, 5.76%, and 3.21% for volunteer data, respectively. Variation of T1 and T2 tended to be larger at higher values outside the range of those typically observed in brain tissue. The highest intrascanner and interscanner CVs for brain tissue volumetry were 2.50% and 5.74%, respectively, for cerebrospinal fluid. In conclusion, quantitative values derived from the QRAPMASTER sequence are overall robust for brain relaxometry and volumetry on 3 T scanners from different ven- dors. Caution is warranted when applying MDME sequence on anatomies with relaxometry values outside the range of those typically observed in brain tissue.

The aim of the second study was to validate the synthetic myelin imaging by comparing it with other myelin imaging methods. Magnetization transfer (MT) imaging has been widely used for estimating myelin content in the brain. Recently, two other approaches, namely SyMRI and the ratio of T1-weighted to T2-weighted images (T1w/T2w ratio), were also proposed as methods for measuring myelin. SyMRI and MT imaging have been reported to correlate well with actual myelin by histology. However, for T1w/T2w ratio, such evidence is limited. Investigation of correlation among different myelin imaging methods is scarce. Specifically, no study has examined the correlation of SyMRI as a myelin imaging tool with other methods. In 20 healthy adults, we examined the correlation between these three methods, using MT saturation index (MTsat) for MT imaging. After calibration, white matter (WM) to gray matter (GM) contrast was the highest for SyMRI among these three metrics. Even though SyMRI and MTsat showed strong correlation in the WM (r = 0.72), only weak correlation was found between T1w/T2w and SyMRI (r = 0.45) or MTsat (r = 0.38) (correlation coefficients significantly different from each other, with P values< 0.001). In subcortical and cortical GM, these measurements showed moderate to strong correlations to each other (r = 0.54 to 0.78). In conclusion, the high correlation between SyMRI and MTsat indicates that both methods are similarly suited to measure myelin in the WM, whereas T1w/T2w ratio may be less optimal.

The purpose of the third study was to evaluate SyMRI myelin imaging model that assesses myelin and edema for characterizing plaques, periplaque white matter, and normal-appearing white matter in patients with MS. We examined 3T SyMRI data from 21 patients with MS. The myelin partial volume, excess parenchymal water partial volume, the inverse of T1 and transverse T2 relaxation times (R1, R2), and proton density were compared among plaques, periplaque white matter, and normal-appearing white matter. All metrics differed significantly across the 3 groups (P < 0.001). Those in plaques differed most from those in normal-appearing white matter. The percentage changes of the metrics in plaques and periplaque white matter relative to normal-appearing white matter were significantly more different from zero for myelin volume fraction (mean, -61.59 ± 20.28% [plaque relative to normal-appearing white matter], and mean, -10.51 ± 11.41% [periplaque white matter relative to normal-appearing white matter]), and excess parenchymal water volume fraction (13.82 × 103 ± 49.47 × 103% and 51.33 × 102 ±155.31 × 102%) than for R1 (-35.23 × 13.93% and -6.08 ± 8.66%), R2 (-21.06 ± 11.39% and - 4.79 ± 6.79%), and PD (23.37 ± 10.30% and 3.37 ± 4.24%). SyMRI captures white matter damage in MS. Myelin volume fraction and excess parenchymal water volume fraction are more sensitive to the MS disease process than R1, R2, and PD. MVF and EPWVF could be useful estimators of disease burden in patients with MS.

In summary, I conclude that QRAPMASTER can perform quantitative measurement of the brain with high accuracy and precision in a short acquisition time. The technique may be clinically useful in the assessment of brain disorders including MS.

この論文で使われている画像

参考文献

1. west J, Aalto A, Tisell , et al. Normal appearing and diffusely abnormal white matter in patients with multiple sclerosis assessed with quantitative MR. PLoS One 9:e95161. 2014.

2. Horsthuis K, Nederveen AJ, de Feiter MW, et al. Mapping of T1-values and Gadolinium- concentrations in MRI as indicator of disease activity in luminal 7rohn's disease: a feasibility study. J Magn Reson Imaging 29:488-93. 2009.

3. wagner M7, Lukas P, Her×og M, et al. MRI and proton-NMR relaxation times in diagnosis and therapeutic monitoring of squamous cell carcinoma. Eur Radiol 4:314-23. 1994.

4. Ma D, Gulani V, Seiberlich N, et al. Magnetic resonance fingerprinting. Nature 495:18V-92. 2013.

5. Newbould RD, Skare ST, Alley MW, et al. Three-dimensional T(1), T(2) and proton density mapping with inversion recovery balanced SSFP. Magn Reson Imaging 28:13V4-82. 2010.

6. Ehses P, Seiberlich N, Ma D, et al. IR TrueFISP with a golden-ratio-based radial readout: fast quantification of T1, T2, and proton density. Magn Reson Med 69:V1-81. 2013.

7. Deoni S7, Rutt BK, Arun T, et al. Gleaning multicomponent T1 and T2 information from steady-state imaging data. Magn Reson Med 60:1372-87. 2008.

8. Hagiwara A, warntjes M, Hori M, et al. SyMRI of the Brain: Rapid Quantification of Relaxation Rates and Proton Density, with Synthetic MRI, Automatic Brain Segmentation, and Myelin Measurement. Invest Radiol 52:647-57. 2017.

9. Lee SM, 7hoi wH, wou SK, et al. Age-Related 7hanges in Wissue Value Properties in 7hildren: Simultaneous Quantification of Relaxation Wimes and Proton Density Using Synthetic Magnetic Resonance Imaging. Invest Radiol 53:236-45. 2018.

10. warntjes JB, Engstrom M, Tisell A, et al. Brain characterization using normalized quantitative magnetic resonance imaging. PLoS One 8:eV0864. 2013.

11. Hagiwara A, Hori M, Suzuki M, et al. 7ontrast-enhanced synthetic MRI for the detection of brain metastases. Acta Radiol Open 5:2058460115626V5V. 2016.

12. Hagiwara A, Nakazawa M, Andica 7, et al. Dural Enhancement in a Patient with Sturge-weber Syndrome Revealed by Double Inversion Recovery 7ontrast Using Synthetic MRI. Magn Reson Med Sci 15:151-2. 2016.

13. Blystad I, warntjes JB, Smedby O, et al. Synthetic MRI of the brain in a clinical setting. Acta Radiol 53:1158-63. 2012.

14. west J, warntjes JB, Lundberg P. Novel whole brain segmentation and volume estimation using quantitative MRI. Eur Radiol 22:998-100V. 2012.

15. warntjes M, Engstrom M, Wisell A, et al. Modeling the Presence of Myelin and Edema in the Brain Based on Multi-Parametric Quantitative MRI. Front Neurol V:16. 2016.

16. Jack 7R, Jr., Shiung MM, Gunter JL, et al. 7omparison of different MRI brain atrophy rate measures with clinical disease progression in AD. Neurology 62:591-600. 2004.

17. Miller DH, Barkhof F, Frank JA, et al. Measurement of atrophy in multiple sclerosis: pathological basis, methodological aspects and clinical relevance. Brain 125:16V6-95. 2002.

18. Andica 7, Hagiwara A, Hori M, et al. Automated brain tissue and myelin volumetry based on quantitative MR imaging with various in-plane resolutions. J Neuroradiol 45:164-68. 2018.

19. warntjes JBM, Persson A, Berge J, et al. Myelin Detection Using Rapid Quantitative MR Imaging 7orrelated to Macroscopically Registered Luxol Fast Blue-Stained Brain Specimens. AJNR Am J Neuroradiol 38:1096-102. 2017.

20. Raunig DL, McShane LM, Pennello G, et al. Quantitative imaging biomarkers: a review of statistical methods for technical performance assessment. Stat Methods Med Res 24:27-67. 2015.

21. warntjes JB, Leinhard OD, west J, et al. Rapid magnetic resonance quantification on the brain: Optimization for clinical usage. Magn Reson Med 60:320-9. 2008.

22. Krauss W, Gunnarsson M, Andersson T, et al. Accuracy and reproducibility of a quantitative magnetic resonance imaging method for concurrent measurements of tissue relaxation times and proton density. Magn Reson Imaging 33:584-91. 2015.

23. Russek S, Boss M, Jackson E, et al. 7haracteri×ation of NISW/ISMRM MRI System Phantom. In Proceedings of the 20th Annual Meeting of ISMRM, Melbourne, Victoria, Austraia, 2012 Abstract 2456.

24. Keenan K, Stupic K, Boss M, et al. Multi-site, multi-vendor comparison of T1 measurement using ISMRM/NISW system phantom. In Proceedings of the 24th Annual Meeting of ISMRM, Singapore, 2016 Abstract 3290.

25. Levesque IR, Pike GB. 7haracterizing healthy and diseased white matter using quantitative magnetization transfer and multicomponent T(2) relaxometry: A unified view via a four-pool model. Magn Reson Med 62:1487-96. 2009.

26. Ambarki K, Lindqvist T, wahlin A, et al. Evaluation of automatic measurement of the intracranial volume based on quantitative MR imaging. AJNR Am J Neuroradiol 33:1951-6. 2012.

27. Stikov N, Boudreau M, Levesque IR, et al. On the accuracy of W1 mapping: searching for common ground. Magn Reson Med 73:514-22. 2015.

28. McPhee K7, wilman AH. Transverse relaxation and flip angle mapping: Evaluation of simultaneous and independent methods using multiple spin echoes. Magn Reson Med 77:2057-65. 2017.

29. whittall KP, MacKay AL, Graeb DA, et al. In vivo measurement of T2 distributions and water contents in normal human brain. Magn Reson Med 37:34-43. 1997.

30. Abbas S, Gras V, Mollenhoff K, et al. Analysis of proton-density bias corrections based on T1 measurement for robust quantification of water content in the brain at 3 Tesla. Magn Reson Med 72:1735-45. 2014.

31. Bauer 7M, Jara H, Killiany R, et al. whole brain quantitative T2 MRI across multiple scanners with dual echo FSE: applications to AD, M7I, and normal aging. Neuroimage 52:508-14. 2010.

32. Deoni S7L, Williams S7R, Jezzard P, et al. Standardized structural magnetic resonance imaging in multicentre studies using quantitative T1 and T2 imaging at 1.5 T. Neuroimage 40:662-71. 2008.

33. Davies GR, Hadjiprocopis A, Altmann DR, et al. Normal-appearing grey and white matter T1 abnormality in early relapsing-remitting multiple sclerosis: a longitudinal study. Mult Scler 13:169-77. 2007.

34. Reitz S7, Hof SM, Fleischer V, et al. Multi-parametric quantitative MRI of normal appearing white matter in multiple sclerosis, and the effect of disease activity on T2. Brain Imaging Behav 11:744-53. 2017.

35. Wang H, wuan H, Shu L, et al. Prolongation of T(2) relaxation times of hippocampus and amygdala in Alzheimer's disease. Neurosci Lett 363:150-3. 2004.

36. Measurement in MRI. In: 7ercignani M, Dowell NG, Tofts PS, eds. Quantitative MRI of the Brain 2nd ed. Tofts PS. NW FL: 7R7 Press; 10-11. 2018.

37. Park S, Kwack KS, Lee WJ, et al. Initial experience with synthetic MRI of the knee at 3T: comparison with conventional T1 weighted imaging and T2 mapping. Br J Radiol 90:20170350. 2017.

38. 7hougar L, Hagiwara A, Andica 7, et al. Synthetic MRI of the knee: new perspectives in musculoskeletal imaging and possible applications for the assessment of bone marrow disorders. Br J Radiol:20170886. 2018.

39. Lee SH, Lee WH, Song HT, et al. Quantitative T2 Mapping of Knee 7artilage: 7omparison between the Synthetic MR Imaging and the 7PMG Sequence. Magn Reson Med Sci2018.

40. Landman BA, Huang AJ, Gifford A, et al. Multi-parametric neuroimaging reproducibility: a 3- T resource study. Neuroimage 54:2854-66. 2011.

41. Sampat MP, Healy B7, Meier DS, et al. Disease modeling in multiple sclerosis: assessment and quantification of sources of variability in brain parenchymal fraction measurements. Neuroimage 52:1367-73. 2010.

42. Huppertz HJ, Kroll-Seger J, Kloppel S, et al. Intra- and interscanner variability of automated voxel-based volumetry based on a 3D probabilistic atlas of human cerebral structures. Neuroimage 49:2216-24. 2010.

43. de Boer R, Vrooman HA, Ikram MA, et al. Accuracy and reproducibility study of automatic MRI brain tissue segmentation methods. Neuroimage 51:1047-56. 2010.

44. Granberg T, Uppman M, Hashim F, et al. 7linical Feasibility of Synthetic MRI in Multiple Sclerosis: A Diagnostic and Volumetric Validation Study. AJNR Am J Neuroradiol 37:1023-9. 2016.

45. Akkus S, Galim×ianova A, Hoogi A, et al. Deep Learning for Brain MRI Segmentation: State of the Art and Future Directions. J Digit Imaging 30:449-59. 2017.

46. Nguyen TD, Deh K, Monohan E, et al. Feasibility and reproducibility of whole brain myelin water mapping in 4 minutes using fast acquisition with spiral trajectory and adiabatic T2prep (FAST- T2) at 3T. Magn Reson Med 76:456-65. 2016.

47. de Hoz L, Simons M. The emerging functions of oligodendrocytes in regulating neuronal network behaviour. Bioessays 37:60-9. 2015.

48. Duval T, Stikov N, 7ohen-Adad J. Modeling white matter microstructure. Funct Neurol 31:217-28. 2016.

49. Wu M, Kumar A, Yang S. Development and aging of superficial white matter myelin from young adulthood to old age: Mapping by vertex-based surface statistics (VBSS). Hum Brain Mapp 37:1759-69. 2016.

50. van Buchem MA, Steens S7, Vrooman HA, et al. Global estimation of myelination in the developing brain on the basis of magnetization transfer imaging: a preliminary study. AJNR Am J Neuroradiol 22:762-6. 2001.

51. Dean D7, 3rd, O'Muircheartaigh J, Dirks H, et al. Estimating the age of healthy infants from quantitative myelin water fraction maps. Hum Brain Mapp 36:1233-44. 2015.

52. Ihara M, Polvikoski TM, Hall R, et al. Quantification of myelin loss in frontal lobe white matter in vascular dementia, Alzheimer's disease, and dementia with Lewy bodies. Acta Neuropathol 119:579-89. 2010.

53. Bakshi R, Thompson AJ, Rocca MA, et al. MRI in multiple sclerosis: current status and future prospects. Lancet Neurol 7:615-25. 2008.

54. McAllister A, Leach J, west H, et al. Quantitative Synthetic MRI in 7hildren: Normative Intracranial Tissue Segmentation Values During Development. AJNR Am J Neuroradiol [Epub ahed of print]2017.

55. Kim HG, Moon WJ, Han J, et al. Quantification of myelin in children using multiparametric quantitative MRI: a pilot study. Neuroradiology [Epub ahead of print]2017.

56. Hagiwara A, Andica 7, Hori M, et al. Synthetic MRI showed increased myelin partial volumem in the white matter of a patient with Sturge-weber syndrome. Neuroradiology [Epub ahead of print]2017.

57. Wallaert L, Hagiwara A, Andica 7, et al. Whe Advantage of SyMRI for the Visualization of Anterior Temporal Pole Lesions by Double Inversion recovery (DIR), Phase-Sensitive Inversion Recovery (PSIR), and Myelin Images in a Patient with 7ADASIL. Magn Reson Med Sci [in press]2017.

58. Alonso-Ortiz E, Levesque IR, Pike GB. MRI-based myelin water imaging: A technical review. Magn Reson Med 73:70-81. 2015.

59. MacKay A, Laule 7, Vavasour I, et al. Insights into brain microstructure from the T2 distribution. Magn Reson Imaging 24:515-25. 2006.

60. Mezer A, Yeatman JD, Stikov N, et al. Quantifying the local tissue volume and composition in individual brains with magnetic resonance imaging. Nat Med 19:1667-72. 2013.

61. Henkelman RM, Stanisz GJ, Graham SJ. Magneti×ation transfer in MRI: a review. NMR Biomed 14:57-64. 2001.

62. 7ampbell JS, Leppert IR, Narayanan S, et al. Promise and pitfalls of g-ratio estimation with MRI. Neuroimage [Epub ahead of print]2017.

63. Schmierer K, Scaravilli F, Altmann DR, et al. Magnetization transfer ratio and myelin in postmortem multiple sclerosis brain. Ann Neurol 56:407-15. 2004.

64. Filippi M, 7ampi A , Dousset V, et al. A magnetization transfer imaging study of normal- appearing white matter in multiple sclerosis. Neurology 45:478-82. 1995.

65. Mottershead JP, Schmierer K, 7lemence M, et al. High field MRI correlates of myelin content and axonal density in multiple sclerosis--a post-mortem study of the spinal cord. J Neurol 250:1293-301. 2003.

66. Harkins KD, Xu J, Dula AN, et al. The microstructural correlates of T1 in white matter. Magn Reson Med 75:1341-5. 2016.

67. Schmierer K, wheeler-Kingshott 7A, Wozer DJ, et al. Quantitative magnetic resonance of postmortem multiple sclerosis brain before and after fixation. Magn Reson Med 59:268-77. 2008.

68. Helms G, Dathe H, Kallenberg K, et al. High-resolution maps of magnetization transfer with inherent correction for RF inhomogeneity and T1 relaxation obtained from 3D FLASH MRI. Magn Reson Med 60:1396-407. 2008.

69. Lema A, Bishop 7, Malik O, et al. A 7omparison of Magneti×ation Wransfer Methods to Assess Brain and 7ervical 7ord Microstructure in Multiple Sclerosis. J Neuroimaging 27:221-26. 2017.

70. Glasser MF, Van Essen D7. Mapping human cortical areas in vivo based on myelin content as revealed by T1- and T2-weighted MRI. J Neurosci 31:11597-616. 2011.

71. Ma S, Shang N. 7ross-population myelination covariance of human cerebral cortex. Hum Brain Mapp 38:4730-43. 2017.

72. Ganzetti M, Wenderoth N, Mantini D. whole brain myelin mapping using T1- and T2- weighted MR imaging data. Front Hum Neurosci 8:671. 2014.

73. Shafee R, Buckner RL, Fischl B. Gray matter myelination of 1555 human brains using partial volume corrected MRI images. Neuroimage 105:473-85. 2015.

74. Grydeland H, Walhovd KB, Tamnes 7K, et al. Intracortical myelin links with performance variability across the human lifespan: results from T1- and T2-weighted MRI myelin mapping and diffusion tensor imaging. J Neurosci 33:18618-30. 2013.

75. Soun JE, Liu MS, 7auley KA, et al. Evaluation of neonatal brain myelination using the T1- and T2-weighted MRI ratio. J Magn Reson Imaging 46:690-96. 2016.

76. Lee K, 7herel M, Budin F, et al. Early Postnatal Myelin 7ontent Estimate of white Matter via T1w/T2w Ratio. Proc SPIE Int Soc Opt Eng 94172015.

77. Arshad M, Stanley JA, Raz N. Test-retest reliability and concurrent validity of in vivo myelin content indices: Myelin water fraction and calibrated T1 w/T2 w image ratio. Hum Brain Mapp 38:1780-90. 2017.

78. Nakamura K, 7hen JT, Ontaneda D, et al. T1-/T2-weighted ratio differs in demyelinated cortex of multiple sclerosis. Ann Neurol [Epub ahead of print]2017.

79. Righart R, Biberacher V, Jonkman LE, et al. 7ortical pathology in MS detected by the T1/T2- weighted ratio from routine MRI. Ann Neurol [Epub ahead of print]2017.

80. Uddin MN, Figley TD, Marrie RA, et al. 7an T1 w/T2 w ratio be used as a myelin-specific measure in subcortical structures? 7omparisons between FSE-based T1 w/T2 w ratios, GRASE-based T1 w/T2 w ratios and multi-echo GRASE-based myelin water fractions. NMR Biomed 312018.

81. Fazekas F, 7hawluk JB, Alavi A, et al. MR signal abnormalities at 1.5 T in Alzheimer's dementia and normal aging. AJR Am J Roentgenol 149:351-6. 1987.

82. Helms G, Dathe H, Dechent P. Modeling the influence of TR and excitation flip angle on the magneti×ation transfer ratio (MTR) in human brain obtained from 3D spoiled gradient echo MRI. Magn Reson Med 64:177-85. 2010.

83. Weiskopf N, Suckling J, Williams ;, et al. Quantitative multi-parameter mapping of R1, PD(2), MT, and R2(2) at 3T: a multi-center validation. Front Neurosci 7:95. 2013.

84. Morrell GR, Schabel M7. An analysis of the accuracy of magnetic resonance flip angle measurement methods. Phys Med Biol 55:6157-74. 2010.

85. Hua K, Shang J, Wakana S, et al. Tract probability maps in stereotaxic spaces: analyses of white matter anatomy and tract-specific quantification. Neuroimage 39:336-47. 2008.

86. Wakana S, 7aprihan A, Panzenboeck MM, et al. Reproducibility of quantitative tractography methods applied to cerebral white matter. Neuroimage 36:630-44. 2007.

87. Tzourio-Mazoyer N, Landeau B, Papathanassiou D, et al. Automated anatomical labeling of activations in SPM using a macroscopic anatomical parcellation of the MNI MRI single-subject brain. Neuroimage 15:273-89. 2002.

88. Schmahmann JD, Doyon J, McDonald D, et al. Three-dimensional MRI atlas of the human cerebellum in proportional stereotaxic space. Neuroimage 10:233-60. 1999.

89. Jenkinson M, Bannister P, Brady M, et al. Improved optimization for the robust and accurate linear registration and motion correction of brain images. Neuroimage 17:825-41. 2002.

90. Jenkinson M, Beckmann 7F, Behrens TE, et al. FSL. Neuroimage 62:782-90. 2012.

91. Shang W, Brady M, Smith S. Segmentation of brain MR images through a hidden Markov random field model and the expectation-maximization algorithm. IEEE Trans Med Imaging 20:45-57. 2001.

92. Mohammadi S, 7arey D, Dick F, et al. whole-Brain In-vivo Measurements of the Axonal G- Ratio in a Group of 37 Healthy Volunteers. Front Neurosci 9:441. 2015.

93. Stikov N, 7ampbell JS, Stroh W, et al. In vivo histology of the myelin g-ratio with magnetic resonance imaging. Neuroimage 118:397-405. 2015.

94. Mukaka MM. Statistics corner: A guide to appropriate use of correlation coefficient in medical research. Malawi Med J 24:69-71. 2012.

95. Steiger JH. Tests for 7omparing Elements of a 7orrelation Matrix. Psychological Bulletin 87:245-51. 1980.

96. Thiessen JD, Shang w, Shang H, et al. Quantitative MRI and ultrastructural examination of the cuprizone mouse model of demyelination. NMR Biomed 26:1562-81. 2013.

97. Hagiwara A, Hori M, Yokoyama K, et al. Analysis of white Matter Damage in Patients with Multiple Sclerosis via a Novel In Vivo Magnetic Resonance Method for Measuring Myelin, Axons, and Gratio. AJNR Am J Neuroradiol [Epub ahead of print]2017.

98. Hagiwara A, Hori M, Yokoyama K, et al. Utility of a Multiparametric Quantitative MRI Model That Assesses Myelin and Edema for Evaluating Plaques, Periplaque white Matter, and Normal-Appearing white Matter in Patients with Multiple Sclerosis: A Feasibility Study. AJNR Am J Neuroradiol 38:237-42. 2017.

99. Sjobeck M, Haglund M, Englund E. Decreasing myelin density reflected increasing white matter pathology in Alzheimer's disease--a neuropathological study. Int J Geriatr Psychiatry 20:919-26. 2005.

100. Khodanovich MY, Sorokina IV, Glazacheva VY, et al. Histological validation of fast macromolecular proton fraction mapping as a quantitative myelin imaging method in the cuprizone demyelination model. Sci Rep 7:46686. 2017.

101. Dula AN, Gochberg DF, Valentine HL, et al. Multiexponential T2, magnetization transfer, and quantitative histology in white matter tracts of rat spinal cord. Magn Reson Med 63:902-9. 2010.

102. Glasser MF, Goyal MS, Preuss TM, et al. Trends and properties of human cerebral cortex: correlations with cortical myelin content. Neuroimage 93:165-75. 2014.

103. Berman S, west KL, Does MD, et al. Evaluating g-ratio weighted changes in the corpus callosum as a function of age and sex. Neuroimage [Epub ahead of print]2017.

104. 7ercignani M, Giulietti G, Dowell NG, et al. 7haracterizing axonal myelination within the healthy population: a tract-by-tract mapping of effects of age and gender on the fiber g-ratio. Neurobiol Aging 49:109-18. 2017.

105. Laule 7, Leung E, Lis DK, et al. Myelin water imaging in multiple sclerosis: quantitative correlations with histopathology. Mult Scler 12:747-53. 2006.

106. Andica 7, Hagiwara A, Hori M, et al. Automated Brain Wissue and Myelin Volumetry Based on Quantitative MR Imaging with Various In-plane Resolutions. J Neuroradiol [Epub ahead of print]2017.

107. Vavasour IM, Laule 7, Li DK, et al. Is the magnetization transfer ratio a marker for myelin in multiple sclerosis? J Magn Reson Imaging 33:713-8. 2011.

108. Polman 7H, Reingold S7, Banwell B, et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann Neurol 69:292-302. 2011.

109. Filippi M, Rocca MA, De Stefano N, et al. Magnetic resonance techniques in multiple sclerosis: the present and the future. Arch Neurol 68:1514-20. 2011.

110. Yoshida M, Hori M, Yokoyama K, et al. Diffusional kurtosis imaging of normal-appearing white matter in multiple sclerosis: preliminary clinical experience. Jpn J Radiol 31:50-5. 2013.

111. Guo A7, MacFall JR, Provenzale JM. Multiple sclerosis: diffusion tensor MR imaging for evaluation of normal-appearing white matter. Radiology 222:729-36. 2002.

112. Hori M, Yoshida M, Yokoyama K, et al. Multiple sclerosis: Benefits of q-space imaging in evaluation of normal-appearing and periplaque white matter. Magn Reson Imaging 32:625-9. 2014.

113. McDonald WI, 7ompston A, Edan G, et al. Recommended diagnostic criteria for multiple sclerosis: guidelines from the International Panel on the diagnosis of multiple sclerosis. Ann Neurol 50:121-7. 2001.

114. Polman 7H, Reingold S7, Edan G, et al. Diagnostic criteria for multiple sclerosis: 2005 revisions to the "McDonald 7riteria". Ann Neurol 58:840-6. 2005.

115. Seewann A, Vrenken H, van der Valk P, et al. Diffusely abnormal white matter in chronic multiple sclerosis: imaging and histopathologic analysis. Arch Neurol 66:601-9. 2009.

116. Kurtzke JF. A new scale for evaluating disability in multiple sclerosis. Neurology 5:580-3. 1955.

117. MacKay A, Whittall K, Adler J, et al. In vivo visualization of myelin water in brain by magnetic resonance. Magn Reson Med 31:673-7. 1994.

118. Nijeholt GJ, Bergers E, Kamphorst W, et al. Post-mortem high-resolution MRI of the spinal cord in multiple sclerosis: a correlative study with conventional MRI, histopathology and clinical phenotype. Brain 124:154-66. 2001.

119. Lieury A, 7hanal M, Androdias G et al. Tissue remodeling in periplaque regions of multiple sclerosis spinal cord lesions. Glia 62:1645-58. 2014.

120. Narayanan S, Fu L, Pioro E, et al. Imaging of axonal damage in multiple sclerosis: spatial distribution of magnetic resonance imaging lesions. Ann Neurol 41:385-91. 1997.

121. Perry VH, Anthony D7. Axon damage and repair in multiple sclerosis. Philos Trans R Soc Lond B Biol Sci 354:1641-7. 1999.

122. Dziedzic W, Metz I, Dallenga T, et al. Wallerian degeneration: a major component of early axonal pathology in multiple sclerosis. Brain Pathol 20:976-85. 2010.

123. Simons M, Misgeld T, Kerschensteiner M. A unified cell biological perspective on axon-myelin injury. J 7ell Biol 206:335-45. 2014.

124. McDonald WI, Miller DH, Barnes D. The pathological evolution of multiple sclerosis. Neuropathol Appl Neurobiol 18:319-34. 1992.

125. Helms G, Stawiarz L, Kivisakk P, et al. Regression analysis of metabolite concentrations estimated from locali×ed proton MR spectra of active and chronic multiple sclerosis lesions. Magn Reson Med 43:102-10. 2000.

126. Kolind S, Matthews L, Johansen-Berg H, et al. Myelin water imaging reflects clinical variability in multiple sclerosis. Neuroimage 60:263-70. 2012.

127. Faizy TD, Thaler 7, Kumar D, et al. Heterogeneity of Multiple Sclerosis Lesions in Multislice Myelin Water Imaging. PLoS One 11:e0151496. 2016.

参考文献をもっと見る