Circulating Metabolome and White Matter Hyperintensities in Women and Men

Background: White matter hyperintensities (WMH), identified on T2-weighted magnetic resonance images of the human brain as areas of enhanced brightness, are a major risk factor of stroke, dementia, and death. There are no large-scale studies testing associations between WMH and circulating metabolites. Methods: We studied up to 9290 individuals (50.7% female, average age 61 years) from 15 populations of 8 community-based cohorts. WMH volume was quantified from T2-weighted or fluid-attenuated inversion recovery images or as hypointensities on T1-weighted images. Circulating metabolomic measures were assessed with mass spectrometry and nuclear magnetic resonance spectroscopy. Associations between WMH and metabolomic measures were tested by fitting linear regression models in the pooled sample and in sex-stratified and statin treatment–stratified subsamples. Our basic models were adjusted for age, sex, age×sex, and technical covariates, and our fully adjusted models were also adjusted for statin treatment, hypertension, type 2 diabetes, smoking, body mass index, and estimated glomerular filtration rate. Population-specific results were meta-analyzed using the fixed-effect inverse variance–weighted method. Associations with false discovery rate (FDR)–adjusted P values (PFDR)<0.05 were considered significant. Results: In the meta-analysis of results from the basic models, we identified 30 metabolomic measures associated with WMH (PFDR<0.05), 7 of which remained significant in the fully adjusted models. The most significant association was with higher level of hydroxyphenylpyruvate in men (PFDR.full.adj=1.40×10−7) and in both the pooled sample (PFDR.full.adj=1.66×10−4) and statin-untreated (PFDR.full.adj=1.65×10−6) subsample. In men, hydroxyphenylpyruvate explained 3% to 14% of variance in WMH. In men and the pooled sample, WMH were also associated with lower levels of lysophosphatidylcholines and hydroxysphingomyelins and a larger diameter of low-density lipoprotein particles, likely arising from higher triglyceride to total lipids and lower cholesteryl ester to total lipids ratios within these particles. In women, the only significant association was with higher level of glucuronate (PFDR=0.047). Conclusions: Circulating metabolomic measures, including multiple lipid measures (eg, lysophosphatidylcholines, hydroxysphingomyelins, low-density lipoprotein size and composition) and nonlipid metabolites (eg, hydroxyphenylpyruvate, glucuronate), associate with WMH in a general population of middle-aged and older adults. Some metabolomic measures show marked sex specificities and explain a sizable proportion of WMH variance.

[1]  J. Wardlaw,et al.  Sex Differences in Cerebral Small Vessel Disease: A Systematic Review and Meta-Analysis , 2021, Frontiers in Neurology.

[2]  F. Vaz,et al.  Sex differences in the association of sphingolipids with age in Dutch and South-Asian Surinamese living in Amsterdam, the Netherlands , 2021, Biology of sex differences.

[3]  N. Eriksson,et al.  Cerebral small vessel disease genomics and its implications across the lifespan , 2020, Nature Communications.

[4]  M. Woodward,et al.  Sex differences in the association between major risk factors and the risk of stroke in the UK Biobank cohort study , 2020, Neurology.

[5]  Sophie V. Eastwood,et al.  Cohort Profile Update: Southall and Brent Revisited (SABRE) study: a UK population-based comparison of cardiovascular disease and diabetes in people of European, South Asian and African Caribbean heritage , 2020, International journal of epidemiology.

[6]  W. M. van der Flier,et al.  Circulating metabolites are associated with brain atrophy and white matter hyperintensities , 2020, Alzheimer's & dementia : the journal of the Alzheimer's Association.

[7]  T. Paus,et al.  Thickness of the cerebral cortex shows positive association with blood levels of triacylglycerols carrying 18-carbon fatty acids , 2020, Communications Biology.

[8]  A. Hainsworth,et al.  Association of White Matter Hyperintensities and Cardiovascular Disease , 2020, Circulation. Cardiovascular imaging.

[9]  Stuart J. Ritchie,et al.  Plasma lipid and liporotein biomarkers in LBC1936: Do they predict general cognitive ability and brain structure? , 2020, bioRxiv.

[10]  Brenda C T Kieboom,et al.  Objectives, design and main findings until 2020 from the Rotterdam Study , 2020, European Journal of Epidemiology.

[11]  J. Wardlaw,et al.  Protocol: The Lacunar Intervention Trial 2 (LACI-2). A trial of two repurposed licenced drugs to prevent progression of cerebral small vessel disease , 2020, European stroke journal.

[12]  J. Broderick,et al.  Temporal Trends in Stroke Incidence Over Time by Sex and Age in the GCNKSS , 2020, Stroke.

[13]  T. Paus,et al.  A variant near DHCR24 associates with microstructural properties of white matter and peripheral lipid metabolism in adolescents , 2020, Molecular Psychiatry.

[14]  C. Jack,et al.  Cardiometabolic Health and Longitudinal Progression of White Matter Hyperintensity: The Mayo Clinic Study of Aging. , 2019, Stroke.

[15]  J. Manson,et al.  Sex Differences in Hypertension and Stroke Risk in the REGARDS Study. , 2019, Hypertension.

[16]  M. Filippi,et al.  Progression of brain white matter hyperintensities in asymptomatic patients with carotid atherosclerotic plaques and no indication for revascularization. , 2019, Atherosclerosis.

[17]  M. Dichgans,et al.  Small vessel disease: mechanisms and clinical implications , 2019, The Lancet Neurology.

[18]  Alonso Barrantes-Freer,et al.  Myelin in the Central Nervous System: Structure, Function, and Pathology. , 2019, Physiological reviews.

[19]  C. Jack,et al.  Plasma Metabolites Associated with Brain MRI Measures of Neurodegeneration in Older Adults in the Atherosclerosis Risk in Communities–Neurocognitive Study (ARIC-NCS) , 2019, International journal of molecular sciences.

[20]  C. Moy,et al.  Sex and Race Differences in the Association of Incident Ischemic Stroke With Risk Factors , 2019, JAMA neurology.

[21]  S. Larsson,et al.  Clinical Significance of Magnetic Resonance Imaging Markers of Vascular Brain Injury: A Systematic Review and Meta-analysis , 2019, JAMA neurology.

[22]  N. Stachenfeld,et al.  Sex differences in endothelial function important to vascular health and overall cardiovascular disease risk across the lifespan. , 2018, American journal of physiology. Heart and circulatory physiology.

[23]  W. Niessen,et al.  Practical Small Vessel Disease Score Relates to Stroke, Dementia, and Death: The Rotterdam Study , 2018, Stroke.

[24]  M. Jorge Cardoso,et al.  Cardiovascular Risk Factors and White Matter Hyperintensities: Difference in Susceptibility in South Asians Compared With Europeans , 2018, Journal of the American Heart Association.

[25]  M. Weiner,et al.  Serum triglycerides in Alzheimer’s disease: Relation to neuroimaging and CSF biomarkers , 2018, bioRxiv.

[26]  Manfred Spraul,et al.  Quantitative Lipoprotein Subclass and Low Molecular Weight Metabolite Analysis in Human Serum and Plasma by 1H NMR Spectroscopy in a Multilaboratory Trial. , 2018, Analytical chemistry.

[27]  T. Paus,et al.  Visceral fat-related systemic inflammation and the adolescent brain: a mediating role of circulating glycerophosphocholines , 2018, International Journal of Obesity.

[28]  V. Salomaa,et al.  Circulating metabolites and general cognitive ability and dementia: Evidence from 11 cohort studies , 2018, Alzheimer's & Dementia.

[29]  I. Deary,et al.  Cohort Profile Update: The Lothian Birth Cohorts of 1921 and 1936 , 2018, International journal of epidemiology.

[30]  A. Saykin,et al.  Brain and blood metabolite signatures of pathology and progression in Alzheimer disease: A targeted metabolomics study , 2018, PLoS medicine.

[31]  David S. Wishart,et al.  HMDB 4.0: the human metabolome database for 2018 , 2017, Nucleic Acids Res..

[32]  O. Franco,et al.  The Rotterdam Study: 2018 update on objectives, design and main results , 2017, European Journal of Epidemiology.

[33]  Kenneth J. Smith,et al.  Understanding a role for hypoxia in lesion formation and location in the deep and periventricular white matter in small vessel disease and multiple sclerosis. , 2017, Clinical science.

[34]  P. Subbaiah,et al.  Dietary docosahexaenoic acid (DHA) as lysophosphatidylcholine, but not as free acid, enriches brain DHA and improves memory in adult mice , 2017, Scientific Reports.

[35]  J. Wardlaw,et al.  Blood-brain barrier failure as a core mechanism in cerebral small vessel disease and dementia: evidence from a cohort study , 2017, Alzheimer's & Dementia.

[36]  Debbie A Lawlor,et al.  Quantitative Serum Nuclear Magnetic Resonance Metabolomics in Large-Scale Epidemiology: A Primer on -Omic Technologies , 2017, American journal of epidemiology.

[37]  Nick C Fox,et al.  Study protocol: Insight 46 – a neuroscience sub-study of the MRC National Survey of Health and Development , 2017, BMC Neurology.

[38]  S. Weis,et al.  Sex‐dependent consequences of neonatal brain hypoxia‐ischemia in the rat , 2017, Journal of neuroscience research.

[39]  G. Fiskum,et al.  Sex‐dependent mitochondrial respiratory impairment and oxidative stress in a rat model of neonatal hypoxic‐ischemic encephalopathy , 2016, Journal of neurochemistry.

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

[41]  Louis Richer,et al.  Cohort Profile: The Saguenay Youth Study (SYS). , 2016, International journal of epidemiology.

[42]  Ramachandran S Vasan,et al.  Cohort Profile: The Framingham Heart Study (FHS): overview of milestones in cardiovascular epidemiology. , 2015, International journal of epidemiology.

[43]  L. Ermini,et al.  Plasma non-esterified docosahexaenoic acid is the major pool supplying the brain , 2015, Scientific Reports.

[44]  Tomás Paus,et al.  Axon diameter and axonal transport: In vivo and in vitro effects of androgens , 2015, NeuroImage.

[45]  Sébastien Ourselin,et al.  Bayesian Model Selection for Pathological Neuroimaging Data Applied to White Matter Lesion Segmentation , 2015, IEEE Transactions on Medical Imaging.

[46]  Tom R. Gaunt,et al.  Metabolite profiling and cardiovascular event risk: a prospective study of 3 population-based cohorts. , 2015, Circulation.

[47]  C. Jack,et al.  Smoking and white matter hyperintensity progression , 2015, Neurology.

[48]  Anders Larsson,et al.  Large-scale Metabolomic Profiling Identifies Novel Biomarkers for Incident Coronary Heart Disease , 2014, PLoS genetics.

[49]  Richard P. Bazinet,et al.  Polyunsaturated fatty acids and their metabolites in brain function and disease , 2014, Nature Reviews Neuroscience.

[50]  J. Magdalou,et al.  The UDP-glucuronosyltransferases of the blood-brain barrier: their role in drug metabolism and detoxication , 2014, Front. Cell. Neurosci..

[51]  M. Spraul,et al.  Precision high-throughput proton NMR spectroscopy of human urine, serum, and plasma for large-scale metabolic phenotyping. , 2014, Analytical chemistry.

[52]  G. Marathe,et al.  To hydrolyze or not to hydrolyze: the dilemma of platelet-activating factor acetylhydrolase , 2014, Journal of Lipid Research.

[53]  D. Weaver Alzheimer's , 2014, Neurology.

[54]  M. Woodward,et al.  Diabetes as a risk factor for stroke in women compared with men: a systematic review and meta-analysis of 64 cohorts, including 775 385 individuals and 12 539 strokes , 2014, The Lancet.

[55]  I. Deary,et al.  Vascular risk factors, large-artery atheroma, and brain white matter hyperintensities , 2014, Neurology.

[56]  Derick R. Peterson,et al.  Plasma phospholipids identify antecedent memory impairment in older adults , 2014, Nature Medicine.

[57]  Henning Hermjakob,et al.  The Reactome pathway knowledgebase , 2013, Nucleic Acids Res..

[58]  Nick C Fox,et al.  Neuroimaging standards for research into small vessel disease and its contribution to ageing and neurodegeneration , 2013, The Lancet Neurology.

[59]  Reino Laatikainen,et al.  High-throughput quantification of circulating metabolites improves prediction of subclinical atherosclerosis. , 2012, European heart journal.

[60]  Jesper Fredriksson,et al.  Brain tissue volumes in the general population of the elderly The AGES-Reykjavik Study , 2012, NeuroImage.

[61]  Nish Chaturvedi,et al.  Southall And Brent REvisited: Cohort profile of SABRE, a UK population-based comparison of cardiovascular disease and diabetes in people of European, Indian Asian and African Caribbean origins. , 2012, International journal of epidemiology.

[62]  K. Suhre,et al.  Procedure for tissue sample preparation and metabolite extraction for high-throughput targeted metabolomics , 2012, Metabolomics.

[63]  I. Deary,et al.  Brain Aging, Cognition in Youth and Old Age and Vascular Disease in the Lothian Birth Cohort 1936: Rationale, Design and Methodology of the Imaging Protocol* , 2011, International journal of stroke : official journal of the International Stroke Society.

[64]  W. Rathmann,et al.  Cohort profile: the study of health in Pomerania. , 2011, International journal of epidemiology.

[65]  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.

[66]  V. Gudnason,et al.  Coronary Artery Calcium, Brain Function and Structure: The AGES-Reykjavik Study , 2010, Stroke.

[67]  C. Lyketsos,et al.  Serum sphingomyelins and ceramides are early predictors of memory impairment , 2010, Neurobiology of Aging.

[68]  R. Skoff,et al.  Sexual dimorphism in the white matter of rodents , 2009, Journal of the Neurological Sciences.

[69]  Wiro J. Niessen,et al.  White matter lesion extension to automatic brain tissue segmentation on MRI , 2009, NeuroImage.

[70]  D. Hartmann,et al.  Absence of 2-Hydroxylated Sphingolipids Is Compatible with Normal Neural Development But Causes Late-Onset Axon and Myelin Sheath Degeneration , 2008, The Journal of Neuroscience.

[71]  P. Visscher,et al.  The Lothian Birth Cohort 1936: a study to examine influences on cognitive ageing from age 11 to age 70 and beyond , 2007, BMC geriatrics.

[72]  A. Hofman,et al.  The Rotterdam Study: objectives and design update , 2007, European Journal of Epidemiology.

[73]  V. Gudnason,et al.  Age, Gene/Environment Susceptibility-Reykjavik Study: multidisciplinary applied phenomics. , 2007, American journal of epidemiology.

[74]  I. Mikhailenko,et al.  Beyond endocytosis: LRP function in cell migration, proliferation and vascular permeability , 2005, Journal of thrombosis and haemostasis : JTH.

[75]  D. Harvey,et al.  Measures of brain morphology and infarction in the framingham heart study: establishing what is normal , 2005, Neurobiology of Aging.

[76]  M. van Buchem,et al.  Different progression rates for deep white matter hyperintensities in elderly men and women , 2004, Neurology.

[77]  Charles DeCarli,et al.  Stroke Risk Profile Predicts White Matter Hyperintensity Volume: The Framingham Study , 2004, Stroke.

[78]  H. Suh,et al.  Therapeutic effects of lysophosphatidylcholine in experimental sepsis , 2004, Nature Medicine.

[79]  S. Ousman,et al.  Lysophosphatidylcholine induces rapid recruitment and activation of macrophages in the adult mouse spinal cord , 2000, Glia.

[80]  J. Heeren,et al.  Lipoprotein lipase (EC 3.1.1.34) targeting of lipoproteins to receptors , 1997, Proceedings of the Nutrition Society.

[81]  H. S. Mason,et al.  Metabolic hypoxia: accumulation of tyrosine metabolites in hepatocytes at low pO2. , 1978, Biochemical and biophysical research communications.

[82]  T. Dawber,et al.  Epidemiological approaches to heart disease: the Framingham Study. , 1951, American journal of public health and the nation's health.

[83]  P. Carmeliet,et al.  Endothelial Cell Metabolism. , 2018, Physiological reviews.

[84]  Self-Concept Variables Sex Differences in , 2016 .

[85]  R Core Team,et al.  R: A language and environment for statistical computing. , 2014 .

[86]  V. Aidinis,et al.  Lysoglycerophospholipids in chronic inflammatory disorders: the PLA(2)/LPC and ATX/LPA axes. , 2013, Biochimica et biophysica acta.

[87]  U. John,et al.  Study of Health in Pomerania (SHIP): A health examination survey in an east German region: Objectives and design , 2005, Sozial- und Präventivmedizin.

[88]  Y. Benjamini,et al.  Controlling the false discovery rate: a practical and powerful approach to multiple testing , 1995 .

[89]  Stuart J. Ritchie,et al.  Associations between vascular risk factors and brain MRI indices in UK Biobank , 2019, bioRxiv.