Diffusion Goes Multi-Modal: Myelin Mapping
Mark D Does1

1Vanderbilt University, United States

Synopsis

Myelin plays a central role in determining MRI contrast in neuronal tissues, and, consequently, there are many MRI approaches measuring myelin content. This presentation will review several common approaches as well as some recent and developing approaches.

Target Audience

Physicians and scientists wanting to explore methodologies for quantitative microstructural imaging techniques for white matter; physicians and scientists engaged in advanced white matter imaging for scientific and clinical research.

Objectives

To provide an overview of current MRI methods that can be used to estimate myelin content in neuronal tissue. To this end, the lecture will cover:

  • the role of myelin in MRI signal models of neuronal tissue
  • pulse sequence and corresponding analysis methods to quantitatively probe these models
  • pros, cons, and unknowns of various methods

Overview

Early MRI studies of human brain revealed strong contrast between white and gray matter, which suggested the importance of myelin in determining water proton nuclear magnetic relaxation rates. Since then, many experimental studies have aimed to model the role of myelin in MRI contrast, and corresponding method developments have aimed to establish robust MRI measures of myelin content. While some model details remain elusive, it is clear that myelin is a prominent contributor to water proton relaxation and several MRI methods have been demonstrated to quantitatively report on myelin content with a least some reasonable level of specificity.

Myelin is comprised of tightly wrapped layers of oligodendrocyte or Schwann cell membranes, with water residing in the relatively thin (~ 3 nm) intra- and extra-cellular spaces between adjacent membrane layers. Consequently, the water density per volume in myelin is relatively low, about 1/2 that in other soft tissue, and in this confined environment, water protons experience relatively frequent interactions with the non-aqueous components of myelin. These interactions result in a relatively rapid transverse and longitudinal relaxation of the water proton magnetization, and include a relatively large exchange of longitudinal magnetization between water and macromolecular protons, commonly referred to a magnetization transfer (MT). Each of these factors (low water density, rapid relaxation, large MT effect) can contribute contrast that reports on myelin content. Considering a more complete model of white matter or nerve, the relationships between these contrasts and myelin content becomes somewhat more complicated.

Water protons in white matter and nerve can be modeled in terms of three micro-anatomical compartments: intra-axonal (i.e., axoplasm), extra-axonal (space in between axons), and myelin. Macromolecular protons, although not directly observed in most MRI due to extremely fast relaxation of transverse magnetization, do affect water proton magnetization through MT. Including macromolecular protons therefore extends the compartmental model to six proton pools with coupling of longitudinal magnetization between the water and macromolecule pools in each anatomical compartment. Inter-compartmental water exchange, particularly between myelin and each of the other two water compartments, may also be relevant, resulting in a six-pool model with at least five coupling pathways. While attempts have been made to parameterize this model (1-3), it is far too complex to directly inform on clinically-practical MRI. Instead, the role of myelin in MRI contrast is best appreciated in terms of a simpler model.

For measurement of transverse relaxation (that is, both T2 and T2* time constants), it is reasonable to ignore the macromolecular protons because their transverse magnetization is effectively zero and time invariant. Further, it is common to ignore the inter-compartmental water exchange on the assumption that it is relatively slow compared with the transverse relaxation. The faster relaxing myelin water signal can then be distinguished from intra- and extra-axonal water signals by multi-exponential signal analysis (4). The most common use of this model is for multi-exponential T2 analysis of multiple spin echo images (5-7), through which the term myelin water fraction (MWF) was coined (5). Multiple experimental studies support the conclusion that MWF is a correlative measure of myelin content (8-15). More recently, similar multi-exponential T2* analysis has been applied to multiple gradient echo images (16-20). The T2*approach has the advantage of using a simpler and more easily accelerated pulse sequence, but comes with the additional complication of needing to correct for macroscopic field variations and, potentially, susceptibility induced frequency shifts (21-24). For either T2 or T2*, there are two shortcomings to this approach: 1) the signal to noise ratio requirement of the multi-exponential analysis are high, and 2) the assumption of slow inter-compartmental water may not always hold, resulting in an systematic underestimation of myelin content by MWF measurement (25-29). This exchange effect is likely greater in smaller axons with thinner myelin, resulting in a possible axon-size dependence of the MWF measurement.

For measurement of longitudinal relaxation, there are a few options for model simplification. Applying the same slow-exchange, water-only model, MWF can be estimated by multi-exponential T1 analysis of inversion-recovery image data (30). However, because T1 > T2, the effects of inter-compartment water exchange can only be worse in this scenario. Also, when considering longitudinal magnetization, the macromolecular proton pool cannot, in general, be ignored. The water and macromolecular longitudinal magnetizations will respond differently to radio frequency (RF) pulses, and this will in-turn effect the MRI signal through MT (31-32). One can try to avoid this effect through appropriate sequence timings, although, as noted below, if the MT effect is also strongly dependent upon myelin, its confound in T1-MWF measurements may not be a practical problem.

Alternatively, rather than trying to mitigate the MT effect, one can make it the target signal, with the reasoning the myelin is the dominate contributing anatomical compartment. This can be implemented in a number of ways, but two common approaches are: 1) saturating the macromolecular proton magnetization independently of the water magnetization using off-resonance RF and then observing the MT effect on the water magnetization, or 2) saturating or inverting the water magnetization independently of the macromolecular magnetization and then, again, observing the effect MT effect on the water magnetization. Of course, myelin is not the only source of MT contrast, but MT contrast has been shown to correlate with myelin content (12,33-35), and a more recent variation on MT contrast known as inhomogeneous MT (ihMT) may offer a greater level of specificity to myelin (36-38).

A third approach for relating myelin to longitudinal relaxation, which is a logical extension of the MT model, is to assume that MT is relatively fast and the dominant contributor to water proton T1. If one again assumes that the dominant source of MT is myelin, then with this model a measure of 1/T1 (=R1) is proportional to the concentration of myelin (39-41) and even appropriately calibrated T1-contrast can do the same (42–44). This model too is an oversimplification, but has gained popularity as a fast, high resolution approach to mapping myelin, particularly in the cortex (45-47).

In summary, myelin is a prominent contributor to both longitudinal and transverse relaxation as well as overall proton density, and so any number of quantitative measures of these characteristic can be related to myelin content. Presently, there is no one approach that is decidedly better than the others, but multi-exponential T2 and quantitative MT likely use the most well founded models and have been the most experimentally well supported, while R1 measures, although perhaps less specific to myelin, offer faster and higher resolution acquisitions. For further reading on the material, the reader is directed to a collection of recent review articles related to probing brain microstructure with MRI (48-50).

Acknowledgements

No acknowledgement found.

References

1. Stanisz, G.J., Kecojevic, A., Bronskill, M.J., Henkelman, R.M., 1999. Characterizing white matter with magnetization transfer and T(2). Magn. Reson. Med. 42 (6), 1128–1136.

2. Bjarnason, T.A., Vavasour, I.M., Chia, C.L.L., MacKay, A.L., 2005. Characterization of the NMR behavior of white matter in bovine brain. Magn. Reson. Med. 54 (5), 1072–1081.

3. Barta, R., Kalantari, S., Laule, C., Vavasour, I.M., MacKay, A.L., Michal, C.A., 2015. Modeling T1 and T2 relaxation in bovine white matter. J. Magn. Reson. 259, 56–67.

4. Whittall, K.P., MacKay, A.L., 1989. Quantitative interpretation of NMR relaxation data. J. Magn. Reson. 84, 134–152.

5. MacKay, A., Whittall, K., Adler, J., Li, D., Paty, D., Graeb, D., 1994. In-vivo visualization of myelin water in brain by magnetic-resonance. Magn. Reson. Med. 31 (6), 673–677.

6. Whittall, K., MacKay, A., Graeb, D., Nugent, R., Li, D., Paty, D., 1997. In vivo measurement of T-2 distributions and water contents in normal human brain. Magn. Reson. Med. 37 (1), 34–43.

7. Prasloski, T., Rauscher, A., Mackay, A.L., Hodgson, M., Vavasour, I.M., Laule, C., M€adler, B., 2012b. Rapid whole cerebrum myelin water imaging using a 3D GRASE sequence. Neuroimage 63 (1), 533–539.

8. Menon, R.S., Rusinko, M.S., Allen, P.S., 1992. Proton relaxation studies of water compartmentalization in a model neurological system. Magn. Reson. Med. 28 (2), 264–274.

9. Stewart, W.A., Mackay, A.L., Whittall, K.P., Moore, G.R.W., Paty, D.W., 1993. Spin-spin relaxation in experimental allergic Encephalomyelitis. Analysis of CPMG data using a non-linear least squares method and linear inverse theory. Magn. Reson. Med. 29 (6), 767–775.

10. Does, M., Snyder, R., 1996. Multiexponential T-2 relaxation in degenerating peripheral nerve. Magn. Reson. Med. 35 (2), 207–213.

11. Webb, S., Munro, C., Midha, R., Stanisz, G., 2003. Is multicomponent T-2 a good measure of myelin content in peripheral nerve? Magn. Reson. Med. 49 (4), 638–645.

12. Odrobina, E.E., Lam, T.Y., Pun, T., Midha, R., Stanisz, G.J., 2005. MR properties of excised neural tissue following experimentally induced demyelination. NMR Biomed. 18 (5), 277–284.

13. Laule, C., Leung, E., Li, D.K., Traboulsee, A.L., Paty, D.W., MacKay, A.L., Moore, G.R., 2006. Myelin water imaging in multiple sclerosis: quantitative correlations with histopathology. Mult. Scler. 12 (6), 747–753.

14. Laule, C., Kozlowski, P., Leung, E., Li, D.K.B., Mackay, A.L., Moore, G.R.W., 2008. Myelin water imaging of multiplesclerosis at 7 T: correlations with histopathology. Neuroimage 40 (4), 1575–1580.

15. West, K.L., Kelm, N.D., Carson, R.P., Gochberg, D.F., Ess, K.C., 2018. Myelin volume fraction imaging with MRI. Neuroimage 182 511–521.

16. Du, Y.P., Chu, R., Hwang, D., Brown, M.S., Kleinschmidt-DeMasters, B.K., Singel, D., Simon, J.H., 2007. Fast multislice mapping of the myelin water fraction using multicompartment analysis of T2* decay at 3T: a preliminary postmortem study. Magn. Reson. Med. 58 (5), 865–870.

17. Hwang, D., Kim, D.-H., Du, Y.P., 2010. In vivo multi-slice mapping of myelin water content using T2* decay. Neuroimage 52 (1), 198–204.

18. Alonso-Ortiz, E., Levesque, I.R., Paquin, R., Pike, G.B., 2017. Field inhomogeneity correction for gradient echo myelin water fraction imaging. Magn. Reson. Med. 78 (1), 49–57.

19. Alonso-Ortiz, E., Levesque, I.R., Pike, G.B., 2018. Impact of magnetic susceptibility anisotropy at 3 T and 7 T on T2*-based myelin water fraction imaging. Neuroimage 182 370-378.

20. Alonso-Ortiz, E., Levesque, I.R., Pike, G.B., 2018. Multi-gradient-echo myelin water fraction imaging: comparison to the multi-echo-spin-echo technique. Magn. Reson. Med 79, 1439-1446

21. van Gelderen, P., de Zwart, J.A., Lee, J., Sati, P., Reich, D.S., Duyn, J.H., 2012. Nonexponential T2* decay in white matter. Magn. Reson. Med. 67 (1), 110–117.

22. Sati, P., van Gelderen, P., Silva, A.C., Reich, D.S., Merkle, H., de Zwart, J.A., Duyn, J.H., 2013. Micro-compartment specific T2* relaxation in the brain. Neuroimage 77, 268–278.

23. Nam, Y., Lee, J., Hwang, D., Kim, D.-H., 2015. Improved estimation of myelin water fraction using complex model fitting. Neuroimage 116, 214–221.

24. Nunes, D., Cruz, T.L., Jespersen, S.N., Shemesh, N., 2017. Mapping axonal density and average diameter using non-monotonic time-dependent gradient-echo MRI. J. Magn. Reson. 277, 117–130.

25. Sled, J.G., Levesque, I., Santos, A.C., Francis, S.J., Narayanan, S., Brass, S.D., Arnold, D.L., Pike, G.B., 2004. Regional variations in normal brain shown by quantitative magnetization transfer imaging. Magn. Reson. Med. 51 (2), 299–303.

26. Levesque, I.R., Pike, G.B., 2009. Characterizing healthy and diseased white matter using quantitative magnetization transfer and multicomponent T2 relaxometry: a unified view via a four-pool model. Magn. Reson. Med. 62 (6), 1487–1496.

27. Dula, A.N., Gochberg, D.F., Valentine, H.L., Valentine, W.M., Does, M.D., 2010. Multiexponential T2, magnetization transfer, and quantitative histology in white matter tracts of rat spinal cord. Magn. Reson. Med. 63 (4), 902–909.

28. Harkins, K.D., Dula, A.N., Does, M.D., 2012. Effect of intercompartmental water exchange on the apparent myelin water fraction in multiexponential T2 measurements of rat spinal cord. Magn. Reson. Med. 67 (3), 793–800.

29. Dortch, R.D., Harkins, K.D., Juttukonda, M.R., Gore, J.C., Does, M.D., 2013a. Characterizing inter-compartmental water exchange in myelinated tissue using relaxation exchange spectroscopy. Magn. Reson. Med. 70 (5), 1450–1459.

30. Labadie, C., Lee, J.-H., Rooney, W.D., Jarchow, S., Aubert-Frecon, M., Springer, C.S., Moeller, H.E., 2014. Myelin water mapping by spatially regularized longitudinal relaxographic imaging at high magnetic fields. Magn. Reson. Med. 71 (1), 375–387.

31. Henkelman R, Huang X, Xiang Q, Stanisz G, Swanson S, Bronskill M. 1993. Quantitative Interpretation of Magnetization-Transfer. Magn. Reson. Med. 29 (6), 759–66.

32. Gochberg, D.F., Gore, J.C., 2003. Quantitative imaging of magnetization transfer using an inversion recovery sequence. Magn. Reson. Med. 49 (3), 501–505.

33. Schmierer, K., Tozer, D., Scaravilli, F., Altmann, D., Barker, G., Tofts, P., Miller, D., 2007. Quantitative magnetization transfer imaging in postmortem multiple sclerosis brain. J. Magn. Reson Imaging 26 (1), 41–51.

34. Janve, V., Zu, Z., Yao, S., Li, K., Zhang, F., Wilson, K., Ou, X., Does, M., Subramaniam, S., Gochberg, D., 2013. The radial diffusivity and magnetization transfer pool size ratio are sensitive markers for demyelination in a rat model of type III multiple sclerosis (MS) lesions. NeuroImage 74, 298–305.

35. Turati, L., Moscatelli, M., Mastropietro, A., Dowell, N., Zucca, I., Erbetta, A., Cordiglieri, C., Brenna, G., Bianchi, B., Mantegazza, R., Cercignani, M., Baggi, F., Minati, L., 2015. In vivo quantitative magnetization transfer imaging correlates with histology during de- and remyelination in cuprizone-treated mice. NMR Biomed. 28 (3), 327–337.

36. Varma, G., Duhamel, G., de Bazelaire, C., Alsop, D., 2015. Magnetization transfer from inhomogeneously broadened lines: a potential marker for myelin. Magn. Reson Med. 73 (2), 614–622.

37. Varma, G., Girard, O., Prevost, V., Grant, A., Duhamel, G., Alsop, D., 2015. Interpretation of magnetization transfer from inhomogeneously broadened lines (ihMT) in tissues as a dipolar order effect within motion restricted molecules. J. Magn. Reson 260, 67–76.

38. Swanson, S., Malyarenko, D., Fabiilli, M., Welsh, R., Nielsen, J., Srinivasan, A., 2017. Molecular, dynamic, and structural origin of inhomogeneous magnetization transfer in lipid membranes. Magn. Reson Med. 77 (3), 1318–1328.

39. Rooney, W.D., Johnson, G., Li, X., Cohen, E.R., Kim, S.-G., Ugurbil, K., Springer, C.S., 2007. Magnetic field and tissue dependencies of human brain longitudinal 1H2O relaxation in vivo. Magn. Reson. Med. 57 (2), 308–318.

40. Callaghan, M.F., Helms, G., Lutti, A., Mohammadi, S., Weiskopf, N., 2015. A general linear relaxometry model of R1 using imaging data. Magn. Reson. Med. 73 (3), 1309–1314.

41. Stüber, C., Morawski, M., Sch€afer, A., Labadie, C., W€ahnert, M., Leuze, C., Streicher, M., Barapatre, N., Reimann, K., Geyer, S., Spemann, D., Turner, R., 2014. Myelin and iron concentration in the human brain: a quantitative study of MRI contrast. Neuroimage 93 (Pt 1), 95–106.

42. Bock, N.A., Hashim, E., Janik, R., Konyer, N.B., Weiss, M., Stanisz, G.J., Turner, R., Geyer, S., 2013. Optimizing T1-weighted imaging of cortical myelin content at 3.0T. Neuroimage 65, 1–12.

43. Bock, N.A., Kocharyan, A., Liu, J.V., Silva, A.C., 2009. Visualizing the entire cortical myelination pattern in marmosets with magnetic resonance imaging. J. Neurosci. Meth. 185 (1), 15–22.

44. Oakden, W., Bock, N.A., Al-Ebraheem, A., Farquharson, M.J., Stanisz, G.J., 2017. Early regional cuprizone-induced demyelination in a rat model revealed with MRI. NMR Biomed. 28 (3), e3743–e3749.

45. Lutti, A., Dick, F., Sereno, M.I., Weiskopf, N., 2014. Using high-resolution quantitative mapping of R1 as an index of cortical myelination. Neuroimage 93 (2), 176–188.

46. Rioux, J.A., Levesque, I.R., Rutt, B.K., 2015. Biexponential longitudinal relaxation in white matter: characterization and impact on T1 mapping with IR-FSE and MP2RAGE. Magn. Reson. Med. 75 (6), 2265–2277.

47. Marques, J.P., Kober, T., Krueger, G., van der Zwaag, W., Van de Moortele, P.-F., Gruetter, R., 2010. MP2RAGE, a self bias-field corrected sequence for improved segmentation and T1-mapping at high field. Neuroimage 49 (2), 1271–1281.

48. Duyn JH. 2018. Studying brain microstructure with magnetic susceptibility contrast at high-field. Neuroimage 182, 152–161.

49. Sled JG. 2018. Modelling and interpretation of magnetization transfer imaging in the brain. Neuroimage 182, 128–135

50. Does MD. 2018. Inferring brain tissue composition and microstructure via MR relaxometry. Neuroimage 182, 136–148

Proc. Intl. Soc. Mag. Reson. Med. 27 (2019)