436 results on '"Schott, JM"'
Search Results
102. Clinical reporting following the quantification of cerebrospinal fluid biomarkers in Alzheimer's disease: An international overview.
- Author
-
Delaby C, Teunissen CE, Blennow K, Alcolea D, Arisi I, Amar EB, Beaume A, Bedel A, Bellomo G, Bigot-Corbel E, Bjerke M, Blanc-Quintin MC, Boada M, Bousiges O, Chapman MD, DeMarco ML, D'Onofrio M, Dumurgier J, Dufour-Rainfray D, Engelborghs S, Esselmann H, Fogli A, Gabelle A, Galloni E, Gondolf C, Grandhomme F, Grau-Rivera O, Hart M, Ikeuchi T, Jeromin A, Kasuga K, Keshavan A, Khalil M, Körtvelyessy P, Kulczynska-Przybik A, Laplanche JL, Lewczuk P, Li QX, Lleó A, Malaplate C, Marquié M, Masters CL, Mroczko B, Nogueira L, Orellana A, Otto M, Oudart JB, Paquet C, Paoletti FP, Parnetti L, Perret-Liaudet A, Peoc'h K, Poesen K, Puig-Pijoan A, Quadrio I, Quillard-Muraine M, Rucheton B, Schraen S, Schott JM, Shaw LM, Suárez-Calvet M, Tsolaki M, Tumani H, Udeh-Momoh CT, Vaudran L, Verbeek MM, Verde F, Vermunt L, Vogelgsang J, Wiltfang J, Zetterberg H, and Lehmann S
- Subjects
- Humans, Biomarkers cerebrospinal fluid, Amyloid beta-Peptides cerebrospinal fluid, tau Proteins cerebrospinal fluid, Peptide Fragments cerebrospinal fluid, Alzheimer Disease diagnosis, Alzheimer Disease cerebrospinal fluid
- Abstract
Introduction: The current practice of quantifying cerebrospinal fluid (CSF) biomarkers as an aid in the diagnosis of Alzheimer's disease (AD) varies from center to center. For a same biochemical profile, interpretation and reporting of results may differ, which can lead to misunderstandings and raises questions about the commutability of tests., Methods: We obtained a description of (pre-)analytical protocols and sample reports from 40 centers worldwide. A consensus approach allowed us to propose harmonized comments corresponding to the different CSF biomarker profiles observed in patients., Results: The (pre-)analytical procedures were similar between centers. There was considerable heterogeneity in cutoff definitions and report comments. We therefore identified and selected by consensus the most accurate and informative comments regarding the interpretation of CSF biomarkers in the context of AD diagnosis., Discussion: This is the first time that harmonized reports are proposed across worldwide specialized laboratories involved in the biochemical diagnosis of AD., (© 2021 The Authors. Alzheimer's & Dementia published by Wiley Periodicals LLC on behalf of Alzheimer's Association.)
- Published
- 2022
- Full Text
- View/download PDF
103. Corrigendum to "Use of the tau protein-to-peptide ratio in CSF to improve diagnostic classification of Alzheimer's disease" [Clin. Mass Spectrom. 14 (Part B) (2019) 74-82].
- Author
-
Hansson K, Dahlén R, Hansson O, Pernevik E, Paterson R, Schott JM, Magdalinou N, Zetterberg H, Blennow K, and Gobom J
- Abstract
[This corrects the article DOI: 10.1016/j.clinms.2019.07.002.]., (© 2022 THE AUTHORS. Publishing services by ELSEVIER B.V. on behalf of MSACL.)
- Published
- 2022
- Full Text
- View/download PDF
104. Factors influencing resilience to postoperative delirium in adults undergoing elective orthopaedic surgery.
- Author
-
Bowman EML, Cardwell C, McAuley DF, McGuinness B, Passmore AP, Beverland D, Zetterberg H, Schott JM, and Cunningham EL
- Subjects
- Adult, Elective Surgical Procedures, Humans, Postoperative Complications, Risk Factors, Delirium etiology, Orthopedic Procedures adverse effects
- Published
- 2022
- Full Text
- View/download PDF
105. Association between carotid atherosclerosis and brain activation patterns during the Stroop task in older adults: An fNIRS investigation.
- Author
-
Mason SA, Al Saikhan L, Jones S, James SN, Murray-Smith H, Rapala A, Williams S, Sudre C, Wong B, Richards M, Fox NC, Hardy R, Schott JM, Chaturvedi N, and Hughes AD
- Subjects
- Aged, Brain physiology, Hemoglobins analysis, Humans, Prefrontal Cortex diagnostic imaging, Prefrontal Cortex physiology, Spectroscopy, Near-Infrared methods, Stroop Test, Atherosclerosis, Carotid Artery Diseases diagnostic imaging, Dementia
- Abstract
There is an increasing body of evidence suggesting that vascular disease could contribute to cognitive decline and overt dementia. Of particular interest is atherosclerosis, as it is not only associated with dementia, but could be a potential mechanism through which cardiovascular disease directly impacts brain health. In this work, we evaluated the differences in functional near infrared spectroscopy (fNIRS)-based measures of brain activation, task performance, and the change in central hemodynamics (mean arterial pressure (MAP) and heart rate (HR)) during a Stroop color-word task in individuals with atherosclerosis, defined as bilateral carotid plaques (n = 33) and healthy age-matched controls (n = 33). In the healthy control group, the left prefrontal cortex (LPFC) was the only region showing evidence of activation when comparing the incongruous with the nominal Stroop test. A smaller extent of brain activation was observed in the Plaque group compared with the healthy controls (1) globally, as measured by oxygenated hemoglobin (p = 0.036) and (2) in the LPFC (p = 0.02) and left sensorimotor cortices (LMC)(p = 0.008) as measured by deoxygenated hemoglobin. There were no significant differences in HR, MAP, or task performance (both in terms of the time required to complete the task and number of errors made) between Plaque and control groups. These results suggest that carotid atherosclerosis is associated with altered functional brain activation patterns despite no evidence of impaired performance of the Stroop task or central hemodynamic changes., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
106. Nomograms of human hippocampal volume shifted by polygenic scores.
- Author
-
Janahi M, Aksman L, Schott JM, Mokrab Y, and Altmann A
- Subjects
- Hippocampus diagnostic imaging, Humans, Magnetic Resonance Imaging, Neuroimaging, Nomograms, Alzheimer Disease diagnostic imaging, Alzheimer Disease genetics, Cognitive Dysfunction diagnostic imaging, Cognitive Dysfunction genetics
- Abstract
Nomograms are important clinical tools applied widely in both developing and aging populations. They are generally constructed as normative models identifying cases as outliers to a distribution of healthy controls. Currently used normative models do not account for genetic heterogeneity. Hippocampal volume (HV) is a key endophenotype for many brain disorders. Here, we examine the impact of genetic adjustment on HV nomograms and the translational ability to detect dementia patients. Using imaging data from 35,686 healthy subjects aged 44-82 from the UK Biobank (UKB), we built HV nomograms using Gaussian process regression (GPR), which - compared to a previous method - extended the application age by 20 years, including dementia critical age ranges. Using HV polygenic scores (HV-PGS), we built genetically adjusted nomograms from participants stratified into the top and bottom 30% of HV-PGS. This shifted the nomograms in the expected directions by ~100 mm
3 (2.3% of the average HV), which equates to 3 years of normal aging for a person aged ~65. Clinical impact of genetically adjusted nomograms was investigated by comparing 818 subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database diagnosed as either cognitively normal (CN), having mild cognitive impairment (MCI) or Alzheimer's disease (AD) patients. While no significant change in the survival analysis was found for MCI-to-AD conversion, an average of 68% relative decrease was found in intra-diagnostic-group variance, highlighting the importance of genetic adjustment in untangling phenotypic heterogeneity., Competing Interests: MJ, LA, JS, YM, AA No competing interests declared, (© 2022, Janahi et al.)- Published
- 2022
- Full Text
- View/download PDF
107. CSF biomarkers for dementia.
- Author
-
Keshavan A, O'Shea F, Chapman MD, Hart MS, Lunn MP, Paterson RW, Rohrer JD, Mummery CJ, Fox NC, Zetterberg H, and Schott JM
- Subjects
- Aged, Biomarkers cerebrospinal fluid, Humans, Alzheimer Disease diagnosis, Amyloid beta-Peptides
- Abstract
Although cerebrospinal fluid (CSF) biomarker testing is incorporated into some current guidelines for the diagnosis of dementia (such as England's National Institute for Health and Care Excellence (NICE)), it is not widely accessible for most patients for whom biomarkers could potentially change management. Here we share our experience of running a clinical cognitive CSF service and discuss recent developments in laboratory testing including the use of the CSF amyloid-β 42/40 ratio and automated assay platforms. We highlight the importance of collaborative working between clinicians and laboratory staff, of preanalytical sample handling, and discuss the various factors influencing interpretation of the results in appropriate clinical contexts. We advocate for broadening access to CSF biomarkers by sharing clinical expertise, protocols and interpretation with colleagues working in psychiatry and elderly care, especially when access to CSF may be part of a pathway to disease-modifying treatments for Alzheimer's disease and other forms of dementia., Competing Interests: Competing interests: RWP is codirector of the NfL consortium (an industry-funded research consortium) and has given educational lectures sponsored by GE Healthcare. JDR has served on a medical advisory board and had a consultancy agreement with Alector, Arkuda Therapeutics, Wave Life Sciences and Prevail Therapeutics, and had a consultancy agreement also with UCB, AC Immune, Astex Pharmaceuticals, Biogen, Takeda and Eisai. CJM has been an advisor to IONIS, Biogen, Lilly and Roche, is on the international steering committee for aducanumab, and has lectured for Biogen. NCF’s research group has received payment for consultancy or for conducting studies from Biogen, Eli Lilly Research Laboratories, Ionis and Roche. NCF received no personal compensation for the aforementioned activities. NCF also serves on a Data Safety Monitoring Board for Biogen. JMS has received research funding from Avid Radiopharmaceuticals (a wholly owned subsidiary of Eli Lilly), has consulted for Roche Pharmaceuticals, Biogen, Merck, and Eli Lilly, given educational lectures sponsored by GE Healthcare, Eli Lilly, and Biogen, and serves on a data safety monitoring committee for Axon Neuroscience SE. HZ has served at scientific advisory boards and/or as a consultant for Abbvie, Alector, Annexon, Artery Therapeutics, AZTherapies, CogRx, Denali, Eisai, Nervgen, Pinteon Therapeutics, Red Abbey Labs, Passage Bio, Roche, Samumed, Siemens Healthineers, Triplet Therapeutics, and Wave; and has given lectures in symposia sponsored by Cellectricon, Fujirebio, Alzecure, Biogen and Roche. HZ is a cofounder of Brain Biomarker Solutions in Gothenburg AB, which is a part of the GU Ventures Incubator Programme., (© Author(s) (or their employer(s)) 2022. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
108. Associations of β-Amyloid and Vascular Burden With Rates of Neurodegeneration in Cognitively Normal Members of the 1946 British Birth Cohort.
- Author
-
Keuss SE, Coath W, Nicholas JM, Poole T, Barnes J, Cash DM, Lane CA, Parker TD, Keshavan A, Buchanan SM, Wagen AZ, Storey M, Harris M, Malone IB, Sudre CH, Lu K, James SN, Street R, Thomas DL, Dickson JC, Murray-Smith H, Wong A, Freiberger T, Crutch S, Richards M, Fox NC, and Schott JM
- Subjects
- Aged, Amyloid beta-Peptides metabolism, Apolipoprotein E4 genetics, Atrophy pathology, Birth Cohort, Brain pathology, Female, Humans, Magnetic Resonance Imaging, Male, Positron-Emission Tomography, Alzheimer Disease pathology, Cerebrovascular Disorders pathology
- Abstract
Background and Objectives: The goals of this work were to quantify the independent and interactive associations of β-amyloid (Aβ) and white matter hyperintensity volume (WMHV), a marker of presumed cerebrovascular disease (CVD), with rates of neurodegeneration and to examine the contributions of APOE ε4 and vascular risk measured at different stages of adulthood in cognitively normal members of the 1946 British Birth Cohort., Methods: Participants underwent brain MRI and florbetapir-Aβ PET as part of Insight 46, an observational population-based study. Changes in whole-brain, ventricular, and hippocampal volume were directly measured from baseline and repeat volumetric T1 MRI with the boundary shift integral. Linear regression was used to test associations with baseline Aβ deposition, baseline WMHV, APOE ε4, and office-based Framingham Heart Study Cardiovascular Risk Score (FHS-CVS) and systolic blood pressure (BP) at ages 36, 53, and 69 years., Results: Three hundred forty-six cognitively normal participants (mean [SD] age at baseline scan 70.5 [0.6] years; 48% female) had high-quality T1 MRI data from both time points (mean [SD] scan interval 2.4 [0.2] years). Being Aβ positive at baseline was associated with 0.87-mL/y faster whole-brain atrophy (95% CI 0.03, 1.72), 0.39-mL/y greater ventricular expansion (95% CI 0.16, 0.64), and 0.016-mL/y faster hippocampal atrophy (95% CI 0.004, 0.027), while each 10-mL additional WMHV at baseline was associated with 1.07-mL/y faster whole-brain atrophy (95% CI 0.47, 1.67), 0.31-mL/y greater ventricular expansion (95% CI 0.13, 0.60), and 0.014-mL/y faster hippocampal atrophy (95% CI 0.006, 0.022). These contributions were independent, and there was no evidence that Aβ and WMHV interacted in their effects. There were no independent associations of APOE ε4 with rates of neurodegeneration after adjustment for Aβ status and WMHV, no clear relationships between FHS-CVS or systolic BP and rates of neurodegeneration when assessed across the whole sample, and no evidence that FHS-CVS or systolic BP acted synergistically with Aβ., Discussion: Aβ and presumed CVD have distinct and additive effects on rates of neurodegeneration in cognitively normal elderly. These findings have implications for the use of MRI measures as biomarkers of neurodegeneration and emphasize the importance of risk management and early intervention targeting both pathways., (Copyright © 2022 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology.)
- Published
- 2022
- Full Text
- View/download PDF
109. The Influence of Orthopedic Surgery on Circulating Metabolite Levels, and their Associations with the Incidence of Postoperative Delirium.
- Author
-
Jung M, Pan X, Cunningham EL, Passmore AP, McGuinness B, McAuley DF, Beverland D, O'Brien S, Mawhinney T, Schott JM, Zetterberg H, and Green BD
- Abstract
The mechanisms underlying the occurrence of postoperative delirium development are unclear and measurement of plasma metabolites may improve understanding of its causes. Participants ( n = 54) matched for age and gender were sampled from an observational cohort study investigating postoperative delirium. Participants were ≥65 years without a diagnosis of dementia and presented for primary elective hip or knee arthroplasty. Plasma samples collected pre- and postoperatively were grouped as either control ( n = 26, aged: 75.8 ± 5.2) or delirium ( n = 28, aged: 76.2 ± 5.7). Widespread changes in plasma metabolite levels occurred following surgery. The only metabolites significantly differing between corresponding control and delirium samples were ornithine and spermine. In delirium cases, ornithine was 17.6% higher preoperatively, and spermine was 12.0% higher postoperatively. Changes were not associated with various perioperative factors. In binary logistic regression modeling, these two metabolites did not confer a significantly increased risk of delirium. These findings support the hypothesis that disturbed polyamine metabolism is an underlying factor in delirium that warrants further investigation.
- Published
- 2022
- Full Text
- View/download PDF
110. Familial British dementia: a clinical and multi-modal imaging case study.
- Author
-
Harris MJ, Lane CA, Coath W, Malone IB, Cash DM, Barnes J, Barkhof F, and Schott JM
- Subjects
- Humans, Multimodal Imaging methods, Cerebral Amyloid Angiopathy, Dementia diagnostic imaging, Dementia genetics
- Published
- 2022
- Full Text
- View/download PDF
111. Development of a sensitive trial-ready poly(GP) CSF biomarker assay for C9orf72 -associated frontotemporal dementia and amyotrophic lateral sclerosis.
- Author
-
Wilson KM, Katona E, Glaria I, Carcolé M, Swift IJ, Sogorb-Esteve A, Heller C, Bouzigues A, Heslegrave AJ, Keshavan A, Knowles K, Patil S, Mohapatra S, Liu Y, Goyal J, Sanchez-Valle R, Laforce RJ, Synofzik M, Rowe JB, Finger E, Vandenberghe R, Butler CR, Gerhard A, Van Swieten JC, Seelaar H, Borroni B, Galimberti D, de Mendonça A, Masellis M, Tartaglia MC, Otto M, Graff C, Ducharme S, Schott JM, Malaspina A, Zetterberg H, Boyanapalli R, Rohrer JD, and Isaacs AM
- Subjects
- Biomarkers cerebrospinal fluid, C9orf72 Protein genetics, DNA Repeat Expansion genetics, Humans, Amyotrophic Lateral Sclerosis cerebrospinal fluid, Amyotrophic Lateral Sclerosis diagnosis, Amyotrophic Lateral Sclerosis genetics, Frontotemporal Dementia diagnosis, Frontotemporal Dementia genetics, Frontotemporal Dementia metabolism
- Abstract
Objective: A GGGGCC repeat expansion in the C9orf72 gene is the most common cause of genetic frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). As potential therapies targeting the repeat expansion are now entering clinical trials, sensitive biomarker assays of target engagement are urgently required. Our objective was to develop such an assay., Methods: We used the single molecule array (Simoa) platform to develop an immunoassay for measuring poly(GP) dipeptide repeat proteins (DPRs) generated by the C9orf72 repeat expansion in cerebrospinal fluid (CSF) of people with C9orf72 -associated FTD/ALS., Results and Conclusions: We show the assay to be highly sensitive and robust, passing extensive qualification criteria including low intraplate and interplate variability, a high precision and accuracy in measuring both calibrators and samples, dilutional parallelism, tolerance to sample and standard freeze-thaw and no haemoglobin interference. We used this assay to measure poly(GP) in CSF samples collected through the Genetic FTD Initiative (N=40 C9orf72 and 15 controls). We found it had 100% specificity and 100% sensitivity and a large window for detecting target engagement, as the C9orf72 CSF sample with the lowest poly(GP) signal had eightfold higher signal than controls and on average values from C9orf72 samples were 38-fold higher than controls, which all fell below the lower limit of quantification of the assay. These data indicate that a Simoa-based poly(GP) DPR assay is suitable for use in clinical trials to determine target engagement of therapeutics aimed at reducing C9orf72 repeat-containing transcripts., Competing Interests: Competing interests: SP, SM, YL, JG and RB were paid employees of Wave Life Sciences during completion of this work. JDR is on a Medical Advisory Board for Wave Life Sciences. JMS has received research funding from Avid Radiopharmaceuticals (a wholly owned subsidiary of Eli Lilly), has consulted for Roche Pharmaceuticals, Biogen, Merck and Eli Lilly, given educational lectures sponsored by GE Healthcare, Eli Lilly, and Biogen, and serves on a Data Safety Monitoring Committee for Axon Neuroscience SE. HZ has served at scientific advisory boards for Alector, Eisai, Denali, Roche Diagnostics, Wave, Samumed, Siemens Healthineers, Pinteon Therapeutics, Nervgen, AZTherapies and CogRx, has given lectures in symposia sponsored by Cellectricon, Fujirebio, Alzecure and Biogen, and is a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program (outside submitted work). JBR has provided consultancy unrelated to the current work for Asceneuron, Astex, Biogen, UCB, SV Health, Curasen., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
112. Blood biomarkers for Alzheimer's disease and related disorders.
- Author
-
Zetterberg H and Schott JM
- Subjects
- Amyloid beta-Peptides cerebrospinal fluid, Biomarkers, Humans, Peptide Fragments, tau Proteins cerebrospinal fluid, Alzheimer Disease diagnostic imaging, Neurodegenerative Diseases
- Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and the single commonest cause of dementia. Many other diseases can, however, cause dementia, and differential diagnosis can be challenging, especially in early disease stages. For most neurodegenerative dementias, accumulation of brain pathologies starts many years before clinical onset; the ability to detect these pathologies paves the way for targeted disease-modifying prevention trials. AD is associated with β-amyloid and tau pathologies, which can be quantified using cerebrospinal fluid and imaging biomarkers and, more recently, using highly sensitive blood tests. While for the most part, specific biomarkers of non-AD neurodegenerative dementias are lacking, non-specific biomarkers of neurodegeneration are available. This review summarizes recent advances in the neurodegenerative dementia blood biomarker research and discusses the next steps required for clinical implementation., (© 2022 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
113. Iatrogenic cerebral amyloid angiopathy: an emerging clinical phenomenon.
- Author
-
Banerjee G, Samra K, Adams ME, Jaunmuktane Z, Parry-Jones AR, Grieve J, Toma AK, Farmer SF, Sylvester R, Houlden H, Rudge P, Mead S, Brandner S, Schott JM, Collinge J, and Werring DJ
- Abstract
In the last 6 years, following the first pathological description of presumed amyloid-beta (Aβ) transmission in humans (in 2015) and subsequent experimental confirmation (in 2018), clinical cases of iatrogenic cerebral amyloid angiopathy (CAA)-attributed to the transmission of Aβ seeds-have been increasingly recognised and reported. This newly described form of CAA is associated with early disease onset (typically in the third to fifth decade), and often presents with intracerebral haemorrhage, but also seizures and cognitive impairment. Although assumed to be rare, it is important that clinicians remain vigilant for potential cases, particularly as the optimal management, prognosis, true incidence and public health implications remain unknown. This review summarises our current understanding of the clinical spectrum of iatrogenic CAA and provides a diagnostic framework for clinicians. We provide clinical details for three patients with pathological evidence of iatrogenic CAA and present a summary of the published cases to date (n=20), identified following a systematic review. Our aims are: (1) To describe the clinical features of iatrogenic CAA, highlighting important similarities and differences between iatrogenic and sporadic CAA; and (2) To discuss potential approaches for investigation and diagnosis, including suggested diagnostic criteria for iatrogenic CAA., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2022. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
114. AD and its comorbidities: An obstacle to develop a clinically efficient treatment?
- Author
-
Baker J and Schott JM
- Subjects
- Amyloid beta-Peptides therapeutic use, Humans, Alzheimer Disease drug therapy, Alzheimer Disease epidemiology
- Abstract
Whilst the development of new drugs designed for the treatment of Alzheimer's disease (AD) has been widely publicised, we do not yet have treatments that are proven to slow the progression of AD. The decision taken by the US Food and Drug Administration (FDA) to grant a licence for the use of aducanumab, based on the premise that β-amyloid removal would result in downstream benefits rather than demonstration of cognitive efficacy per se contrasts with that made by the European Medicines Agency (EMA), who declined to grant a licence, citing lack of evidence of clinical improvement, and a failure to demonstrate that the treatment was sufficiently safe. Multiple factors have complicated the search for new and effective treatments for the management of AD. It is a complex neurodegenerative condition in which multiple comorbidities are common in the affected population. However, such conditions are commonly exclusion criteria in clinical trials for new treatments. Here we discuss how some of these comorbidities impact the development of clinically efficient treatments for AD. Firstly, we will examine what is meant by AD, and how definitions of this condition have changed and continue to evolve. Secondly, we describe some of the most important comorbid conditions accompanying and in some cases mimicking AD. Finally, we will examine how the inclusion, or exclusion, of these conditions from AD research may have had an effect on treatment trials, the implications of co-morbidities on "real-life" use of novel therapeutics especially when these have been trialled in patients with relatively pure disease, and how clinical trials may need to adapt to account for comorbidities in the future., (Copyright © 2022. Published by Elsevier Masson SAS.)
- Published
- 2022
- Full Text
- View/download PDF
115. Circulating Metabolome and White Matter Hyperintensities in Women and Men.
- Author
-
Sliz E, Shin J, Ahmad S, Williams DM, Frenzel S, Gauß F, Harris SE, Henning AK, Hernandez MV, Hu YH, Jiménez B, Sargurupremraj M, Sudre C, Wang R, Wittfeld K, Yang Q, Wardlaw JM, Völzke H, Vernooij MW, Schott JM, Richards M, Proitsi P, Nauck M, Lewis MR, Launer L, Hosten N, Grabe HJ, Ghanbari M, Deary IJ, Cox SR, Chaturvedi N, Barnes J, Rotter JI, Debette S, Ikram MA, Fornage M, Paus T, Seshadri S, and Pausova Z
- Subjects
- Aged, Brain pathology, Female, Humans, Magnetic Resonance Imaging methods, Male, Metabolome, Middle Aged, Diabetes Mellitus, Type 2 pathology, White Matter diagnostic imaging
- Abstract
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 ( P
FDR )<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.- Published
- 2022
- Full Text
- View/download PDF
116. Sex-related differences in whole brain volumes at age 70 in association with hyperglycemia during adult life.
- Author
-
Fatih N, Chaturvedi N, Lane CA, Parker TD, Lu K, Cash DM, Malone IB, Silverwood R, Wong A, Barnes J, Sudre CH, Richards M, Fox NC, Schott JM, Hughes A, and James SN
- Subjects
- Adult, Aged, Brain diagnostic imaging, Female, Humans, Magnetic Resonance Imaging, Male, Positron-Emission Tomography, Hyperglycemia diagnostic imaging, Sex Characteristics
- Abstract
Longitudinal studies of the relationship between hyperglycemia and brain health are rare and there is limited information on sex differences in associations. We investigated whether glycosylated hemoglobin (HbA1c) measured at ages of 53, 60-64 and 69 years, and cumulative glycemic index (CGI), a measure of cumulative glycemic burden, were associated with metrics of brain health in later life. Participants were from Insight 46, a substudy of the Medical Research Council National Survey of Health and Development (NSHD) who undertook volumetric MRI, florbetapir amyloid-PET imaging and cognitive assessments at ages of 69-71. Analyses were performed using linear and logistic regression as appropriate, with adjustment for potential confounders. We observed a sex interaction between HbA1c and whole brain volume (WBV) at all 3 time points. Following stratification of our sample, we observed that HbA1c at all ages, and CGI were positively associated with lower WBV exclusively in females. HbA1c (or CGI) was not associated with amyloid status, white matter hyperintensities (WMHs), hippocampal volumes (HV) or cognitive outcomes in either sex. Higher HbA1c in adulthood is associated with smaller WBV at 69-71 years in females but not in males. This suggests that there may be preferential target organ damage in the brain for females with hyperglycemia., Competing Interests: Declaration of competing interest JS has received research funding from Avid Radiopharmaceuticals (a wholly owned subsidiary of Eli Lilly), has consulted for Roche Pharmaceuticals, Biogen and Eli Lilly, and serves on a Data Safety Monitoring Committee for Axon Neuroscience SE. AVID Radiopharmaceuticals (a wholly owned subsidiary of Eli Lilly) provide the PET β-amyloid tracer for Insight 46 (Florbetapir) but had no part in the design of the study. All other authors have no competing interests to declare., (Crown Copyright © 2021. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
117. Amyloid processing in COVID-19-associated neurological syndromes.
- Author
-
Ziff OJ, Ashton NJ, Mehta PR, Brown R, Athauda D, Heaney J, Heslegrave AJ, Benedet AL, Blennow K, Checkley AM, Houlihan CF, Gauthier S, Rosa-Neto P, Fox NC, Schott JM, Zetterberg H, Benjamin LA, and Paterson RW
- Subjects
- Amyloid beta-Peptides metabolism, Amyloid beta-Protein Precursor cerebrospinal fluid, Biomarkers cerebrospinal fluid, Cohort Studies, Cross-Sectional Studies, Humans, Pilot Projects, Prospective Studies, SARS-CoV-2, Alzheimer Disease cerebrospinal fluid, Amyloidosis, COVID-19 complications
- Abstract
SARS-CoV-2 infection can damage the nervous system with multiple neurological manifestations described. However, there is limited understanding of the mechanisms underlying COVID-19 neurological injury. This is a cross-sectional exploratory prospective biomarker cohort study of 21 patients with COVID-19 neurological syndromes (Guillain-Barre Syndrome [GBS], encephalitis, encephalopathy, acute disseminated encephalomyelitis [ADEM], intracranial hypertension, and central pain syndrome) and 23 healthy COVID-19 negative controls. We measured cerebrospinal fluid (CSF) and serum biomarkers of amyloid processing, neuronal injury (neurofilament light), astrocyte activation (GFAp), and neuroinflammation (tissue necrosis factor [TNF] ɑ, interleukin [IL]-6, IL-1β, IL-8). Patients with COVID-19 neurological syndromes had significantly reduced CSF soluble amyloid precursor protein (sAPP)-ɑ (p = 0.004) and sAPPβ (p = 0.03) as well as amyloid β (Aβ) 40 (p = 5.2 × 10
-8 ), Aβ42 (p = 3.5 × 10-7 ), and Aβ42/Aβ40 ratio (p = 0.005) compared to controls. Patients with COVID-19 neurological syndromes showed significantly increased neurofilament light (NfL, p = 0.001) and this negatively correlated with sAPPɑ and sAPPβ. Conversely, GFAp was significantly reduced in COVID-19 neurological syndromes (p = 0.0001) and this positively correlated with sAPPɑ and sAPPβ. COVID-19 neurological patients also displayed significantly increased CSF proinflammatory cytokines and these negatively correlated with sAPPɑ and sAPPβ. A sensitivity analysis of COVID-19-associated GBS revealed a non-significant trend toward greater impairment of amyloid processing in COVID-19 central than peripheral neurological syndromes. This pilot study raises the possibility that patients with COVID-19-associated neurological syndromes exhibit impaired amyloid processing. Altered amyloid processing was linked to neuronal injury and neuroinflammation but reduced astrocyte activation., (© 2022 The Authors. Journal of Neurochemistry published by John Wiley & Sons Ltd on behalf of International Society for Neurochemistry.)- Published
- 2022
- Full Text
- View/download PDF
118. New insights into the genetic etiology of Alzheimer's disease and related dementias.
- Author
-
Bellenguez C, Küçükali F, Jansen IE, Kleineidam L, Moreno-Grau S, Amin N, Naj AC, Campos-Martin R, Grenier-Boley B, Andrade V, Holmans PA, Boland A, Damotte V, van der Lee SJ, Costa MR, Kuulasmaa T, Yang Q, de Rojas I, Bis JC, Yaqub A, Prokic I, Chapuis J, Ahmad S, Giedraitis V, Aarsland D, Garcia-Gonzalez P, Abdelnour C, Alarcón-Martín E, Alcolea D, Alegret M, Alvarez I, Álvarez V, Armstrong NJ, Tsolaki A, Antúnez C, Appollonio I, Arcaro M, Archetti S, Pastor AA, Arosio B, Athanasiu L, Bailly H, Banaj N, Baquero M, Barral S, Beiser A, Pastor AB, Below JE, Benchek P, Benussi L, Berr C, Besse C, Bessi V, Binetti G, Bizarro A, Blesa R, Boada M, Boerwinkle E, Borroni B, Boschi S, Bossù P, Bråthen G, Bressler J, Bresner C, Brodaty H, Brookes KJ, Brusco LI, Buiza-Rueda D, Bûrger K, Burholt V, Bush WS, Calero M, Cantwell LB, Chene G, Chung J, Cuccaro ML, Carracedo Á, Cecchetti R, Cervera-Carles L, Charbonnier C, Chen HH, Chillotti C, Ciccone S, Claassen JAHR, Clark C, Conti E, Corma-Gómez A, Costantini E, Custodero C, Daian D, Dalmasso MC, Daniele A, Dardiotis E, Dartigues JF, de Deyn PP, de Paiva Lopes K, de Witte LD, Debette S, Deckert J, Del Ser T, Denning N, DeStefano A, Dichgans M, Diehl-Schmid J, Diez-Fairen M, Rossi PD, Djurovic S, Duron E, Düzel E, Dufouil C, Eiriksdottir G, Engelborghs S, Escott-Price V, Espinosa A, Ewers M, Faber KM, Fabrizio T, Nielsen SF, Fardo DW, Farotti L, Fenoglio C, Fernández-Fuertes M, Ferrari R, Ferreira CB, Ferri E, Fin B, Fischer P, Fladby T, Fließbach K, Fongang B, Fornage M, Fortea J, Foroud TM, Fostinelli S, Fox NC, Franco-Macías E, Bullido MJ, Frank-García A, Froelich L, Fulton-Howard B, Galimberti D, García-Alberca JM, García-González P, Garcia-Madrona S, Garcia-Ribas G, Ghidoni R, Giegling I, Giorgio G, Goate AM, Goldhardt O, Gomez-Fonseca D, González-Pérez A, Graff C, Grande G, Green E, Grimmer T, Grünblatt E, Grunin M, Gudnason V, Guetta-Baranes T, Haapasalo A, Hadjigeorgiou G, Haines JL, Hamilton-Nelson KL, Hampel H, Hanon O, Hardy J, Hartmann AM, Hausner L, Harwood J, Heilmann-Heimbach S, Helisalmi S, Heneka MT, Hernández I, Herrmann MJ, Hoffmann P, Holmes C, Holstege H, Vilas RH, Hulsman M, Humphrey J, Biessels GJ, Jian X, Johansson C, Jun GR, Kastumata Y, Kauwe J, Kehoe PG, Kilander L, Ståhlbom AK, Kivipelto M, Koivisto A, Kornhuber J, Kosmidis MH, Kukull WA, Kuksa PP, Kunkle BW, Kuzma AB, Lage C, Laukka EJ, Launer L, Lauria A, Lee CY, Lehtisalo J, Lerch O, Lleó A, Longstreth W Jr, Lopez O, de Munain AL, Love S, Löwemark M, Luckcuck L, Lunetta KL, Ma Y, Macías J, MacLeod CA, Maier W, Mangialasche F, Spallazzi M, Marquié M, Marshall R, Martin ER, Montes AM, Rodríguez CM, Masullo C, Mayeux R, Mead S, Mecocci P, Medina M, Meggy A, Mehrabian S, Mendoza S, Menéndez-González M, Mir P, Moebus S, Mol M, Molina-Porcel L, Montrreal L, Morelli L, Moreno F, Morgan K, Mosley T, Nöthen MM, Muchnik C, Mukherjee S, Nacmias B, Ngandu T, Nicolas G, Nordestgaard BG, Olaso R, Orellana A, Orsini M, Ortega G, Padovani A, Paolo C, Papenberg G, Parnetti L, Pasquier F, Pastor P, Peloso G, Pérez-Cordón A, Pérez-Tur J, Pericard P, Peters O, Pijnenburg YAL, Pineda JA, Piñol-Ripoll G, Pisanu C, Polak T, Popp J, Posthuma D, Priller J, Puerta R, Quenez O, Quintela I, Thomassen JQ, Rábano A, Rainero I, Rajabli F, Ramakers I, Real LM, Reinders MJT, Reitz C, Reyes-Dumeyer D, Ridge P, Riedel-Heller S, Riederer P, Roberto N, Rodriguez-Rodriguez E, Rongve A, Allende IR, Rosende-Roca M, Royo JL, Rubino E, Rujescu D, Sáez ME, Sakka P, Saltvedt I, Sanabria Á, Sánchez-Arjona MB, Sanchez-Garcia F, Juan PS, Sánchez-Valle R, Sando SB, Sarnowski C, Satizabal CL, Scamosci M, Scarmeas N, Scarpini E, Scheltens P, Scherbaum N, Scherer M, Schmid M, Schneider A, Schott JM, Selbæk G, Seripa D, Serrano M, Sha J, Shadrin AA, Skrobot O, Slifer S, Snijders GJL, Soininen H, Solfrizzi V, Solomon A, Song Y, Sorbi S, Sotolongo-Grau O, Spalletta G, Spottke A, Squassina A, Stordal E, Tartan JP, Tárraga L, Tesí N, Thalamuthu A, Thomas T, Tosto G, Traykov L, Tremolizzo L, Tybjærg-Hansen A, Uitterlinden A, Ullgren A, Ulstein I, Valero S, Valladares O, Broeckhoven CV, Vance J, Vardarajan BN, van der Lugt A, Dongen JV, van Rooij J, van Swieten J, Vandenberghe R, Verhey F, Vidal JS, Vogelgsang J, Vyhnalek M, Wagner M, Wallon D, Wang LS, Wang R, Weinhold L, Wiltfang J, Windle G, Woods B, Yannakoulia M, Zare H, Zhao Y, Zhang X, Zhu C, Zulaica M, Farrer LA, Psaty BM, Ghanbari M, Raj T, Sachdev P, Mather K, Jessen F, Ikram MA, de Mendonça A, Hort J, Tsolaki M, Pericak-Vance MA, Amouyel P, Williams J, Frikke-Schmidt R, Clarimon J, Deleuze JF, Rossi G, Seshadri S, Andreassen OA, Ingelsson M, Hiltunen M, Sleegers K, Schellenberg GD, van Duijn CM, Sims R, van der Flier WM, Ruiz A, Ramirez A, and Lambert JC
- Subjects
- Genome-Wide Association Study, Humans, tau Proteins genetics, Alzheimer Disease genetics, Alzheimer Disease pathology, Cognitive Dysfunction psychology
- Abstract
Characterization of the genetic landscape of Alzheimer's disease (AD) and related dementias (ADD) provides a unique opportunity for a better understanding of the associated pathophysiological processes. We performed a two-stage genome-wide association study totaling 111,326 clinically diagnosed/'proxy' AD cases and 677,663 controls. We found 75 risk loci, of which 42 were new at the time of analysis. Pathway enrichment analyses confirmed the involvement of amyloid/tau pathways and highlighted microglia implication. Gene prioritization in the new loci identified 31 genes that were suggestive of new genetically associated processes, including the tumor necrosis factor alpha pathway through the linear ubiquitin chain assembly complex. We also built a new genetic risk score associated with the risk of future AD/dementia or progression from mild cognitive impairment to AD/dementia. The improvement in prediction led to a 1.6- to 1.9-fold increase in AD risk from the lowest to the highest decile, in addition to effects of age and the APOE ε4 allele., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
119. Cerebrospinal fluid metallomics in cerebral amyloid angiopathy: an exploratory analysis.
- Author
-
Banerjee G, Forsgard N, Ambler G, Keshavan A, Paterson RW, Foiani MS, Toombs J, Heslegrave A, Thompson EJ, Lunn MP, Fox NC, Zetterberg H, Schott JM, and Werring DJ
- Subjects
- Amyloid beta-Peptides cerebrospinal fluid, Biomarkers cerebrospinal fluid, Cerebral Hemorrhage complications, Humans, Alzheimer Disease complications, Cerebral Amyloid Angiopathy complications
- Abstract
Introduction: Cerebral amyloid angiopathy (CAA) is associated with symptomatic intracerebral haemorrhage. Biomarkers of clinically silent bleeding events, such as cerebrospinal fluid (CSF) ferritin and iron, might provide novel measures of disease presence and severity., Methods: We performed an exploratory study comparing CSF iron, ferritin, and other metal levels in patients with CAA, control subjects (CS) and patients with Alzheimer's disease (AD). Ferritin was measured using a latex fixation test; metal analyses were performed using inductively coupled plasma mass spectrometry., Results: CAA patients (n = 10) had higher levels of CSF iron than the AD (n = 20) and CS (n = 10) groups (medians 23.42, 15.48 and 17.71 μg/L, respectively, p = 0.0015); the difference between CAA and AD groups was significant in unadjusted and age-adjusted analyses. We observed a difference in CSF ferritin (medians 10.10, 7.77 and 8.01 ng/ml, for CAA, AD and CS groups, respectively, p = 0.01); the difference between the CAA and AD groups was significant in unadjusted, but not age-adjusted, analyses. We also observed differences between the CAA and AD groups in CSF nickel and cobalt (unadjusted analyses)., Conclusions: In this exploratory study, we provide preliminary evidence for a distinct CSF metallomic profile in patients with CAA. Replication and validation of these results in larger cohorts is needed., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
120. Straight and Divergent Pathways to Cognitive State: Seven Decades of Follow-Up in the British 1946 Birth Cohort.
- Author
-
Richards M, James SN, Lu K, Livingston G, Schott JM, Lane CA, Barnes J, Parker TD, Sudre CH, Cash DM, Coath W, Fox N, and Davis DHJ
- Subjects
- Cognition, Follow-Up Studies, Humans, Memory Disorders, Neuropsychological Tests, Reproducibility of Results, Birth Cohort, Cognitive Dysfunction diagnostic imaging
- Abstract
Background: Using the British 1946 birth cohort we previously estimated life course paths to the Addenbrooke's Cognitive Examination (ACE-III)., Objective: We now compared those whose ACE-III scores were expected, worse and better than predicted from the path model on a range of independent variables including clinical ratings of cognitive impairment and neuroimaging measures., Methods: Predicted ACE-III scores were categorized into three groups: those with Expected (between -1.5 and 1.5 standard deviation; SD); Worse (< -1.5 SD); and Better (>1.5 SD) scores. Differences in the independent variables were then tested between these three groups., Results: Compared with the Expected group, those in the Worse group showed independent evidence of progressive cognitive impairment: faster memory decline, more self-reported memory difficulties, more functional difficulties, greater likelihood of being independently rated by experienced specialist clinicians as having a progressive cognitive impairment, and a cortical thinning pattern suggestive of preclinical Alzheimer's disease. Those in the Better group showed slower verbal memory decline and absence of independently rated progressive cognitive impairment compared to the Expected group, but no differences in any of the other independent variables including the neuroimaging variables., Conclusion: The residual approach shows that life course features can map directly to clinical diagnoses. One future challenge is to translate this into a readily usable algorithm to identify high-risk individuals in preclinical state, when preventive strategies and therapeutic interventions may be most effective.
- Published
- 2022
- Full Text
- View/download PDF
121. Not all voxels are created equal: Reducing estimation bias in regional NODDI metrics using tissue-weighted means.
- Author
-
Parker CS, Veale T, Bocchetta M, Slattery CF, Malone IB, Thomas DL, Schott JM, Cash DM, and Zhang H
- Subjects
- Adult, Bias, Humans, Image Processing, Computer-Assisted, Models, Neurological, Phenotype, Alzheimer Disease cerebrospinal fluid, Alzheimer Disease diagnostic imaging, Diffusion Tensor Imaging methods, Neurites ultrastructure, White Matter diagnostic imaging
- Abstract
Neurite orientation dispersion and density imaging (NODDI) estimates microstructural properties of brain tissue relating to the organisation and processing capacity of neurites, which are essential elements for neuronal communication. Descriptive statistics of NODDI tissue metrics are commonly analyzed in regions-of-interest (ROI) to identify brain-phenotype associations. Here, the conventional method to calculate the ROI mean weights all voxels equally. However, this produces biased estimates in the presence of CSF partial volume. This study introduces the tissue-weighted mean, which calculates the mean NODDI metric across the tissue within an ROI, utilising the tissue fraction estimate from NODDI to reduce estimation bias. We demonstrate the proposed mean in a study of white matter abnormalities in young onset Alzheimer's disease (YOAD). Results show the conventional mean induces significant bias that correlates with CSF partial volume, primarily affecting periventricular regions and more so in YOAD subjects than in healthy controls. Due to the differential extent of bias between healthy controls and YOAD subjects, the conventional mean under- or over-estimated the effect size for group differences in many ROIs. This demonstrates the importance of using the correct estimation procedure when inferring group differences in studies where the extent of CSF partial volume differs between groups. These findings are robust across different acquisition and processing conditions. Bias persists in ROIs at higher image resolution, as demonstrated using data obtained from the third phase of the Alzheimer's disease neuroimaging initiative (ADNI); and when performing ROI analysis in template space. This suggests that conventional ROI means of NODDI metrics are biased estimates under most contemporary experimental conditions, the correction of which requires the proposed tissue-weighted mean. The tissue-weighted mean produces accurate estimates of ROI means and group differences when ROIs contain voxels with CSF partial volume. In addition to NODDI, the technique can be applied to other multi-compartment models that account for CSF partial volume, such as the free water elimination method. We expect the technique to help generate new insights into normal and abnormal variation in tissue microstructure of regions typically confounded by CSF partial volume, such as those in individuals with larger ventricles due to atrophy associated with neurodegenerative disease., (Copyright © 2021. Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
122. Beyond the average patient: how neuroimaging models can address heterogeneity in dementia.
- Author
-
Verdi S, Marquand AF, Schott JM, and Cole JH
- Subjects
- Alzheimer Disease diagnostic imaging, Alzheimer Disease epidemiology, Case-Control Studies, Databases, Factual statistics & numerical data, Dementia epidemiology, Humans, Neuroimaging standards, Dementia diagnostic imaging, Models, Neurological, Neuroimaging methods
- Abstract
Dementia is a highly heterogeneous condition, with pronounced individual differences in age of onset, clinical presentation, progression rates and neuropathological hallmarks, even within a specific diagnostic group. However, the most common statistical designs used in dementia research studies and clinical trials overlook this heterogeneity, instead relying on comparisons of group average differences (e.g. patient versus control or treatment versus placebo), implicitly assuming within-group homogeneity. This one-size-fits-all approach potentially limits our understanding of dementia aetiology, hindering the identification of effective treatments. Neuroimaging has enabled the characterization of the average neuroanatomical substrates of dementias; however, the increasing availability of large open neuroimaging datasets provides the opportunity to examine patterns of neuroanatomical variability in individual patients. In this update, we outline the causes and consequences of heterogeneity in dementia and discuss recent research that aims to tackle heterogeneity directly, rather than assuming that dementia affects everyone in the same way. We introduce spatial normative modelling as an emerging data-driven technique, which can be applied to dementia data to model neuroanatomical variation, capturing individualized neurobiological 'fingerprints'. Such methods have the potential to detect clinically relevant subtypes, track an individual's disease progression or evaluate treatment responses, with the goal of moving towards precision medicine for dementia., (© The Author(s) (2021). Published by Oxford University Press on behalf of the Guarantors of Brain.)
- Published
- 2021
- Full Text
- View/download PDF
123. Loss and dispersion of superficial white matter in Alzheimer's disease: a diffusion MRI study.
- Author
-
Veale T, Malone IB, Poole T, Parker TD, Slattery CF, Paterson RW, Foulkes AJM, Thomas DL, Schott JM, Zhang H, Fox NC, and Cash DM
- Abstract
Pathological cerebral white matter changes in Alzheimer's disease have been shown using diffusion tensor imaging. Superficial white matter changes are relatively understudied despite their importance in cortico-cortical connections. Measuring superficial white matter degeneration using diffusion tensor imaging is challenging due to its complex organizational structure and proximity to the cortex. To overcome this, we investigated diffusion MRI changes in young-onset Alzheimer's disease using standard diffusion tensor imaging and Neurite Orientation Dispersion and Density Imaging to distinguish between disease-related changes that are degenerative (e.g. loss of myelinated fibres) and organizational (e.g. increased fibre dispersion). Twenty-nine young-onset Alzheimer's disease patients and 22 healthy controls had both single-shell and multi-shell diffusion MRI. We calculated fractional anisotropy, mean diffusivity, neurite density index, orientation dispersion index and tissue fraction (1-free water fraction). Diffusion metrics were sampled in 15 a priori regions of interest at four points along the cortical profile: cortical grey matter, grey/white boundary, superficial white matter (1 mm below grey/white boundary) and superficial/deeper white matter (2 mm below grey/white boundary). To estimate cross-sectional group differences, we used average marginal effects from linear mixed effect models of participants' diffusion metrics along the cortical profile. The superficial white matter of young-onset Alzheimer's disease individuals had lower neurite density index compared to controls in five regions (superior and inferior parietal, precuneus, entorhinal and parahippocampus) (all P < 0.05), and higher orientation dispersion index in three regions (fusiform, entorhinal and parahippocampus) (all P < 0.05). Young-onset Alzheimer's disease individuals had lower fractional anisotropy in the entorhinal and parahippocampus regions (both P < 0.05) and higher fractional anisotropy within the postcentral region ( P < 0.05). Mean diffusivity was higher in the young-onset Alzheimer's disease group in the parahippocampal region ( P < 0.05) and lower in the postcentral, precentral and superior temporal regions (all P < 0.05). In the overlying grey matter, disease-related changes were largely consistent with superficial white matter findings when using neurite density index and fractional anisotropy, but appeared at odds with orientation dispersion and mean diffusivity. Tissue fraction was significantly lower across all grey matter regions in young-onset Alzheimer's disease individuals (all P < 0.001) but group differences reduced in magnitude and coverage when moving towards the superficial white matter. These results show that microstructural changes occur within superficial white matter and along the cortical profile in individuals with young-onset Alzheimer's disease. Lower neurite density and higher orientation dispersion suggests underlying fibres undergo neurodegeneration and organizational changes, two effects previously indiscernible using standard diffusion tensor metrics in superficial white matter., (© The Author(s) (2021). Published by Oxford University Press on behalf of the Guarantors of Brain.)
- Published
- 2021
- Full Text
- View/download PDF
124. Mild Parkinsonian Signs: A Systematic Review of Clinical, Imaging, and Pathological Associations.
- Author
-
Buchanan SM, Richards M, Schott JM, and Schrag A
- Subjects
- Aged, Aged, 80 and over, Humans, Lewy Bodies pathology, Substantia Nigra pathology, Neurodegenerative Diseases pathology, Parkinson Disease complications, Parkinson Disease diagnostic imaging, Parkinson Disease epidemiology, Parkinsonian Disorders complications, Parkinsonian Disorders diagnostic imaging, Parkinsonian Disorders epidemiology
- Abstract
Mild parkinsonian signs (MPS) have been widely studied during the past 3 decades and proposed as a risk marker for neurodegenerative disease. This systematic review explores the epidemiology, clinical and prognostic associations, radiological features, and pathological findings associated with MPS in older adults free from neurodegenerative disease. We find that MPS as currently defined are strongly associated with increasing age and increased risk of development of Parkinson's disease (PD), all-cause dementia, disability, and death. Positive associations with later PD are found mainly in younger populations and those with other features of prodromal PD. There are currently no consistent radiological findings for MPS, and pathological studies have shown that MPS, at least in the oldest old, are often underpinned by mixed neuropathologies, including those associated with Alzheimer's disease, cerebrovascular disease, nigral neuronal loss, and Lewy bodies. Different subcategories of MPS appear to convey varying risk and specificity for PD and other outcomes. MPS overall are not specific for parkinsonian disorders and, although associated with increased risk of PD, can reflect multiple pathologies, particularly in older individuals. "Mild motor signs" appears a more appropriate term to avoid prognostic and pathological implications, and larger future studies to prospectively examine outcomes and associations of specific MPS subcategories are required. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society., (© 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.)
- Published
- 2021
- Full Text
- View/download PDF
125. Subjective cognitive complaints at age 70: associations with amyloid and mental health.
- Author
-
Pavisic IM, Lu K, Keuss SE, James SN, Lane CA, Parker TD, Keshavan A, Buchanan SM, Murray-Smith H, Cash DM, Coath W, Wong A, Fox NC, Crutch SJ, Richards M, and Schott JM
- Subjects
- Aged, Anxiety diagnostic imaging, Anxiety metabolism, Brain diagnostic imaging, Brain metabolism, Cross-Sectional Studies, Depression diagnostic imaging, Depression metabolism, Female, Humans, Magnetic Resonance Imaging, Male, Neuropsychological Tests, Positron-Emission Tomography, Amyloid beta-Peptides metabolism, Anxiety psychology, Cognition physiology, Depression psychology, Mental Health
- Abstract
Objective: To investigate subjective cognitive decline (SCD) in relation to β-amyloid pathology and to test for associations with anxiety, depression, objective cognition and family history of dementia in the Insight 46 study., Methods: Cognitively unimpaired ~70-year-old participants, all born in the same week in 1946 (n=460, 49% female, 18% amyloid-positive), underwent assessments including the SCD-Questionnaire (MyCog). MyCog scores were evaluated with respect to
18 F-Florbetapir-PET amyloid status (positive/negative). Associations with anxiety, depression, objective cognition (measured by the Preclinical Alzheimer Cognitive Composite, PACC) and family history of dementia were also investigated. The informant's perspective on SCD was evaluated in relation to MyCog score., Results: Anxiety (mean (SD) trait anxiety score: 4.4 (3.9)) was associated with higher MyCog scores, especially in women. MyCog scores were higher in amyloid-positive compared with amyloid-negative individuals (adjusted means (95% CIs): 5.3 (4.4 to 6.1) vs 4.3 (3.9 to 4.7), p=0.044), after accounting for differences in anxiety. PACC (mean (SD) -0.05 (0.68)) and family history of dementia (prevalence: 23.9%) were not independently associated with MyCog scores. The informant's perception of SCD was generally in accordance with that of the participant., Conclusions: This cross-sectional study demonstrates that symptoms of SCD are associated with both β-amyloid pathology, and more consistently, trait anxiety in a population-based cohort of older adults, at an age when those who are destined to develop dementia are still likely to be some years away from symptoms. This highlights the necessity of considering anxiety symptoms when assessing Alzheimer's disease pathology and SCD., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY. Published by BMJ.)- Published
- 2021
- Full Text
- View/download PDF
126. Dissociable effects of APOE -ε4 and β-amyloid pathology on visual working memory.
- Author
-
Lu K, Nicholas JM, Pertzov Y, Grogan J, Husain M, Pavisic IM, James SN, Parker TD, Lane CA, Keshavan A, Keuss SE, Buchanan SM, Murray-Smith H, Cash DM, Malone IB, Sudre CH, Coath W, Wong A, Henley SMD, Fox NC, Richards M, Schott JM, and Crutch SJ
- Subjects
- Humans, Amyloid beta-Peptides genetics, Memory, Short-Term, Apolipoprotein E4 genetics, Genotype, Alzheimer Disease diagnostic imaging
- Abstract
Although APOE -ε4 carriers are at significantly higher risk of developing Alzheimer's disease than non-carriers
1 , controversial evidence suggests that APOE -ε4 might confer some advantages, explaining the survival of this gene (antagonistic pleiotropy)2,3 . In a population-based cohort born in one week in 1946 (assessed aged 69-71), we assessed differential effects of APOE -ε4 and β-amyloid pathology (quantified using18 F-Florbetapir-PET) on visual working memory (object-location binding). In 398 cognitively normal participants, APOE -ε4 and β-amyloid had opposing effects on object identification, predicting better and poorer recall respectively. ε4-carriers also recalled locations more precisely, with a greater advantage at higher β-amyloid burden. These results provide evidence of superior visual working memory in ε4-carriers, showing that some benefits of this genotype are demonstrable in older age, even in the preclinical stages of Alzheimer's disease., Competing Interests: Competing Interests Statement All authors declare no competing interests.- Published
- 2021
- Full Text
- View/download PDF
127. Reply: Functional cognitive disorder: dementia's blind spot.
- Author
-
Ball HA, McWhirter L, Ballard C, Bhome R, Blackburn DJ, Edwards MJ, Fox NC, Howard R, Huntley J, Isaacs JD, Larner AJ, Nicholson TR, Pennington CM, Poole N, Price G, Price JP, Reuber M, Ritchie C, Rossor MN, Schott JM, Venneri A, Stone J, and Carson AJ
- Subjects
- Cognition, Humans, Cognition Disorders etiology, Dementia complications
- Published
- 2021
- Full Text
- View/download PDF
128. The global Alzheimer's Association round robin study on plasma amyloid β methods.
- Author
-
Pannee J, Shaw LM, Korecka M, Waligorska T, Teunissen CE, Stoops E, Vanderstichele HMJ, Mauroo K, Verberk IMW, Keshavan A, Pesini P, Sarasa L, Pascual-Lucas M, Fandos N, Allué JA, Portelius E, Andreasson U, Yoda R, Nakamura A, Kaneko N, Yang SY, Liu HC, Palme S, Bittner T, Mawuenyega KG, Ovod V, Bollinger J, Bateman RJ, Li Y, Dage JL, Stomrud E, Hansson O, Schott JM, Blennow K, and Zetterberg H
- Abstract
Introduction: Blood-based assays to measure brain amyloid beta (Aβ) deposition are an attractive alternative to the cerebrospinal fluid (CSF)-based assays currently used in clinical settings. In this study, we examined different blood-based assays to measure Aβ and how they compare among centers and assays., Methods: Aliquots from 81 plasma samples were distributed to 10 participating centers. Seven immunological assays and four mass-spectrometric methods were used to measure plasma Aβ concentrations., Results: Correlations were weak for Aβ42 while Aβ40 correlations were stronger. The ratio Aβ42/Aβ40 did not improve the correlations and showed weak correlations., Discussion: The poor correlations for Aβ42 in plasma might have several potential explanations, such as the high levels of plasma proteins (compared to CSF), sensitivity to pre-analytical sample handling and specificity, and cross-reactivity of different antibodies. Different methods might also measure different pools of plasma Aβ42. We, however, hypothesize that greater correlations might be seen in future studies because many of the methods have been refined during completion of this study., Competing Interests: OH has acquired research support (for the institution) from Avid Radiopharmaceuticals, Biogen, Eli Lilly, Eisai, GE Healthcare, Pfizer, and Roche. In the past 2 years, he has received consultancy/speaker fees from AC Immune, Alzpath, Biogen, Cerveau, and Roche. SP is a full‐time employee of Roche Diagnostics GmbH and holds shares in Roche. TB is a full‐time employee of and owns stock in F. Hoffmann‐La Roche Ltd. KB has served as a consultant, on advisory boards, or on data monitoring committees for Abcam, Axon, Biogen, JOMDD/Shimadzu, Julius Clinical, Lilly, MagQu, Novartis, Prothena, Roche Diagnostics, and Siemens Healthineers, and is a co‐founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program. HZ has served on scientific advisory boards for Alector, Eisai, Denali, Roche Diagnostics, Wave, Samumed, Siemens Healthineers, Pinteon Therapeutics, Nervgen, AZTherapies and CogRx. JLD is an employee and stockholder of Eli Lilly and Company. ELECSYS is a trademark of Roche. E Stoops and KM are full‐time paid employees of ADx NeuroSciences. JMS has received research funding from Avid Radiopharmaceuticals (a wholly owned subsidiary of Eli Lilly and Company); has consulted for Roche Pharmaceuticals, Biogen, Merck, and Eli Lilly; given educational lectures sponsored by GE Healthcare, Eli Lilly, and Biogen; and serves on a Data Safety Monitoring Committee for Axon Neuroscience SE. RJB cofounded C2N Diagnostics. Washington University and Dr. Bateman have equity ownership interest in C2N Diagnostics and receive royalty income based on technology (stable isotope labeling kinetics and blood plasma assay) licensed by Washington University to C2N Diagnostics. He receives income from C2N Diagnostics for serving on the scientific advisory board. Washington University, with RJB as co‐inventor, has submitted the US provisional patent application “Plasma Based Methods for Detecting CNS Amyloid Deposition.” He has received consultant fees from Roche, C2N Diagnostics, Genentech, AbbVie, Pfizer, Boehringer‐Ingelheim, Eisai, AC Immune, Janssen, and Merck. He serves as principal investigator of the DIAN‐TU, which is supported by the Alzheimer's Association, GHR Foundation, Eisai, an anonymous organization ,and the DIAN‐TU Pharma Consortium. HV is a founder of Biomarkable and a co‐founder of ADx NeuroSciences. RY and NK are full‐time employees of Shimadzu Corporation. NK holds stock in Shimadzu Corporation and has received payment for manuscript writing from Rinshohoushasen. CET has a collaboration contract with ADx Neurosciences and Quanterix; performed contract research or received grants from AC‐Immune, Axon Neurosciences, Biogen, Brainstorm Therapeutics, Celgene, EIP Pharma, Eisai, PeopleBio, Roche, Toyama, Vivoryon; received honoraria from Medidact Neurologie. LMS has received honorarium from Biogen for teaching. LS and JA have submitted patents for “Methods for quantification of amyloid beta peptides in plasma by mass spectrometry.” AN received honoraria from The Educational Program for Dementia Experts in Hokuriku (NINPRO), The Japan Society for the Promotion of Science (JSPS), Translational Research Center for Medical Innovation (TRI), Eisai Co. Ltd. SY is an employee and shareholder of MagQu Co., Ltd. KGM, VO, and JB have submitted patent application “Plasma Based Methods for Detecting CNS Amyloid Deposition” and may receive royalties based on blood plasma assay technology licensed to C2N Diagnostics. ES has received payment (to institution) from Roche Diagnostics for medical writing., (© 2021 The Authors. Alzheimer's & Dementia: Diagnosis, Assessment & Disease Monitoring published by Wiley Periodicals, LLC on behalf of Alzheimer's Association.)
- Published
- 2021
- Full Text
- View/download PDF
129. Negotiating Time and Space When Working From Home: Experiences During COVID-19.
- Author
-
Fukumura YE, Schott JM, Lucas GM, Becerik-Gerber B, and Roll SC
- Subjects
- Adult, Demography, Female, Humans, Male, Surveys and Questionnaires, United States, COVID-19, Environment, SARS-CoV-2, Social Isolation, Teleworking
- Abstract
Stay-at-home mandates following the COVID-19 pandemic increased work from home (WFH). While WFH offers many benefits, navigating work in nontraditional contexts can be a challenge. The objective of this study was to explore the benefits and challenges of WFH during COVID-19 to identify supports and resources necessary. Comments from two free-response questions on a survey regarding experiences of WFH ( N = 648, N = 366) were analyzed using inductive qualitative content analysis. Four themes emerged: time use, considerations of working in the home space, intersections between work-life and home-life, and temporality of WFH as situated within a pandemic. Across all themes were concerns related to participation in both work and home roles, work performance, and well-being. Findings highlight the importance of support during times of disruption of occupational patterns, roles, and routines. Despite challenges, many individuals hoped to continue WFH. Organizations should consider the complex intersections of work-life and home-life to develop supportive policies and resources.
- Published
- 2021
- Full Text
- View/download PDF
130. Evaluation of plasma tau and neurofilament light chain biomarkers in a 12-year clinical cohort of human prion diseases.
- Author
-
Thompson AGB, Anastasiadis P, Druyeh R, Whitworth I, Nayak A, Nihat A, Mok TH, Rudge P, Wadsworth JDF, Rohrer J, Schott JM, Heslegrave A, Zetterberg H, Collinge J, Jackson GS, and Mead S
- Subjects
- Biomarkers, Humans, Neurofilament Proteins genetics, Prospective Studies, tau Proteins, Intermediate Filaments, Prion Diseases genetics
- Abstract
Prion diseases are fatal neurodegenerative conditions with highly accurate CSF and imaging diagnostic tests, but major unmet needs for blood biomarkers. Using ultrasensitive immuno-assays, we measured tau and neurofilament light chain (NfL) protein concentrations in 709 plasma samples taken from 377 individuals with prion disease during a 12 year prospective clinical study, alongside healthy and neurological control groups. This provides an unprecedented opportunity to evaluate their potential as biomarkers. Plasma tau and NfL were increased across all prion disease types. For distinguishing sCJD from control groups including clinically-relevant "CJD mimics", both show considerable diagnostic value. In sCJD, NfL was substantially elevated in every sample tested, including during early disease with minimal functional impairment and in all follow-up samples. Plasma tau was independently associated with rate of clinical progression in sCJD, while plasma NfL showed independent association with severity of functional impairment. In asymptomatic PRNP mutation carriers, plasma NfL was higher on average in samples taken within 2 years of symptom onset than in samples taken earlier. We present biomarker trajectories for nine mutation carriers healthy at enrolment who developed symptoms during follow-up. NfL started to rise as early as 2 years before onset in those with mutations typically associated with more slowly progressive clinical disease. This shows potential for plasma NfL as a "proximity marker", but further work is needed to establish predictive value on an individual basis, and how this varies across different PRNP mutations. We conclude that plasma tau and NfL have potential to fill key unmet needs for biomarkers in prion disease: as a secondary outcome for clinical trials (NfL and tau); for predicting onset in at-risk individuals (NfL); and as an accessible test for earlier identification of patients that may have CJD and require more definitive tests (NfL). Further studies should evaluate their performance directly in these specific roles., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
131. Correction to: Grip strength from midlife as an indicator of later-life brain health and cognition: evidence from a British birth cohort.
- Author
-
Dercon Q, Nicholas JM, James SN, Schott JM, and Richards M
- Published
- 2021
- Full Text
- View/download PDF
132. Transitioning from cerebrospinal fluid to blood tests to facilitate diagnosis and disease monitoring in Alzheimer's disease.
- Author
-
Alawode DOT, Heslegrave AJ, Ashton NJ, Karikari TK, Simrén J, Montoliu-Gaya L, Pannee J, O Connor A, Weston PSJ, Lantero-Rodriguez J, Keshavan A, Snellman A, Gobom J, Paterson RW, Schott JM, Blennow K, Fox NC, and Zetterberg H
- Subjects
- Amyloid beta-Peptides, Biomarkers blood, Humans, Peptide Fragments, tau Proteins, Alzheimer Disease diagnosis, Cerebrospinal Fluid, Hematologic Tests
- Abstract
Alzheimer's disease (AD) is increasingly prevalent worldwide, and disease-modifying treatments may soon be at hand; hence, now, more than ever, there is a need to develop techniques that allow earlier and more secure diagnosis. Current biomarker-based guidelines for AD diagnosis, which have replaced the historical symptom-based guidelines, rely heavily on neuroimaging and cerebrospinal fluid (CSF) sampling. While these have greatly improved the diagnostic accuracy of AD pathophysiology, they are less practical for application in primary care, population-based and epidemiological settings, or where resources are limited. In contrast, blood is a more accessible and cost-effective source of biomarkers in AD. In this review paper, using the recently proposed amyloid, tau and neurodegeneration [AT(N)] criteria as a framework towards a biological definition of AD, we discuss recent advances in biofluid-based biomarkers, with a particular emphasis on those with potential to be translated into blood-based biomarkers. We provide an overview of the research conducted both in CSF and in blood to draw conclusions on biomarkers that show promise. Given the evidence collated in this review, plasma neurofilament light chain (N) and phosphorylated tau (p-tau; T) show particular potential for translation into clinical practice. However, p-tau requires more comparisons to be conducted between its various epitopes before conclusions can be made as to which one most robustly differentiates AD from non-AD dementias. Plasma amyloid beta (A) would prove invaluable as an early screening modality, but it requires very precise tests and robust pre-analytical protocols., (© 2021 The Authors. Journal of Internal Medicine published by John Wiley & Sons Ltd on behalf of Association for Publication of The Journal of Internal Medicine.)
- Published
- 2021
- Full Text
- View/download PDF
133. Antiphospholipid antibodies and neurological manifestations in acute COVID-19: A single-centre cross-sectional study.
- Author
-
Benjamin LA, Paterson RW, Moll R, Pericleous C, Brown R, Mehta PR, Athauda D, Ziff OJ, Heaney J, Checkley AM, Houlihan CF, Chou M, Heslegrave AJ, Chandratheva A, Michael BD, Blennow K, Vivekanandam V, Foulkes A, Mummery CJ, Lunn MP, Keddie S, Spyer MJ, Mckinnon T, Hart M, Carletti F, Jäger HR, Manji H, Zandi MS, Werring DJ, Nastouli E, Simister R, Solomon T, Zetterberg H, Schott JM, Cohen H, and Efthymiou M
- Abstract
Background: A high prevalence of antiphospholipid antibodies has been reported in case series of patients with neurological manifestations and COVID-19; however, the pathogenicity of antiphospholipid antibodies in COVID-19 neurology remains unclear., Methods: This single-centre cross-sectional study included 106 adult patients: 30 hospitalised COVID-neurological cases, 47 non-neurological COVID-hospitalised controls, and 29 COVID-non-hospitalised controls, recruited between March and July 2020. We evaluated nine antiphospholipid antibodies: anticardiolipin antibodies [aCL] IgA, IgM, IgG; anti-beta-2 glycoprotein-1 [aβ
2 GPI] IgA, IgM, IgG; anti-phosphatidylserine/prothrombin [aPS/PT] IgM, IgG; and anti-domain I β2 GPI (aD1β2GPI) IgG., Findings: There was a high prevalence of antiphospholipid antibodies in the COVID-neurological (73.3%) and non-neurological COVID-hospitalised controls (76.6%) in contrast to the COVID-non-hospitalised controls (48.2%). aPS/PT IgG titres were significantly higher in the COVID-neurological group compared to both control groups ( p < 0.001). Moderate-high titre of aPS/PT IgG was found in 2 out of 3 (67%) patients with acute disseminated encephalomyelitis [ADEM]. aPS/PT IgG titres negatively correlated with oxygen requirement (FiO2 R =-0.15 p = 0.040) and was associated with venous thromboembolism ( p = 0.043). In contrast, aCL IgA ( p < 0.001) and IgG ( p < 0.001) was associated with non-neurological COVID-hospitalised controls compared to the other groups and correlated positively with d-dimer and creatinine but negatively with FiO2 ., Interpretation: Our findings show that aPS/PT IgG is associated with COVID-19-associated ADEM. In contrast, aCL IgA and IgG are seen much more frequently in non-neurological hospitalised patients with COVID-19. Characterisation of antiphospholipid antibody persistence and potential longitudinal clinical impact are required to guide appropriate management., Funding: This work is supported by UCL Queen Square Biomedical Research Centre (BRC) and Moorfields BRC grants (#560441 and #557595). LB is supported by a Wellcome Trust Fellowship (222102/Z/20/Z). RWP is supported by an Alzheimer's Association Clinician Scientist Fellowship (AACSF-20-685780) and the UK Dementia Research Institute. KB is supported by the Swedish Research Council (#2017-00915) and the Swedish state under the agreement between the Swedish government and the County Councils, the ALF-agreement (#ALFGBG-715986). HZ is a Wallenberg Scholar supported by grants from the Swedish Research Council (#2018-02532), the European Research Council (#681712), Swedish State Support for Clinical Research (#ALFGBG-720931), the Alzheimer Drug Discovery Foundation (ADDF), USA (#201809-2016862), and theUK Dementia Research Institute at UCL. BDM is supported by grants from the MRC/UKRI (MR/V007181/1), MRC (MR/T028750/1) and Wellcome (ISSF201902/3). MSZ, MH and RS are supported by the UCL/UCLH NIHR Biomedical Research Centre and MSZ is supported by Queen Square National Brain Appeal., Competing Interests: LAB reports grants from GlaxoSmithKline, grants from Wellcome Trust, outside the submitted work.; RWP reports grants from Neurofilament light consortium, personal fees from GE healthcare educational talk, outside the submitted work.; CP is co-inventor for a patented Domain I-based potential therapeutic for APS.; KB reports personal fees from Abcam (advisory board / consultant), personal fees from Axon (advisory board / consultant), personal fees from Biogen (advisory board / consultant), personal fees from Julius Clinical (data monitoring committee), personal fees from Lilly (advisory board / consultant), personal fees from MagQu (advisory board / consultant), personal fees from Novartis (data monitoring committee), personal fees from Roche Diagnostic (advisory board / consultant), personal fees from Siemens Healthineers (advisory board / consultant), outside the submitted work; and is co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program.; CM reports personal fees from Biogen (sit on steering committee for aducanumab fees for time spent), personal fees from Roche (advisory board member fees for time spent), personal fees from Washington University (sit on therapeutic evalutation committee fees for time spent), outside the submitted work.; BS reports grants from UKRI/DHSC Global Effort on COVID-19 Research (Medical Research Council), non-financial support from UK National Institute for Health Research Global Health Research Group on Brain Infections, outside the submitted work.; MSZ reports receiving personal fees from UCB, for lecturing, outside the submitted work.; DJW reports personal fees from Bayer, personal fees from Alnylam, personal fees from Portola, outside the submitted work.; TS is on the Data Safety Monitoring Committee of Study to Evaluate the Safety and Immunogenicity of a Candidate Ebola Vaccine in Children GSK3390107A (ChAd3 EBO-Z) vaccine from GSK, is a panel member of Covid-19 Vaccine Benefit Risk Expert Working Group, which assesses the benefits and risks of Covid-19 vaccines at the Medicines and Healthcare Regulatory Agency (UK), is a member of COVID-19 Therapeutics Advisory Panel at the United Kingdom Department of Health & Social Care, and is Chair/Co-Chair of theCOVID-19 Rapid Response and Rolling Funding Initiatives, which supported development of the Oxford-Astra Zeneca Covid-19 vaccine, at the National Institute for Health Research, outside the submitted work. In addition, TS has a patent Test for bacterial meningitis based on a blood test, filed for patent pending.; HZ reports personal fees from Wave (Advisory board / consultant), personal fees from Roche Diagnostics (Advisory board / consultant), personal fees from Biogen (Sponsored lecture), personal fees (Advisory board / consultant) from Samumed, Eisai, Denali, Nervgen, Roche Diagnostics, Siemens Healthineers, Pinteon Therapeutics, AZTherapies, CogRx, personal fees (Sponsored lecture) from Alzecure, Cellectricon, and Fujirebio, outside the submitted work; and is Co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program.; JMS reports work support from National Institute for Health Research University College London Hospitals Biomedical Research Centre, during the conduct of the study; personal fees from Roche Pharmaceuticals (consulting), personal fees from Eli Lilly (consulting, lecturing), Drug Safety Monitoring Board at Axon Neuroscience, non-financial support from AVID Radiopharmaceuticals (PET tracer provision), consulting on educational activities from Biogen, personal fees from Merck (consulting), royalties from Oxford University Press, royalties from Henry Stewart Talks, grants from Wolfson Foundation, grants from Engineering and Physical Sciences Research Council (EP/J020990/1), grants from Medical Research Council Dementias Platform UK (MR/L023784/1), grants from Alzheimer's Research UK (ARUK-Network 2012-6-ICE; ARUK-PG2017-1946; ARUK-PG2017-1946), grants from Brain Research UK (UCC14191), grants from Weston Brain Institute (UB170045), grants from European Union's Horizon 2020 research and innovation programme (Grant 666,992), grants from British Heart Foundation (PG/17/90/33,415), personal fees from Alzheimer's Research UK (Chief Medical Officer), personal fees from UK Dementia Research Institute (Medical Advisor), outside the submitted work.; HC reports institutional research support and support to attend scientific meetings, and honoraria for lectures paid to UCLH charities from Bayer Healthcare, and consultancy fees paid to University College London Hospitals Charity from UCB Biopharma, outside the submitted work.; All other authors have nothing to disclose., (© 2021 The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
134. Grip strength from midlife as an indicator of later-life brain health and cognition: evidence from a British birth cohort.
- Author
-
Dercon Q, Nicholas JM, James SN, Schott JM, and Richards M
- Subjects
- Aged, Aging, Cohort Studies, Hand Strength, Humans, Brain diagnostic imaging, Cognition
- Abstract
Background: Grip strength is an indicator of physical function with potential predictive value for health in ageing populations. We assessed whether trends in grip strength from midlife predicted later-life brain health and cognition., Methods: 446 participants in an ongoing British birth cohort study, the National Survey of Health and Development (NSHD), had their maximum grip strength measured at ages 53, 60-64, and 69, and subsequently underwent neuroimaging as part of a neuroscience sub-study, referred to as "Insight 46", at age 69-71. A group-based trajectory model identified latent groups of individuals in the whole NSHD cohort with below- or above-average grip strength over time, plus a reference group. Group assignment, plus standardised grip strength levels and change from midlife were each related to measures of whole-brain volume (WBV) and white matter hyperintensity volume (WMHV), plus several cognitive tests. Models were adjusted for sex, body size, head size (where appropriate), sociodemographics, and behavioural and vascular risk factors., Results: Lower grip strength from midlife was associated with smaller WBV and lower matrix reasoning scores at age 69-71, with findings consistent between analysis of individual time points and analysis of trajectory groups. There was little evidence of an association between grip strength and other cognitive test scores. Although greater declines in grip strength showed a weak association with higher WMHV at age 69-71, trends in the opposite direction were seen at individual time points with higher grip strength at ages 60-64, and 69 associated with higher WMHV., Conclusions: This study provides preliminary evidence that maximum grip strength may have value in predicting brain health. Future work should assess to what extent age-related declines in grip strength from midlife reflect concurrent changes in brain structure., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
135. Aducanumab: a new phase in therapeutic development for Alzheimer's disease?
- Author
-
Lalli G, Schott JM, Hardy J, and De Strooper B
- Subjects
- Amyloid beta-Peptides, Antibodies, Monoclonal, Humanized, Humans, Immunotherapy, Alzheimer Disease drug therapy
- Abstract
On 7 June
, the FDA approved aducanumab, the first new drug for Alzheimer's disease in almost 20 years-and notably, the first drug with a putative disease-modifying mechanism for the treatment of this devastating disorder, namely the removal of β-amyloid (or Aβ) plaques from the brain., (© 2021 The Authors. Published under the terms of the CC BY 4.0 license.)- Published
- 2021
- Full Text
- View/download PDF
136. Suspecting dementia: canaries, chameleons and zebras.
- Author
-
Johnson JCS, McWhirter L, Hardy CJD, Crutch SJ, Marshall CR, Mummery CJ, Rohrer JD, Rossor MN, Schott JM, Weil RS, Fox NC, and Warren JD
- Abstract
The early and accurate diagnosis of dementia is more important than ever before but remains challenging. Dementia is increasingly the business of neurologists and, with ageing populations worldwide, will become even more so in future. Here we outline a practical, symptom-led, bedside approach to suspecting dementia and its likely diagnosis, inspired by clinical experience and based on recognition of characteristic syndromic patterns. We show how clinical intuition reflects underlying signature profiles of brain involvement by the diseases that cause dementia and suggest next steps that can be taken to define the diagnosis. We propose 'canaries' that provide an early warning signal of emerging dementia and highlight the 'chameleons' that disguise or mimic this, as well as the 'zebras' that herald a rare (and sometimes curable) diagnostic opportunity., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
137. Automated quantitative MRI volumetry reports support diagnostic interpretation in dementia: a multi-rater, clinical accuracy study.
- Author
-
Pemberton HG, Goodkin O, Prados F, Das RK, Vos SB, Moggridge J, Coath W, Gordon E, Barrett R, Schmitt A, Whiteley-Jones H, Burd C, Wattjes MP, Haller S, Vernooij MW, Harper L, Fox NC, Paterson RW, Schott JM, Bisdas S, White M, Ourselin S, Thornton JS, Yousry TA, Cardoso MJ, and Barkhof F
- Subjects
- Aged, Aged, 80 and over, Atrophy, Gray Matter, Humans, Magnetic Resonance Imaging, Middle Aged, Alzheimer Disease diagnostic imaging, Frontotemporal Dementia diagnostic imaging
- Abstract
Objectives: We examined whether providing a quantitative report (QReport) of regional brain volumes improves radiologists' accuracy and confidence in detecting volume loss, and in differentiating Alzheimer's disease (AD) and frontotemporal dementia (FTD), compared with visual assessment alone., Methods: Our forced-choice multi-rater clinical accuracy study used MRI from 16 AD patients, 14 FTD patients, and 15 healthy controls; age range 52-81. Our QReport was presented to raters with regional grey matter volumes plotted as percentiles against data from a normative population (n = 461). Nine raters with varying radiological experience (3 each: consultants, registrars, 'non-clinical image analysts') assessed each case twice (with and without the QReport). Raters were blinded to clinical and demographic information; they classified scans as 'normal' or 'abnormal' and if 'abnormal' as 'AD' or 'FTD'., Results: The QReport improved sensitivity for detecting volume loss and AD across all raters combined (p = 0.015* and p = 0.002*, respectively). Only the consultant group's accuracy increased significantly when using the QReport (p = 0.02*). Overall, raters' agreement (Cohen's κ) with the 'gold standard' was not significantly affected by the QReport; only the consultant group improved significantly (κ
s 0.41➔0.55, p = 0.04*). Cronbach's alpha for interrater agreement improved from 0.886 to 0.925, corresponding to an improvement from 'good' to 'excellent'., Conclusion: Our QReport referencing single-subject results to normative data alongside visual assessment improved sensitivity, accuracy, and interrater agreement for detecting volume loss. The QReport was most effective in the consultants, suggesting that experience is needed to fully benefit from the additional information provided by quantitative analyses., Key Points: • The use of quantitative report alongside routine visual MRI assessment improves sensitivity and accuracy for detecting volume loss and AD vs visual assessment alone. • Consultant neuroradiologists' assessment accuracy and agreement (kappa scores) significantly improved with the use of quantitative atrophy reports. • First multi-rater radiological clinical evaluation of visual quantitative MRI atrophy report for use as a diagnostic aid in dementia.- Published
- 2021
- Full Text
- View/download PDF
138. When dementia is misdiagnosed.
- Author
-
Howard R and Schott JM
- Subjects
- Diagnosis, Differential, Diagnostic Errors, Humans, Dementia diagnosis
- Published
- 2021
- Full Text
- View/download PDF
139. Investigating the Relationship Between IGF-I, IGF-II, and IGFBP-3 Concentrations and Later-Life Cognition and Brain Volume.
- Author
-
Salzmann A, James SN, Williams DM, Richards M, Cadar D, Schott JM, Coath W, Sudre CH, Chaturvedi N, and Garfield V
- Subjects
- Age Factors, Aging physiology, Brain diagnostic imaging, Cohort Studies, Female, History, 20th Century, History, 21st Century, Humans, Insulin-Like Growth Factor Binding Protein 3 analysis, Insulin-Like Growth Factor I analysis, Insulin-Like Growth Factor II analysis, Male, Middle Aged, Organ Size, United Kingdom, Brain anatomy & histology, Cognition physiology, Insulin-Like Growth Factor Binding Protein 3 blood, Insulin-Like Growth Factor I metabolism, Insulin-Like Growth Factor II metabolism
- Abstract
Background: The insulin/insulin-like signaling (IIS) pathways, including insulin-like growth factors (IGFs), vary with age. However, their association with late-life cognition and neuroimaging parameters is not well characterized., Methods: Using data from the British 1946 birth cohort, we investigated associations of IGF-I, IGF-II and IGF binding protein 3 (IGFBP-3; measured at 53 and 60-64 years of age) with cognitive performance [word-learning test (WLT) and visual letter search (VLS) at 60-64 years and 69 years of age] and cognitive state [Addenbrooke's Cognitive Exam III (ACE-III) at 69-71 years of age], and in a proportion, quantified neuroimaging measures [whole brain volume (WBV), white matter hyperintensity volume (WMHV), hippocampal volume (HV)]. Regression models included adjustments for demographic, lifestyle, and health factors., Results: Higher IGF-I and IGF-II at 53 years of age was associated with higher ACE-III scores [ß 0.07 95% confidence interval (CI) (0.02, 0.12); scoreACE-III 89.48 (88.86, 90.1), respectively). IGF-II at 53 years of age was additionally associated with higher WLT scores [scoreWLT 20 (19.35, 20.65)]. IGFBP-3 at 60 to 64 years of age was associated with favorable VLS score at 60 to 64 and 69 years of age [ß 0.07 (0.01, 0.12); ß 0.07 (0.02, 0.12), respectively], higher memory and cognitive state at 69 years of age [ß 0.07 (0.01, 0.12); ß 0.07 (0.01, 0.13), respectively], and reduced WMHV [ß -0.1 (-0.21, -0.00)]. IGF-I/IGFBP-3 at 60 to 64 years of was associated with lower VLS scores at 69 years of age [ß -0.08 (-0.15, -0.02)]., Conclusions: Increased measure in IIS parameters (IGF-I, IGF-II, and IGFBP-3) relate to better cognitive state in later life. There were apparent associations with specific cognitive domains (IGF-II relating to memory; IGFBP-3 relating to memory, processing speed, and WMHV; and IGF-I/IGFBP-3 molar ratio related to slower processing speed). IGFs and IGFBP-3 are associated with favorable cognitive function outcomes., (© The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2021
- Full Text
- View/download PDF
140. Serum and cerebrospinal fluid biomarker profiles in acute SARS-CoV-2-associated neurological syndromes.
- Author
-
Paterson RW, Benjamin LA, Mehta PR, Brown RL, Athauda D, Ashton NJ, Leckey CA, Ziff OJ, Heaney J, Heslegrave AJ, Benedet AL, Blennow K, Checkley AM, Houlihan CF, Mummery CJ, Lunn MP, Manji H, Zandi MS, Keddie S, Chou M, Vinayan Changaradil D, Solomon T, Keshavan A, Barker S, Jäger HR, Carletti F, Simister R, Werring DJ, Spyer MJ, Nastouli E, Gauthier S, Rosa-Neto P, Zetterberg H, and Schott JM
- Abstract
Preliminary pathological and biomarker data suggest that SARS-CoV-2 infection can damage the nervous system. To understand what, where and how damage occurs, we collected serum and CSF from patients with COVID-19 and characterized neurological syndromes involving the PNS and CNS ( n = 34). We measured biomarkers of neuronal damage and neuroinflammation, and compared these with non-neurological control groups, which included patients with ( n = 94) and without ( n = 24) COVID-19. We detected increased concentrations of neurofilament light, a dynamic biomarker of neuronal damage, in the CSF of those with CNS inflammation (encephalitis and acute disseminated encephalomyelitis) [14 800 pg/ml (400, 32 400)], compared to those with encephalopathy [1410 pg/ml (756, 1446)], peripheral syndromes (Guillain-Barré syndrome) [740 pg/ml (507, 881)] and controls [872 pg/ml (654, 1200)]. Serum neurofilament light levels were elevated across patients hospitalized with COVID-19, irrespective of neurological manifestations. There was not the usual close correlation between CSF and serum neurofilament light, suggesting serum neurofilament light elevation in the non-neurological patients may reflect peripheral nerve damage in response to severe illness. We did not find significantly elevated levels of serum neurofilament light in community cases of COVID-19 arguing against significant neurological damage. Glial fibrillary acidic protein, a marker of astrocytic activation, was not elevated in the CSF or serum of any group, suggesting astrocytic activation is not a major mediator of neuronal damage in COVID-19., (© The Author(s) (2021). Published by Oxford University Press on behalf of the Guarantors of Brain.)
- Published
- 2021
- Full Text
- View/download PDF
141. KL∗VS heterozygosity reduces brain amyloid in asymptomatic at-risk APOE∗4 carriers.
- Author
-
Belloy ME, Eger SJ, Le Guen Y, Napolioni V, Deters KD, Yang HS, Scelsi MA, Porter T, James SN, Wong A, Schott JM, Sperling RA, Laws SM, Mormino EC, He Z, Han SS, Altmann A, and Greicius MD
- Subjects
- Aged, Aged, 80 and over, Alzheimer Disease diagnostic imaging, Female, Humans, Klotho Proteins, Male, Middle Aged, Positron-Emission Tomography, Risk, Alzheimer Disease genetics, Amyloidogenic Proteins metabolism, Apolipoprotein E4 genetics, Brain metabolism, Carrier State, Glucuronidase genetics, Heterozygote
- Abstract
KLOTHO∗VS heterozygosity (KL∗VS
HET+ ) was recently shown to be associated with reduced risk of Alzheimer's disease (AD) in APOE∗4 carriers. Additional studies suggest that KL∗VSHET+ protects against amyloid burden in cognitively normal older subjects, but sample sizes were too small to draw definitive conclusions. We performed a well-powered meta-analysis across 5 independent studies, comprising 3581 pre-clinical participants ages 60-80, to investigate whether KL∗VSHET+ reduces the risk of having an amyloid-positive positron emission tomography scan. Analyses were stratified by APOE∗4 status. KL∗VSHET+ reduced the risk of amyloid positivity in APOE∗4 carriers (odds ratio = 0.67 [0.52-0.88]; p = 3.5 × 10-3 ), but not in APOE∗4 non-carriers (odds ratio = 0.94 [0.73-1.21]; p = 0.63). The combination of APOE∗4 and KL∗VS genotypes should help enrich AD clinical trials for pre-symptomatic subjects at increased risk of developing amyloid aggregation and AD. KL-related pathways may help elucidate protective mechanisms against amyloid accumulation and merit exploration for novel AD drug targets. Future investigation of the biological mechanisms by which KL interacts with APOE∗4 and AD are warranted., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
142. Investigating the relationship between BMI across adulthood and late life brain pathologies.
- Author
-
Lane CA, Barnes J, Nicholas JM, Baker JW, Sudre CH, Cash DM, Parker TD, Malone IB, Lu K, James SN, Keshavan A, Buchanan S, Keuss S, Murray-Smith H, Wong A, Gordon E, Coath W, Modat M, Thomas D, Hardy R, Richards M, Fox NC, and Schott JM
- Subjects
- Adult, Aged, Amyloid beta-Peptides metabolism, Body Mass Index, Brain diagnostic imaging, Brain metabolism, Cross-Sectional Studies, Humans, Middle Aged, Alzheimer Disease
- Abstract
Background: In view of reported associations between high adiposity, particularly in midlife and late-life dementia risk, we aimed to determine associations between body mass index (BMI), and BMI changes across adulthood and brain structure and pathology at age 69-71 years., Methods: Four hundred sixty-five dementia-free participants from Insight 46, a sub-study of the British 1946 birth cohort, who had cross-sectional T1/FLAIR volumetric MRI, and florbetapir amyloid-PET imaging at age 69-71 years, were included in analyses. We quantified white matter hyperintensity volume (WMHV) using T1 and FLAIR 3D-MRI; β-amyloid (Aβ) positivity/negativity using a SUVR approach; and whole brain (WBV) and hippocampal volumes (HV) using 3D T1-MRI. We investigated the influence of BMI, and BMI changes at and between 36, 43, 53, 60-64, 69 and 71 years, on late-life WMHV, Aβ-status, WBV and mean HV. Analyses were repeated using overweight and obese status., Results: At no time-point was BMI, change in BMI or overweight/obese status associated with WMHV or WBV at age 69-71 years. Decreasing BMI in the 1-2 years before imaging was associated with an increased odds of being β-amyloid positive (OR 1.45, 95% confidence interval 1.09, 1.92). There were associations between being overweight and larger mean HV at ages 60-64 (β = 0.073 ml, 95% CI 0.009, 0.137), 69 (β = 0.076 ml, 95% CI 0.012, 0.140) and 71 years (β = 0.101 ml, 95% CI 0.037, 0.165). A similar, albeit weaker, trend was seen with obese status., Conclusions: Using WMHV, β-amyloid status and brain volumes as indicators of brain health, we do not find evidence to explain reported associations between midlife obesity and late-life dementia risk. Declining BMI in later life may reflect preclinical Alzheimer's disease.
- Published
- 2021
- Full Text
- View/download PDF
143. A population-based study of head injury, cognitive function and pathological markers.
- Author
-
James SN, Nicholas JM, Lane CA, Parker TD, Lu K, Keshavan A, Buchanan SM, Keuss SE, Murray-Smith H, Wong A, Cash DM, Malone IB, Barnes J, Sudre CH, Coath W, Prosser L, Ourselin S, Modat M, Thomas DL, Cardoso J, Heslegrave A, Zetterberg H, Crutch SJ, Schott JM, Richards M, and Fox NC
- Subjects
- Aged, Cohort Studies, Female, Humans, Magnetic Resonance Imaging, Male, Positron-Emission Tomography, Aging physiology, Cognitive Dysfunction diagnosis, Cognitive Dysfunction etiology, Cognitive Dysfunction pathology, Cognitive Dysfunction physiopathology, Craniocerebral Trauma complications, Craniocerebral Trauma pathology, Craniocerebral Trauma physiopathology, Unconsciousness etiology
- Abstract
Objective: To assess associations between head injury (HI) with loss of consciousness (LOC), ageing and markers of later-life cerebral pathology; and to explore whether those effects may help explain subtle cognitive deficits in dementia-free individuals., Methods: Participants (n = 502, age = 69-71) from the 1946 British Birth Cohort underwent cognitive testing (subtests of Preclinical Alzheimer Cognitive Composite),
18 F-florbetapir Aβ-PET and MR imaging. Measures include Aβ-PET status, brain, hippocampal and white matter hyperintensity (WMH) volumes, normal appearing white matter (NAWM) microstructure, Alzheimer's disease (AD)-related cortical thickness, and serum neurofilament light chain (NFL). LOC HI metrics include HI occurring: (i) >15 years prior to the scan (ii) anytime up to age 71., Results: Compared to those with no evidence of an LOC HI, only those reporting an LOC HI>15 years prior (16%, n = 80) performed worse on cognitive tests at age 69-71, taking into account premorbid cognition, particularly on the digit-symbol substitution test (DSST). Smaller brain volume (BV) and adverse NAWM microstructural integrity explained 30% and 16% of the relationship between HI and DSST, respectively. We found no evidence that LOC HI was associated with Aβ load, hippocampal volume, WMH volume, AD-related cortical thickness or NFL (all p > 0.01)., Interpretation: Having a LOC HI aged 50's and younger was linked with lower later-life cognitive function at age ~70 than expected. This may reflect a damaging but small impact of HI; explained in part by smaller BV and different microstructure pathways but not via pathology related to AD (amyloid, hippocampal volume, AD cortical thickness) or ongoing neurodegeneration (serum NFL)., (© 2021 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association.)- Published
- 2021
- Full Text
- View/download PDF
144. Population-based blood screening for preclinical Alzheimer's disease in a British birth cohort at age 70.
- Author
-
Keshavan A, Pannee J, Karikari TK, Rodriguez JL, Ashton NJ, Nicholas JM, Cash DM, Coath W, Lane CA, Parker TD, Lu K, Buchanan SM, Keuss SE, James SN, Murray-Smith H, Wong A, Barnes A, Dickson JC, Heslegrave A, Portelius E, Richards M, Fox NC, Zetterberg H, Blennow K, and Schott JM
- Subjects
- Aged, Alzheimer Disease metabolism, Biomarkers blood, Early Diagnosis, Female, Hematologic Tests methods, Humans, Male, Prospective Studies, Sensitivity and Specificity, Alzheimer Disease blood, Alzheimer Disease diagnosis, Amyloid beta-Peptides blood, Peptide Fragments blood
- Abstract
Alzheimer's disease has a preclinical stage when cerebral amyloid-β deposition occurs before symptoms emerge, and when amyloid-β-targeted therapies may have maximum benefits. Existing amyloid-β status measurement techniques, including amyloid PET and CSF testing, are difficult to deploy at scale, so blood biomarkers are increasingly considered for screening. We compared three different blood-based techniques-liquid chromatography-mass spectrometry measures of plasma amyloid-β, and single molecule array (Simoa) measures of plasma amyloid-β and phospho-tau181-to detect cortical 18F-florbetapir amyloid PET positivity (defined as a standardized uptake value ratio of >0.61 between a predefined cortical region of interest and eroded subcortical white matter) in dementia-free members of Insight 46, a substudy of the population-based British 1946 birth cohort. We used logistic regression models with blood biomarkers as predictors of amyloid PET status, with or without age, sex and APOE ε4 carrier status as covariates. We generated receiver operating characteristics curves and quantified areas under the curves to compare the concordance of the different blood tests with amyloid PET. We determined blood test cut-off points using Youden's index, then estimated numbers needed to screen to obtain 100 amyloid PET-positive individuals. Of the 502 individuals assessed, 441 dementia-free individuals with complete data were included; 82 (18.6%) were amyloid PET-positive. The area under the curve for amyloid PET status using a base model comprising age, sex and APOE ε4 carrier status was 0.695 (95% confidence interval: 0.628-0.762). The two best-performing Simoa plasma biomarkers were amyloid-β42/40 (0.620; 0.548-0.691) and phospho-tau181 (0.707; 0.646-0.768), but neither outperformed the base model. Mass spectrometry plasma measures performed significantly better than any other measure (amyloid-β1-42/1-40: 0.817; 0.770-0.864 and amyloid-β composite: 0.820; 0.775-0.866). At a cut-off point of 0.095, mass spectrometry measures of amyloid-β1-42/1-40 detected amyloid PET positivity with 86.6% sensitivity and 71.9% specificity. Without screening, to obtain 100 PET-positive individuals from a population with similar amyloid PET positivity prevalence to Insight 46, 543 PET scans would need to be performed. Screening using age, sex and APOE ε4 status would require 940 individuals, of whom 266 would proceed to scan. Using mass spectrometry amyloid-β1-42/1-40 alone would reduce these numbers to 623 individuals and 243 individuals, respectively. Across a theoretical range of amyloid PET positivity prevalence of 10-50%, mass spectrometry measures of amyloid-β1-42/1-40 would consistently reduce the numbers proceeding to scans, with greater cost savings demonstrated at lower prevalence., (© The Author(s) (2021). Published by Oxford University Press on behalf of the Guarantors of Brain.)
- Published
- 2021
- Full Text
- View/download PDF
145. A Clinicopathologic Study of Movement Disorders in Frontotemporal Lobar Degeneration.
- Author
-
de Pablo-Fernández E, González-Herrero B, Cerdán Santacruz D, Rossor MN, Schott JM, Lashley T, Holton JL, Fox NC, Revesz T, Warren JD, Jaunmuktane Z, Rohrer JD, and Warner TT
- Subjects
- Cohort Studies, Humans, Male, Retrospective Studies, Frontotemporal Dementia, Frontotemporal Lobar Degeneration, Movement Disorders etiology
- Abstract
Background: Despite the considerable overlap with atypical parkinsonism, a systematic characterization of the movement disorders associated with frontotemporal lobar degeneration (FTLD) is lacking., Objective: The aim of this study is to provide a detailed description of the phenomenology and neuropathologic correlations of movement disorders in FTLD., Methods: In this cohort study, movement disorder clinical data were retrospectively collected from medical records of consecutive patients with a postmortem diagnosis of FTLD from the Queen Square Brain Bank between January 2010 and December 2018. At postmortem, neurodegenerative pathologies were systematically evaluated following consensus criteria. Degeneration of the substantia nigra was assessed as a marker of presynaptic dopaminergic parkinsonism using semiquantitative methods., Results: A total of 55 patients (35 men [64%]) were included with median (interquartile range) age at diagnosis of 58.8 (52.6-63.9) years and a disease duration of 9.6 (6.2-12.9) years. Movement disorders were present in 19 (35%) patients without differences among disease subtypes. The most common syndromes were parkinsonism (9 patients [16%]), usually as an additional late feature, and corticobasal syndrome (CBS, 7 patients [13%]), commonly as a presenting feature. Substantia nigra degeneration was present in 37 (67%) patients although it did not show a good clinical correlation with movement disorders. Those with Pick's disease showed milder substantia nigra degeneration and better response to levodopa., Conclusions: Movement disorders can present in all FTLD subtypes, more commonly as a late additional feature (parkinsonism) or as a presenting symptom (CBS). The underlying pathophysiology is complex and likely to involve structures outside the presynaptic striatonigral system. © 2020 International Parkinson and Movement Disorder Society., (© 2020 International Parkinson and Movement Disorder Society.)
- Published
- 2021
- Full Text
- View/download PDF
146. New insights into atypical Alzheimer's disease in the era of biomarkers.
- Author
-
Graff-Radford J, Yong KXX, Apostolova LG, Bouwman FH, Carrillo M, Dickerson BC, Rabinovici GD, Schott JM, Jones DT, and Murray ME
- Subjects
- Age of Onset, Aged, Aged, 80 and over, Alzheimer Disease classification, Alzheimer Disease diagnostic imaging, Alzheimer Disease epidemiology, Female, Humans, Male, Middle Aged, Neuroimaging methods, Alzheimer Disease diagnosis, Biomarkers
- Abstract
Most patients with Alzheimer's disease present with amnestic problems; however, a substantial proportion, over-represented in young-onset cases, have atypical phenotypes including predominant visual, language, executive, behavioural, or motor dysfunction. In the past, these individuals often received a late diagnosis; however, availability of CSF and PET biomarkers of Alzheimer's disease pathologies and incorporation of atypical forms of Alzheimer's disease into new diagnostic criteria increasingly allows them to be more confidently diagnosed early in their illness. This early diagnosis in turn allows patients to be offered tailored information, appropriate care and support, and individualised treatment plans. These advances will provide improved access to clinical trials, which often exclude atypical phenotypes. Research into atypical Alzheimer's disease has revealed previously unrecognised neuropathological heterogeneity across the Alzheimer's disease spectrum. Neuroimaging, genetic, biomarker, and basic science studies are providing key insights into the factors that might drive selective vulnerability of differing brain networks, with potential mechanistic implications for understanding typical late-onset Alzheimer's disease., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
147. Visuomotor integration deficits are common to familial and sporadic preclinical Alzheimer's disease.
- Author
-
Lu K, Nicholas JM, Weston PSJ, Stout JC, O'Regan AM, James SN, Buchanan SM, Lane CA, Parker TD, Keuss SE, Keshavan A, Murray-Smith H, Cash DM, Sudre CH, Malone IB, Coath W, Wong A, Richards M, Henley SMD, Fox NC, Schott JM, and Crutch SJ
- Abstract
We investigated whether subtle visuomotor deficits were detectable in familial and sporadic preclinical Alzheimer's disease. A circle-tracing task-with direct and indirect visual feedback, and dual-task subtraction-was completed by 31 individuals at 50% risk of familial Alzheimer's disease (19 presymptomatic mutation carriers; 12 non-carriers) and 390 cognitively normal older adults (members of the British 1946 Birth Cohort, all born during the same week; age range at assessment = 69-71 years), who also underwent β-amyloid-PET/MRI to derive amyloid status (positive/negative), whole-brain volume and white matter hyperintensity volume. We compared preclinical Alzheimer's groups against controls cross-sectionally (mutation carriers versus non-carriers; amyloid-positive versus amyloid-negative) on speed and accuracy of circle-tracing and subtraction. Mutation carriers (mean 7 years before expected onset) and amyloid-positive older adults traced disproportionately less accurately than controls when visual feedback was indirect, and were slower at dual-task subtraction. In the older adults, the same pattern of associations was found when considering amyloid burden as a continuous variable (Standardized Uptake Value Ratio). The effect of amyloid was independent of white matter hyperintensity and brain volumes, which themselves were associated with different aspects of performance: greater white matter hyperintensity volume was also associated with disproportionately poorer tracing accuracy when visual feedback was indirect, whereas larger brain volume was associated with faster tracing and faster subtraction. Mutation carriers also showed evidence of poorer tracing accuracy when visual feedback was direct. This study provides the first evidence of visuomotor integration deficits common to familial and sporadic preclinical Alzheimer's disease, which may precede the onset of clinical symptoms by several years., (© The Author(s) (2021). Published by Oxford University Press on behalf of the Guarantors of Brain.)
- Published
- 2021
- Full Text
- View/download PDF
148. Mild cognitive impairment: the Manchester consensus.
- Author
-
Dunne RA, Aarsland D, O'Brien JT, Ballard C, Banerjee S, Fox NC, Isaacs JD, Underwood BR, Perry RJ, Chan D, Dening T, Thomas AJ, Schryer J, Jones AM, Evans AR, Alessi C, Coulthard EJ, Pickett J, Elton P, Jones RW, Mitchell S, Hooper N, Kalafatis C, Rasmussen JGC, Martin H, Schott JM, and Burns A
- Subjects
- Activities of Daily Living, Amyloid beta-Peptides, Biomarkers, Consensus, Disease Progression, Humans, Neuropsychological Tests, Peptide Fragments, State Medicine, Alzheimer Disease, Cognitive Dysfunction diagnosis, Cognitive Dysfunction epidemiology, Neurodegenerative Diseases
- Abstract
Given considerable variation in diagnostic and therapeutic practice, there is a need for national guidance on the use of neuroimaging, fluid biomarkers, cognitive testing, follow-up and diagnostic terminology in mild cognitive impairment (MCI). MCI is a heterogenous clinical syndrome reflecting a change in cognitive function and deficits on neuropsychological testing but relatively intact activities of daily living. MCI is a risk state for further cognitive and functional decline with 5-15% of people developing dementia per year. However, ~50% remain stable at 5 years and in a minority, symptoms resolve over time. There is considerable debate about whether MCI is a useful clinical diagnosis, or whether the use of the term prevents proper inquiry (by history, examination and investigations) into underlying causes of cognitive symptoms, which can include prodromal neurodegenerative disease, other physical or psychiatric illness, or combinations thereof. Cognitive testing, neuroimaging and fluid biomarkers can improve the sensitivity and specificity of aetiological diagnosis, with growing evidence that these may also help guide prognosis. Diagnostic criteria allow for a diagnosis of Alzheimer's disease to be made where MCI is accompanied by appropriate biomarker changes, but in practice, such biomarkers are not available in routine clinical practice in the UK. This would change if disease-modifying therapies became available and required a definitive diagnosis but would present major challenges to the National Health Service and similar health systems. Significantly increased investment would be required in training, infrastructure and provision of fluid biomarkers and neuroimaging. Statistical techniques combining markers may provide greater sensitivity and specificity than any single disease marker but their practical usefulness will depend on large-scale studies to ensure ecological validity and that multiple measures, e.g. both cognitive tests and biomarkers, are widely available for clinical use. To perform such large studies, we must increase research participation amongst those with MCI., (© The Author(s) 2020. Published by Oxford University Press on behalf of the British Geriatrics Society.)
- Published
- 2021
- Full Text
- View/download PDF
149. Genetic testing in dementia - utility and clinical strategies.
- Author
-
Koriath CAM, Kenny J, Ryan NS, Rohrer JD, Schott JM, Houlden H, Fox NC, Tabrizi SJ, and Mead S
- Subjects
- Age of Onset, Dementia diagnosis, Dementia therapy, High-Throughput Nucleotide Sequencing, Humans, Mutation, Phenotype, Dementia genetics, Genetic Testing
- Abstract
Techniques for clinical genetic testing in dementia disorders have advanced rapidly but remain to be more widely implemented in practice. A positive genetic test offers a precise molecular diagnosis, can help members of an affected family to determine personal risk, provides a basis for reproductive choices and can offer options for clinical trials. The likelihood of identifying a specific genetic cause of dementia depends on the clinical condition, the age at onset and family history. Attempts to match phenotypes to single genes are mostly inadvisable owing to clinical overlap between the dementias, genetic heterogeneity, pleiotropy and concurrent mutations. Currently, the appropriate genetic test in most cases of dementia is a next-generation sequencing gene panel, though some conditions necessitate specific types of test such as repeat expansion testing. Whole-exome and whole-genome sequencing are becoming financially feasible but raise or exacerbate complex issues such as variants of uncertain significance, secondary findings and the potential for re-analysis in light of new information. However, the capacity for data analysis and counselling is already restricting the provision of genetic testing. Patients and their relatives need to be given reliable information to enable them to make informed choices about tests, treatments and data sharing; the ability of patients with dementia to make decisions must be considered when providing this information.
- Published
- 2021
- Full Text
- View/download PDF
150. Predictors for a dementia gene mutation based on gene-panel next-generation sequencing of a large dementia referral series.
- Author
-
Koriath C, Kenny J, Adamson G, Druyeh R, Taylor W, Beck J, Quinn L, Mok TH, Dimitriadis A, Norsworthy P, Bass N, Carter J, Walker Z, Kipps C, Coulthard E, Polke JM, Bernal-Quiros M, Denning N, Thomas R, Raybould R, Williams J, Mummery CJ, Wild EJ, Houlden H, Tabrizi SJ, Rossor MN, Hummerich H, Warren JD, Rowe JB, Rohrer JD, Schott JM, Fox NC, Collinge J, and Mead S
- Subjects
- Aged, Genomics, Humans, Mutation genetics, Referral and Consultation, Dementia genetics, High-Throughput Nucleotide Sequencing
- Abstract
Next-generation genetic sequencing (NGS) technologies facilitate the screening of multiple genes linked to neurodegenerative dementia, but there are few reports about their use in clinical practice. Which patients would most profit from testing, and information on the likelihood of discovery of a causal variant in a clinical syndrome, are conspicuously absent from the literature, mostly for a lack of large-scale studies. We applied a validated NGS dementia panel to 3241 patients with dementia and healthy aged controls; 13,152 variants were classified by likelihood of pathogenicity. We identified 354 deleterious variants (DV, 12.6% of patients); 39 were novel DVs. Age at clinical onset, clinical syndrome and family history each strongly predict the likelihood of finding a DV, but healthcare setting and gender did not. DVs were frequently found in genes not usually associated with the clinical syndrome. Patients recruited from primary referral centres were compared with those seen at higher-level research centres and a national clinical neurogenetic laboratory; rates of discovery were comparable, making selection bias unlikely and the results generalisable to clinical practice. We estimated penetrance of DVs using large-scale online genomic population databases and found 71 with evidence of reduced penetrance. Two DVs in the same patient were found more frequently than expected. These data should provide a basis for more informed counselling and clinical decision making.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.