Assessment of white matter hyperintensity severity using multimodal MRI in Alzheimer’s Disease

White matter hyperintensities (WMHs) are clinically significant MRI abnormalities often detected in the elderly and early stages of Alzheimer’s Disease. They are indicative of vascular pathology but represent a mixture of microstructural tissue alterations that is highly variable between individuals. To better understand these alterations, we leveraged the signal of different MRI contrasts sampled within WMHs, which have differential sensitivity to microstructural properties. Subsequently, we sought to examine the asso of these WMH signal measures to clinically-relevant measures such as cortical and global brain atrophy, cognitive function, diagnostic and demographic differences, and Alzheimer’s Disease-relevant cardiovascular risk factors. Our sample of 118 subjects was composed of healthy controls (n=30), high-risk of Alzheimer’s Disease due to familial history (n=47), mild cognitive impairment (n=32), and clinical Alzheimer’s Disease (n=9) as a means of ascertaining a spectrum of impairment. We sampled the median signal within WMHs on weighted MRI images that are commonly acquired (T1-weighted [T1w], T2-weighted [T2w], T1w/T2w ratio, Fluid-Attenuated Inversion Recovery [FLAIR]), and the relaxation times from quantitative T1 (qT1) and T2* (qT2*) images. Main analyses were performed with a periventricular/deep/superficial white matter parcellation and were repeated with a lobar white matter parcellation. We demonstrated that the correlations between WMH signal measures were variable, suggesting that they are likely influenced by different microstructural properties. We observed that the WMH qT2* and FLAIR measures displayed different age- and disease-related trends compared to normal-appearing white matter, highlighting sensitivity to WMH-specific tissue deterioration. Further, WMH qT2* particularly in periventricular and occipital white matter regions was consistently associated with several of our clinical variables of interest using both parcellation schemes in univariate analyses, and further showed high contributions to a pattern of brain variables that was associated with age and cognitive variables in multivariate Partial Least Squares Correlation analyses. qT1 and FLAIR measures showed consistent clinical relationships in multivariate analyses only, while T1w, T2w, and T1w/T2w ratio measures were not consistently associated with clinical variables. We observed that the qT2* signal was sensitive to clinically-relevant microstructural tissue alterations specific to WMHs. Combining volumetric and signal measures of WMH, particularly qT2* and to a lesser extent qT1 and FLAIR, should be considered to more precisely characterize the severity of WMHs in vivo. These findings may have implications in determining the reversibility of WMHs and potential efficacy of cardio- and cerebrovascular treatments.

[1]  J. Vogelstein,et al.  Suboptimal phenotypic reliability impedes reproducible human neuroscience , 2022, bioRxiv.

[2]  C. Gerloff,et al.  Free-water diffusion MRI detects structural alterations surrounding white matter hyperintensities in the early stage of cerebral small vessel disease , 2022, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.

[3]  V. Hachinski,et al.  White matter hyperintensities and longitudinal cognitive decline in cognitively normal populations and across diagnostic categories: A meta‐analysis, systematic review, and recommendations for future study harmonization , 2022, Alzheimer's & dementia : the journal of the Alzheimer's Association.

[4]  Timothy O. Laumann,et al.  Reproducible brain-wide association studies require thousands of individuals , 2022, Nature.

[5]  Mahanand Belathur Suresh,et al.  The canonical pattern of Alzheimer's disease atrophy is linked to white matter hyperintensities in normal controls, differently in normal controls compared to in AD , 2022, Neurobiology of Aging.

[6]  Christine L. Tardif,et al.  High spatial overlap but diverging age-related trajectories of cortical MRI markers aiming to represent intracortical myelin and microstructure , 2022, bioRxiv.

[7]  D. L. Collins,et al.  White matter hyperintensities are associated with grey matter atrophy and cognitive decline in Alzheimer's disease and frontotemporal dementia , 2021, Neurobiology of Aging.

[8]  S. Duchesne,et al.  White matter hyperintensity distribution differences in aging and neurodegenerative disease cohorts , 2021, NeuroImage: Clinical.

[9]  Sarah N. Kraeutner,et al.  In vivo myelin imaging and tissue microstructure in white matter hyperintensities and perilesional white matter , 2021, medRxiv.

[10]  Chrisopher C. Steele,et al.  Analyses of microstructural variation in the human striatum using non-negative matrix factorization , 2021, NeuroImage.

[11]  I. Maldonado,et al.  Cerebral white matter vasculature: still uncharted? , 2021, Brain : a journal of neurology.

[12]  L. Winchester,et al.  Associations Between Brain Volumes and Cognitive Tests with Hypertensive Burden in UK Biobank , 2021, Journal of Alzheimer's disease : JAD.

[13]  Christine L. Tardif,et al.  Open science datasets from PREVENT-AD, a longitudinal cohort of pre-symptomatic Alzheimer’s disease , 2021, NeuroImage: Clinical.

[14]  Christine L. Tardif,et al.  Hippocampal shape across the healthy lifespan and its relationship with cognition , 2021, Neurobiology of Aging.

[15]  Klaus P. Ebmeier,et al.  White matter hyperintensities classified according to intensity and spatial location reveal specific associations with cognitive performance , 2020, NeuroImage: Clinical.

[16]  Christine L. Tardif,et al.  Hippocampal subfield volumes across the healthy lifespan and the effects of MR sequence on estimates , 2020, NeuroImage.

[17]  B. Mišić,et al.  Molecular signatures of cognition and affect , 2020, bioRxiv.

[18]  Ali R. Khan,et al.  Post-mortem 7 Tesla MRI detection of white matter hyperintensities: A multidisciplinary voxel-wise comparison of imaging and histological correlates , 2020, NeuroImage: Clinical.

[19]  Jongho Lee,et al.  So You Want to Image Myelin Using MRI: An Overview and Practical Guide for Myelin Water Imaging , 2020, Journal of magnetic resonance imaging : JMRI.

[20]  M. Mallar Chakravarty,et al.  Investigating microstructural variation in the human hippocampus using non-negative matrix factorization , 2019, NeuroImage.

[21]  Nadim Joni Shah,et al.  Quantitative MRI of cerebral white matter hyperintensities: A new approach towards understanding the underlying pathology , 2019, NeuroImage.

[22]  Christine L. Tardif,et al.  MR‐based age‐related effects on the striatum, globus pallidus, and thalamus in healthy individuals across the adult lifespan , 2019, Human brain mapping.

[23]  Stefan Ropele,et al.  The role of iron and myelin in orientation dependent R2* of white matter , 2019, NMR in biomedicine.

[24]  Alzheimer's Disease Neuroimaging Initiative,et al.  White matter in different regions evolves differently during progression to dementia , 2019, Neurobiology of Aging.

[25]  D. Louis Collins,et al.  A clinical-anatomical signature of Parkinson's disease identified with partial least squares and magnetic resonance imaging , 2017, NeuroImage.

[26]  Won-Jin Moon,et al.  Myelin loss in white matter hyperintensities and normal-appearing white matter of cognitively impaired patients: a quantitative synthetic magnetic resonance imaging study , 2018, European Radiology.

[27]  A. Hofman,et al.  Cerebral small vessel disease and the risk of dementia: A systematic review and meta-analysis of population-based evidence , 2018, Alzheimer's & Dementia.

[28]  Rikesh M. Rajani,et al.  Reversal of endothelial dysfunction reduces white matter vulnerability in cerebral small vessel disease in rats , 2018, Science Translational Medicine.

[29]  Chu-Chung Huang,et al.  Nonlinear pattern of the emergence of white matter hyperintensity in healthy Han Chinese: an adult lifespan study , 2018, Neurobiology of Aging.

[30]  D. Norris,et al.  Progression of White Matter Hyperintensities Preceded by Heterogeneous Decline of Microstructural Integrity , 2018, Stroke.

[31]  A. Brickman,et al.  The effect of white matter hyperintensities on cognition is mediated by cortical atrophy , 2018, Neurobiology of Aging.

[32]  Mahsa Dadar,et al.  Validation of T1w‐based segmentations of white matter hyperintensity volumes in large‐scale datasets of aging , 2018, Human brain mapping.

[33]  Ludovica Griffanti,et al.  Classification and characterization of periventricular and deep white matter hyperintensities on MRI: A study in older adults , 2017, NeuroImage.

[34]  D. Louis Collins,et al.  Performance comparison of 10 different classification techniques in segmenting white matter hyperintensities in aging , 2017, NeuroImage.

[35]  Alan J. Thomas,et al.  Parietal white matter lesions in Alzheimer’s disease are associated with cortical neurodegenerative pathology, but not with small vessel disease , 2017, Acta Neuropathologica.

[36]  F. de Leeuw,et al.  Disease progression and regression in sporadic small vessel disease-insights from neuroimaging. , 2017, Clinical science.

[37]  C. DeCarli,et al.  Impact of multiple pathologies on the threshold for clinically overt dementia , 2017, Acta Neuropathologica.

[38]  D. Louis Collins,et al.  Validation of a Regression Technique for Segmentation of White Matter Hyperintensities in Alzheimer’s Disease , 2017, IEEE Transactions on Medical Imaging.

[39]  Eleni Sakka,et al.  Integrity of normal-appearing white matter: Influence of age, visible lesion burden and hypertension in patients with small-vessel disease , 2017, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.

[40]  Melissa F. Holmes,et al.  Peripheral inflammatory markers indicate microstructural damage within periventricular white matter hyperintensities in Alzheimer's disease: A preliminary report , 2017, Alzheimer's & dementia.

[41]  T. Hortobágyi,et al.  Vascular cognitive impairment neuropathology guidelines (VCING): the contribution of cerebrovascular pathology to cognitive impairment. , 2016, Brain : a journal of neurology.

[42]  K. Jellinger,et al.  Post-mortem assessment in vascular dementia: advances and aspirations , 2016, BMC Medicine.

[43]  Norbert Schuff,et al.  Early role of vascular dysregulation on late-onset Alzheimer's disease based on multifactorial data-driven analysis , 2016, Nature Communications.

[44]  Nick C Fox,et al.  White matter hyperintensities are a core feature of Alzheimer's disease: Evidence from the dominantly inherited Alzheimer network , 2016, Annals of neurology.

[45]  Christos Davatzikos,et al.  White matter hyperintensities and imaging patterns of brain ageing in the general population. , 2016, Brain : a journal of neurology.

[46]  L. Wolfson,et al.  Cardiovascular risk factors and small vessel disease of the brain: Blood pressure, white matter lesions, and functional decline in older persons , 2016, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.

[47]  J. Poirier,et al.  Rationale and Structure for a New Center for Studies on Prevention of Alzheimer's Disease (StoP-AD). , 2016, The journal of prevention of Alzheimer's disease.

[48]  Christos Davatzikos,et al.  Finding imaging patterns of structural covariance via Non-Negative Matrix Factorization , 2015, NeuroImage.

[49]  Benjamin S. Aribisala,et al.  White matter hyperintensities and normal-appearing white matter integrity in the aging brain , 2015, Neurobiology of Aging.

[50]  A. Brickman,et al.  Reconsidering harbingers of dementia: progression of parietal lobe white matter hyperintensities predicts Alzheimer's disease incidence , 2015, Neurobiology of Aging.

[51]  P. Nederkoorn,et al.  Presence and progression of white matter hyperintensities and cognition , 2014, Neurology.

[52]  Robert Turner,et al.  Myelin and iron concentration in the human brain: A quantitative study of MRI contrast , 2014, NeuroImage.

[53]  Sven Haller,et al.  Do brain T2/FLAIR white matter hyperintensities correspond to myelin loss in normal aging? A radiologic-neuropathologic correlation study , 2013, Acta neuropathologica communications.

[54]  Wiro J. Niessen,et al.  Changes in Normal-Appearing White Matter Precede Development of White Matter Lesions , 2013, Stroke.

[55]  A. McIntosh,et al.  Multivariate statistical analyses for neuroimaging data. , 2013, Annual review of psychology.

[56]  D. Louis Collins,et al.  BEaST: Brain extraction based on nonlocal segmentation technique , 2012, NeuroImage.

[57]  Peter Bühlmann,et al.  MissForest - non-parametric missing value imputation for mixed-type data , 2011, Bioinform..

[58]  D. V. van Essen,et al.  Mapping Human Cortical Areas In Vivo Based on Myelin Content as Revealed by T1- and T2-Weighted MRI , 2011, The Journal of Neuroscience.

[59]  C. DeCarli,et al.  Midlife vascular risk factor exposure accelerates structural brain aging and cognitive decline , 2011, Alzheimer's & Dementia.

[60]  Anthony Randal McIntosh,et al.  Partial Least Squares (PLS) methods for neuroimaging: A tutorial and review , 2011, NeuroImage.

[61]  David G Norris,et al.  Diffusion Tensor Imaging and Gait in Elderly Persons With Cerebral Small Vessel Disease , 2011, Stroke.

[62]  Arno Klein,et al.  A reproducible evaluation of ANTs similarity metric performance in brain image registration , 2011, NeuroImage.

[63]  Nikolaus Weiskopf,et al.  Unified segmentation based correction of R1 brain maps for RF transmit field inhomogeneities (UNICORT) , 2011, NeuroImage.

[64]  U. Klose,et al.  The in vivo influence of white matter fiber orientation towards B0 on T2* in the human brain , 2010, NMR in biomedicine.

[65]  W. M. van der Flier,et al.  Heterogeneity of small vessel disease: a systematic review of MRI and histopathology correlations , 2010, Journal of Neurology, Neurosurgery & Psychiatry.

[66]  H. Markus,et al.  The clinical importance of white matter hyperintensities on brain magnetic resonance imaging: systematic review and meta-analysis , 2010, BMJ : British Medical Journal.

[67]  J. Finsterbusch,et al.  Optimization and Validation of Methods for Mapping of the Radiofrequency Transmit Field at 3T , 2010, Magnetic resonance in medicine.

[68]  Brian B. Avants,et al.  N4ITK: Improved N3 Bias Correction , 2010, IEEE Transactions on Medical Imaging.

[69]  Sean C L Deoni,et al.  Quantitative Relaxometry of the Brain , 2010, Topics in magnetic resonance imaging : TMRI.

[70]  Tobias Kober,et al.  MP2RAGE, a self bias-field corrected sequence for improved segmentation and T1-mapping at high field , 2010, NeuroImage.

[71]  Pierrick Coupé,et al.  Author manuscript, published in "Journal of Magnetic Resonance Imaging 2010;31(1):192-203" DOI: 10.1002/jmri.22003 Adaptive Non-Local Means Denoising of MR Images with Spatially Varying Noise Levels , 2010 .

[72]  D. Loewenstein,et al.  Association of White Matter Hyperintensity Measurements on Brain MR Imaging with Cognitive Status, Medial Temporal Atrophy, and Cardiovascular Risk Factors , 2009, American Journal of Neuroradiology.

[73]  C Caltagirone,et al.  Characterization of white matter fiber bundles with T2* relaxometry and DTI , 2009, NeuroImage.

[74]  Alan C. Evans,et al.  Depth potential function for folding pattern representation, registration and analysis , 2009, Medical Image Anal..

[75]  B. Todorich,et al.  Oligodendrocytes and myelination: The role of iron , 2009, Glia.

[76]  W. M. van der Flier,et al.  Heterogeneity of White Matter Hyperintensities in Alzheimer's Disease: Post-Mortem Quantitative MRI and Neuropathology , 2008, Brain : a journal of neurology.

[77]  J. MacFall,et al.  Classification of White Matter Lesions on Magnetic Resonance Imaging in Elderly Persons , 2008, Biological Psychiatry.

[78]  J. O'Brien,et al.  White matter lesions in an unselected cohort of the elderly: astrocytic, microglial and oligodendrocyte precursor cell responses , 2007, Neuropathology and applied neurobiology.

[79]  Alan C. Evans,et al.  Automated 3-D extraction and evaluation of the inner and outer cortical surfaces using a Laplacian map and partial volume effect classification , 2005, NeuroImage.

[80]  J. Cummings,et al.  The Montreal Cognitive Assessment, MoCA: A Brief Screening Tool For Mild Cognitive Impairment , 2005, Journal of the American Geriatrics Society.

[81]  D. Harvey,et al.  Anatomical Mapping of White Matter Hyperintensities (WMH): Exploring the Relationships Between Periventricular WMH, Deep WMH, and Total WMH Burden , 2005, Stroke.

[82]  Alan C. Evans,et al.  Cortical thickness analysis examined through power analysis and a population simulation , 2005, NeuroImage.

[83]  Alexander Hammers,et al.  Three‐dimensional maximum probability atlas of the human brain, with particular reference to the temporal lobe , 2003, Human brain mapping.

[84]  Thomas E. Nichols,et al.  Thresholding of Statistical Maps in Functional Neuroimaging Using the False Discovery Rate , 2002, NeuroImage.

[85]  C. Randolph,et al.  The Repeatable Battery for the Assessment of Neuropsychological Status (RBANS): preliminary clinical validity. , 1998, Journal of clinical and experimental neuropsychology.

[86]  Alan C. Evans,et al.  A nonparametric method for automatic correction of intensity nonuniformity in MRI data , 1998, IEEE Transactions on Medical Imaging.

[87]  R. Weinshilboum,et al.  Human catechol-O-methyltransferase pharmacogenetics: description of a functional polymorphism and its potential application to neuropsychiatric disorders. , 1996, Pharmacogenetics.

[88]  M. A. Bell,et al.  Features of the cerebral vascular pattern that predict vulnerability to perfusion or oxygenation deficiency: an anatomic study. , 1990, AJNR. American journal of neuroradiology.

[89]  Y Horikawa,et al.  Significance of proton relaxation time measurement in brain edema, cerebral infarction and brain tumors. , 1986, Magnetic resonance imaging.