1
|
Rajicic A, Giannini LAA, Gerrits E, van Buuren R, Melhem S, Slotman JA, Rozemuller AJM, Eggen BJL, van Swieten JC, Seelaar H. WDR49-Positive Astrocytes Mark Severity of Neurodegeneration in Frontotemporal Lobar Degeneration and Alzheimer's Disease. Glia 2025; 73:948-968. [PMID: 39705191 PMCID: PMC11920684 DOI: 10.1002/glia.24663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 12/22/2024]
Abstract
A subpopulation of astrocytes expressing WD Repeat Domain 49 (WDR49) was recently identified in frontotemporal lobar degeneration (FTLD) with GRN pathogenic variants. This is the first study to investigate their expression and relation to pathology in other FTLD subtypes and Alzheimer's disease (AD). In a postmortem cohort of TDP-43 proteinopathies (12 GRN, 11 C9orf72, 9 sporadic TDP-43), tauopathies (13 MAPT, 8 sporadic tau), 10 AD, and four controls, immunohistochemistry and immunofluorescence were performed for WDR49 and pathological inclusions on frontal, temporal, and occipital cortical sections. WDR49-positive cell counts (adjusted per mm2) were examined and related to digitally quantified percentage areas of TDP-43/tau pathology and semiquantitative scores of neurodegeneration. Quantitative colocalization analysis of WDR49 and pathological inclusions was done. WDR49-positive astrocytes were present across FTLD subtypes and AD in the brain parenchyma and (peri-)vascular space, with distinct morphological patterns, and were particularly enriched in gray matter. In controls, sporadic WDR49-positive cells were found enveloping vessels. WDR49-positive astrocytes were most abundant in the frontal cortex (FC) of GRN cases and temporal cortex in GRN, AD, and sporadic primary tauopathy. In the occipital cortex, only a few cells were found across groups. WDR49-positive astrocyte counts positively correlated with the severity of neurodegeneration and TDP-43 pathology but not tauopathy. Furthermore, in frontotemporal cortices, WDR49 partly colocalized with TDP-43 (14%-21%) and tau (31%-45%). In conclusion, WDR49 is a marker for a subset of astrocytes with different morphologies across FTLD and AD, reflecting the severity of neurodegeneration. These astrocytes may become activated in neurodegeneration in response to pathological damage and migrate from the vessel wall to the parenchyma.
Collapse
Affiliation(s)
- Ana Rajicic
- Department of Neurology and Alzheimer Centre Erasmus MCErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Lucia A. A. Giannini
- Department of Neurology and Alzheimer Centre Erasmus MCErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Emma Gerrits
- Department of NeuroscienceKarolinska InstitutetStockholmSweden
| | - Renee van Buuren
- Department of Neurology and Alzheimer Centre Erasmus MCErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Shamiram Melhem
- Department of Neurology and Alzheimer Centre Erasmus MCErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Johan A. Slotman
- Department of Pathology and Erasmus Optical Imaging CenterErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Annemieke J. M. Rozemuller
- Department of Pathology, Amsterdam NeuroscienceAmsterdam University Medical CenterAmsterdamThe Netherlands
| | - Bart J. L. Eggen
- Department of Biomedical Sciences of Cells & Systems, Section of Molecular NeurobiologyUniversity of Groningen and University Medical Center GroningenGroningenNetherlands
| | - John C. van Swieten
- Department of Neurology and Alzheimer Centre Erasmus MCErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Harro Seelaar
- Department of Neurology and Alzheimer Centre Erasmus MCErasmus MC University Medical CenterRotterdamthe Netherlands
| |
Collapse
|
2
|
Reijner N, Frigerio I, Bouwman MMA, Boon BDC, Guizard N, Jubault T, Hoozemans JJM, Rozemuller AJM, Bouwman FH, Barkhof F, Gordon E, van de Berg WDJ, Jonkman LE. Clinical phenotypes of Alzheimer's disease: investigating atrophy patterns and their pathological correlates. Alzheimers Res Ther 2025; 17:93. [PMID: 40281562 PMCID: PMC12032798 DOI: 10.1186/s13195-025-01727-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND In Alzheimer's disease (AD), MRI atrophy patterns can distinguish between amnestic (typical) and non-amnestic (atypical) clinical phenotypes and are increasingly used for diagnosis and outcome measures in clinical trials. However, understanding how protein accumulation and other key features of neurodegeneration influence these imaging measurements, are lacking. The current study aimed to assess regional MRI patterns of cortical atrophy across clinical AD phenotypes, and their association with amyloid-beta (Aβ), phosphorylated tau (pTau), neuro-axonal degeneration and microvascular deterioration. METHODS Post-mortem in-situ 3DT1 3 T-MRI data was obtained from 33 AD (17 typical, 16 atypical) and 16 control brain donors. Additionally, ante-mortem 3DT1 3 T-MRI scans of brain donors were collected if available. Regional volumes were obtained from MRI scans using an atlas based parcellation software. Eight cortical brain regions were selected from formalin-fixed right hemispheres of brain donors and then immunostained for Aβ, pTau, neurofilament light, and collagen IV. Group comparisons and volume-pathology associations were analyzed using linear mixed models corrected for age, sex, post-mortem delay, and intracranial volume. RESULTS Compared to controls, both typical and atypical AD showed volume loss in the temporo-occipital cortex, while typical AD showed additional volume loss in the parietal cortex. Posterior cingulate volume was lower in typical AD compared to atypical AD (- 6.9%, p = 0.043). In AD, a global positive association between MRI cortical volume and Aβ load (βs = 0.21, p = 0.010), and a global negative association with NfL load (βs = - 0.18, p = 0.018) were observed. Regionally, higher superior parietal gyrus volume was associated with higher Aβ load in typical AD (βs = 0.47, p = 0.004), lower middle frontal gyrus volume associated with higher NfL load in atypical AD (βs = - 0.50, p < 0.001), and lower hippocampal volume associated with higher COLIV load in typical AD (βs = - 1.69, p < 0.001). Comparing post-mortem with ante-mortem scans showed minimal volume differences at scan-intervals within 2 years, highlighting the translational aspect of this study. CONCLUSION For both clinical phenotypes, cortical volume is affected by Aβ and neuro-axonal damage, but in opposing directions. Differences in volume-pathology relationships between clinical phenotypes are region-specific. The findings of this study could improve the interpretation of MRI datasets in heterogenous AD cohorts, both in research and clinical settings.
Collapse
Affiliation(s)
- Niels Reijner
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam, 1081 HZ, Netherlands.
- Programs of Neurodegeneration, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands.
- Programs of Brain Imaging, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands.
| | - I Frigerio
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam, 1081 HZ, Netherlands
- Programs of Neurodegeneration, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
- Programs of Brain Imaging, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
| | - M M A Bouwman
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam, 1081 HZ, Netherlands
- Programs of Neurodegeneration, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
- Programs of Brain Imaging, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
| | - B D C Boon
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, USA
| | - N Guizard
- Qynapse, 2 - 10 Rue d'Oradour-Sur-Glane, Paris, 75015, France
| | - T Jubault
- Qynapse, 2 - 10 Rue d'Oradour-Sur-Glane, Paris, 75015, France
| | - J J M Hoozemans
- Programs of Neurodegeneration, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - A J M Rozemuller
- Programs of Neurodegeneration, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - F H Bouwman
- Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - F Barkhof
- Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Institutes of Neurology and Healthcare Engineering, University College London, Gower Street, London, UK
| | - E Gordon
- Qynapse, 2 - 10 Rue d'Oradour-Sur-Glane, Paris, 75015, France
| | - W D J van de Berg
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam, 1081 HZ, Netherlands
- Programs of Neurodegeneration, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
| | - L E Jonkman
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, Amsterdam, 1081 HZ, Netherlands
- Programs of Neurodegeneration, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
- Programs of Brain Imaging, Amsterdam Neuroscience, Boelelaan 1117, Amsterdam, Netherlands
| |
Collapse
|
3
|
Rash BG, Ramdas KN, Agafonova N, Naioti E, McClain-Moss L, Zainul Z, Varnado B, Peterson K, Brown M, Leal T, Kopcho S, Carballosa R, Patel P, Brody M, Herskowitz B, Fuquay A, Rodriguez S, Jacobson AF, Leon R, Pfeffer M, Schwartzbard JB, Botbyl J, Oliva AA, Hare JM. Allogeneic mesenchymal stem cell therapy with laromestrocel in mild Alzheimer's disease: a randomized controlled phase 2a trial. Nat Med 2025; 31:1257-1266. [PMID: 40065171 PMCID: PMC12003194 DOI: 10.1038/s41591-025-03559-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 01/29/2025] [Indexed: 04/18/2025]
Abstract
Alzheimer's disease (AD) is characterized by progressive cognitive decline, severe brain atrophy and neuroinflammation. We conducted a randomized, double-blind, placebo-controlled, parallel-group phase 2a clinical trial that tested the safety and efficacy of laromestrocel, a bone-marrow-derived, allogeneic mesenchymal stem-cell therapy, in slowing AD clinical progression, atrophy and neuroinflammation. Participants across ten centers in the United States were randomly assigned 1:1:1:1 to four infusion groups: group 1 (placebo; four monthly infusions, n = 12); group 2 (25 million cells, one infusion followed by three monthly infusions of placebo, n = 13); group 3 (25 million cells; four monthly doses, n = 13); and group 4 (100 million cells; four monthly doses, n = 11). The study met its primary end point of safety; the rate of treatment-emergent serious adverse events within 4 weeks of any infusion was similar in all four groups: group 1, 0% (95% CI 0-26.5%); group 2, 7.7% (95% CI 0.2-36%); group 3, 7.7% (95% CI 0.2-36%) and group 4, 9.1% (95% CI 0.2-41.3%). Additionally, there were no reported infusion-related reactions, hypersensitivities or amyloid-related imaging abnormalities. Laromestrocel improved clinical assessments at 39 weeks compared to placebo, as measured by a composite AD score (secondary end point was met: group 2 versus placebo change: 0.38; 95% CI -0.06-0.82), Montreal cognitive assessment and the Alzheimer's Disease Cooperative Study Activities of Daily Living. At 39 weeks, Laromestrocel slowed the decline of whole brain volume compared to placebo (n = 10) by 48.4% for all treatment groups combined (groups 2-4: P = 0.005; n = 32) and left hippocampal volume by 61.9% (groups 2-4, P = 0.021; n = 32), and reduced neuroinflammation as measured by diffusion tensor imaging. The change in bilateral hippocampal atrophy correlated with the change in mini-mental state exam scores (R = 0.41, P = 0.0075) in all study patients (N = 42). Collectively these results support safety of single and multiple doses of laromestrocel treatment for mild AD and provide indications of efficacy in combating decline of brain volume and potentially cognitive function. Larger-scale clinical trials of laromestrocel in AD are warranted. ClinicalTrials.gov registration: NCT05233774 .
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Mark Brody
- Brain Matters Research, Delray Beach, FL, USA
| | | | - Ana Fuquay
- Brain Matters Research, Delray Beach, FL, USA
| | | | - Alan F Jacobson
- Allied Clinical Trials, Miami, FL, USA
- Fusion Medical & Research Clinic, Miami, FL, USA
| | | | | | | | | | | | - Joshua M Hare
- Longeveron, Miami, FL, USA.
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
4
|
Ren Y, Pieper AA, Cheng F. Utilization of precision medicine digital twins for drug discovery in Alzheimer's disease. Neurotherapeutics 2025; 22:e00553. [PMID: 39965994 PMCID: PMC12047495 DOI: 10.1016/j.neurot.2025.e00553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/11/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
Alzheimer's disease (AD) presents significant challenges in drug discovery and development due to its complex and poorly understood pathology and etiology. Digital twins (DTs) are recently developed virtual real-time representations of physical entities that enable rapid assessment of the bidirectional interaction between the virtual and physical domains. With recent advances in artificial intelligence (AI) and the growing accumulation of multi-omics and clinical data, application of DTs in healthcare is gaining traction. Digital twin technology, in the form of multiscale virtual models of patients or organ systems, can track health status in real time with continuous feedback, thereby driving model updates that enhance clinical decision-making. Here, we posit an additional role for DTs in drug discovery, with particular utility for complex diseases like AD. In this review, we discuss salient challenges in AD drug development, including complex disease pathology and comorbidities, difficulty in early diagnosis, and the current high failure rate of clinical trials. We also review DTs and discuss potential applications for predicting AD progression, discovering biomarkers, identifying new drug targets and opportunities for drug repurposing, facilitating clinical trials, and advancing precision medicine. Despite significant hurdles in this area, such as integration and standardization of dynamic medical data and issues of data security and privacy, DTs represent a promising approach for revolutionizing drug discovery in AD.
Collapse
Affiliation(s)
- Yunxiao Ren
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland 44106, OH, USA; Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA.
| |
Collapse
|
5
|
Baker LD, Pa JA, Katula JA, Aslanyan V, Salmon DP, Jacobs DM, Chmelo EA, Hodge H, Morrison R, Matthews G, Brewer J, Jung Y, Rissman RA, Taylor C, Léger GC, Messer K, Evans AC, Okonkwo OC, Shadyab AH, Zou J, Jin S, Thomas RG, Zhang J, La Croix AZ, Cotman CW, Feldman HH. Effects of exercise on cognition and Alzheimer's biomarkers in a randomized controlled trial of adults with mild cognitive impairment: The EXERT study. Alzheimers Dement 2025; 21:e14586. [PMID: 40271888 PMCID: PMC12019696 DOI: 10.1002/alz.14586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/04/2024] [Accepted: 01/12/2025] [Indexed: 04/25/2025]
Abstract
INTRODUCTION The EXERT study (Exercise in Adults with Mild Memory Problems) was a Phase 3, multicenter, randomized controlled trial that examined effects of exercise on cognition and other measures of brain health in sedentary older adults with amnestic mild cognitive impairment (MCI). METHODS Participants were randomized to moderate-high intensity aerobic training (AX) or low-intensity stretching/balance/range of motion (SBR) for 18 months. Exercise was supervised for the first 12 months. Assessments were administered at baseline and every 6 months. The primary outcome was a global cognitive composite. RESULTS A total of 296 participants were enrolled, and intervention adherence was high (supervised session attendance: AX = 81%, SBR = 87%). Intervention effects on cognition did not differ for AX and SBR (regression = -0.078, standard error [SE] = 0.074; p = 0.3). Notably, there was no 12 month cognition decline for either group, and mean 12 month hippocampal volume loss for both groups was low at 0.51%. DISCUSSION Exercise intensity did not differentially affect cognitive trajectory. Intervention delivery was successful (high adherence) and cognition remained stable over 12 months for both MCI groups, an association that warrants further study. HIGHLIGHTS Exercise in Adults with Mild Memory Problems (EXERT) was a large multisite randomized controlled trial of moderate-high intensity aerobic training versus lower-intensity flexibility and balance exercise in sedentary older adults with amnestic mild cognitive impairment (MCI). A sensitive and validated measure of global cognitive function, the Alzheimer's Disease Assessment Scale-Cognition supplemented with tests of executive function (ADAS-Cog-Exec), was used to assess intervention efficacy with 12 months of supervised exercise. There was no intervention group difference on the 12-month cognitive trajectory of the ADAS-Cog-Exec. Intervention delivery was successful (high adherence), and cognition remained stable over 12 months for both exercise groups. Regular supported moderate-high or lower-intensity exercise may stall decline in adults with amnestic MCI, but further investigation is needed.
Collapse
Affiliation(s)
- Laura D. Baker
- Department of Internal Medicine‐GeriatricsWake Forest University School of MedicineWinston SalemNorth CarolinaUSA
- Departments of Social Science & Health Policy, and Epidemiology, Division of Public Health SciencesWake Forest University Health SciencesWinston SalemNorth CarolinaUSA
| | - Judy A. Pa
- Alzheimer's Disease Cooperative StudyUniversity of CaliforniaLa JollaCaliforniaUSA
- Department of NeurosciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Jeffrey A. Katula
- Department of Health and Exercise ScienceWake Forest UniversityWinston SalemNorth CarolinaUSA
| | - Vahan Aslanyan
- Department of Population and Public Health Sciences, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - David P. Salmon
- Alzheimer's Disease Cooperative StudyUniversity of CaliforniaLa JollaCaliforniaUSA
- Department of NeurosciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Diane M. Jacobs
- Alzheimer's Disease Cooperative StudyUniversity of CaliforniaLa JollaCaliforniaUSA
- Department of NeurosciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Elizabeth A. Chmelo
- Department of Internal Medicine‐GeriatricsWake Forest University School of MedicineWinston SalemNorth CarolinaUSA
| | | | - Rosemary Morrison
- Alzheimer's Disease Cooperative StudyUniversity of CaliforniaLa JollaCaliforniaUSA
- Department of NeurosciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Genevieve Matthews
- Alzheimer's Disease Cooperative StudyUniversity of CaliforniaLa JollaCaliforniaUSA
- Department of NeurosciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - James Brewer
- Alzheimer's Disease Cooperative StudyUniversity of CaliforniaLa JollaCaliforniaUSA
- Department of NeurosciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Youngkyoo Jung
- Department of Radiology, Medical PhysicsUniversity of California Davis HealthDavisCaliforniaUSA
| | - Robert A. Rissman
- Department of Physiology and Neurosciences, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Curtis Taylor
- Alzheimer's Disease Cooperative StudyUniversity of CaliforniaLa JollaCaliforniaUSA
- Department of NeurosciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Gabriel C. Léger
- Alzheimer's Disease Cooperative StudyUniversity of CaliforniaLa JollaCaliforniaUSA
- Department of NeurosciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Karen Messer
- Herbert Wertheim School of Public Health and Human Longevity ScienceUniversity of California San DiegoLa JollaCaliforniaUSA
- Moors Cancer CenterUniversity of California San DiegoLa JollaCaliforniaUSA
| | - A. Carol Evans
- Alzheimer's Disease Cooperative StudyUniversity of CaliforniaLa JollaCaliforniaUSA
- Department of NeurosciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Ozioma C. Okonkwo
- Department of Medicine, Division of Geriatrics and GerontologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Aladdin H. Shadyab
- Herbert Wertheim School of Public Health and Human Longevity ScienceUniversity of California San DiegoLa JollaCaliforniaUSA
- Department of Medicine, Division of Geriatrics, Gerontology, and Palliative CareUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Jingjing Zou
- Alzheimer's Disease Cooperative StudyUniversity of CaliforniaLa JollaCaliforniaUSA
- Herbert Wertheim School of Public Health and Human Longevity ScienceUniversity of California San DiegoLa JollaCaliforniaUSA
- Moors Cancer CenterUniversity of California San DiegoLa JollaCaliforniaUSA
- Department of Medicine, Division of Geriatrics, Gerontology, and Palliative CareUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Sheila Jin
- Alzheimer's Disease Cooperative StudyUniversity of CaliforniaLa JollaCaliforniaUSA
- Department of NeurosciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Ronald G. Thomas
- Alzheimer's Disease Cooperative StudyUniversity of CaliforniaLa JollaCaliforniaUSA
- Herbert Wertheim School of Public Health and Human Longevity ScienceUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Jin Zhang
- Moors Cancer CenterUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Andrea Z. La Croix
- Herbert Wertheim School of Public Health and Human Longevity ScienceUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Carl W. Cotman
- Shiley‐Marcos Alzheimer's Disease Research CenterUniversity of California San DiegoLa JollaCaliforniaUSA
- Department of Neurobiology and Behavior; Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Howard H. Feldman
- Alzheimer's Disease Cooperative StudyUniversity of CaliforniaLa JollaCaliforniaUSA
- Department of NeurosciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | | |
Collapse
|
6
|
Oltra J, Kalpouzos G, Ekström I, Larsson M, Li Y, Qiu C, Laukka EJ. Cerebrovascular burden and neurodegeneration linked to 15-year odor identification decline in older adults. Front Aging Neurosci 2025; 17:1539508. [PMID: 40196179 PMCID: PMC11973317 DOI: 10.3389/fnagi.2025.1539508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
Background The mechanisms underlying olfactory decline in aging need further investigation. Noticeably, the longitudinal relationship of biological markers with olfaction remains underexplored. We investigated whether baseline levels and progression of microvascular lesions and brain atrophy are associated with odor identification (OID) decline. Methods The association between structural MRI markers and OID decline was examined in participants from the SNAC-K MRI study who were free from dementia at baseline (n = 401, mean age = 70.2 years, 60% females). OID was repeatedly assessed over 15 years. Presence of lacunes, white matter hyperintensities (WMH), perivascular spaces (PVS), and lateral ventricular, hippocampal, amygdalar, and total gray matter (GM) volumes were assessed up to 6 years, concurrent with the first 6 years of olfactory assessments. Results Higher PVS count and lower hippocampal and GM volumes at baseline were associated with accelerated OID decline (pFWE < 0.05). Longitudinally (n = 225), presence of lacunes at follow-up, faster WMH volume and PVS count increases, faster lateral ventricular enlargement, and faster hippocampal, amygdalar, and GM atrophy were associated with accelerated OID decline (p FWE < 0.05). Conclusion Olfactory decline is related to both increased cerebrovascular burden and accelerated brain atrophy over time.
Collapse
Affiliation(s)
- Javier Oltra
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Grégoria Kalpouzos
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Ingrid Ekström
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Maria Larsson
- Gösta Ekman Laboratory, Department of Psychology, Stockholm University, Stockholm, Sweden
| | - Yuanjing Li
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Chengxuan Qiu
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Erika J. Laukka
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Stockholm Gerontology Research Center, Stockholm, Sweden
| |
Collapse
|
7
|
Soelter TM, Howton TC, Wilk EJ, Whitlock JH, Clark AD, Birnbaum A, Patterson DC, Cortes CJ, Lasseigne BN. Evaluation of altered cell-cell communication between glia and neurons in the hippocampus of 3xTg-AD mice at two time points. J Cell Commun Signal 2025; 19:e70006. [PMID: 40026671 PMCID: PMC11870853 DOI: 10.1002/ccs3.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 03/05/2025] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by progressive memory loss and cognitive decline, affecting behavior, speech, and motor abilities. The neuropathology of AD includes the formation of extracellular amyloid-β plaques and intracellular neurofibrillary tangles of phosphorylated tau, along with neuronal loss. Although neuronal loss is an AD hallmark, cell-cell communication between neuronal and non-neuronal cell populations maintains neuronal health and brain homeostasis. To study changes in cell-cell communication during disease progression, we performed snRNA-sequencing of the hippocampus from female 3xTg-AD and wild-type littermates at 6 and 12 months. We inferred differential cell-cell communication between 3xTg-AD and wild-type mice across time points and between senders (astrocytes, microglia, oligodendrocytes, and OPCs) and receivers (excitatory and inhibitory neurons) of interest. We also assessed the downstream effects of altered glia-neuron communication using pseudobulk differential gene expression, functional enrichment, and gene regulatory analyses. We found that glia-neuron communication is increasingly dysregulated in 12-month 3xTg-AD mice. We also identified 23 AD-associated ligand-receptor pairs that are upregulated in the 12-month-old 3xTg-AD hippocampus. Our results suggest increased AD association of interactions originating from microglia. Signaling mediators were not significantly differentially expressed but showed altered gene regulation and transcription factor activity. Our findings indicate that altered glia-neuron communication is increasingly dysregulated and affects the gene regulatory mechanisms in neurons of 12-month-old 3xTg-AD mice.
Collapse
Affiliation(s)
- Tabea M. Soelter
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Timothy C. Howton
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Elizabeth J. Wilk
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Jordan H. Whitlock
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Amanda D. Clark
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Allison Birnbaum
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
- Department of Molecular, Cell and Developmental BiologyUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Dalton C. Patterson
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - Constanza J. Cortes
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Brittany N. Lasseigne
- Department of Cell, Developmental and Integrative BiologyHeersink School of MedicineThe University of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
8
|
Blasutto B, Fattapposta F, Casagrande M. Mild Behavioral Impairment and cognitive functions: A systematic review and meta-analysis. Ageing Res Rev 2025; 105:102668. [PMID: 39875064 DOI: 10.1016/j.arr.2025.102668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 01/09/2025] [Accepted: 01/19/2025] [Indexed: 01/30/2025]
Abstract
Mild behavioral impairment (MBI) represents a recently introduced diagnostic concept that focuses on behavioral and personality changes occurring in late life and associated with cognitive decline. Nevertheless, the relationship between these dimensions remains unclear. This systematic review and meta-analysis aim to analyze the relationship between MBI and cognitive functioning. The review process was conducted according to the PRISMA-Statement. Restrictions were made, selecting the studies published in peer-review journals, including at least one cognitive measure and presenting the measurement of MBI. Studies that included participants with neurological disorders, dementia, or psychiatric disorders or that only did a neuroimaging or genetic study were excluded. Twenty-two studies were included in the systematic review, while in the meta-analysis seventeen studies featured data to be included in the analyses. The results were classified according to the following cognitive domains: global cognitive functioning, memory, language, attention executive functions, visuospatial skills, and processing speed. In the quantitative analysis, only global cognitive functioning, executive function, attention, and memory were evaluated. The results of both qualitative and quantitative analysis indicate that individuals with MBI exhibited diminished performance on cognitive tasks when compared to those without MBI symptoms. These results are stronger when evaluating the various domains individually (particularly memory and executive functions) than when a global assessment was made. These findings highlight the potential role of MBI symptoms as early indicators of neurodegenerative processes, reinforcing the necessity for comprehensive assessments that encompass both behavioral and cognitive evaluations. The early detection of these symptoms in prodromal phases can be very useful for the development of non-pharmacological interventions and may provide relevant guidelines for clinicians in the management and diagnosis of neurodegenerative disorders.
Collapse
Affiliation(s)
- Barbara Blasutto
- Department of Psychology, University of Rome "Sapienza", Rome 00185, Italy.
| | - Francesco Fattapposta
- Department of Human Neuroscience, "Sapienza" University of Rome, Viale dell'Università 30, Rome 00185, Italy
| | - Maria Casagrande
- Department of Dynamic and Clinical Psychology and Health, University of Rome "Sapienza", Rome 00185, Italy.
| |
Collapse
|
9
|
Fuller OK, McLennan ED, Egan CL, Perera N, Terry LV, Pyun J, de Mendonca M, Telles GD, Smeuninx B, Burrows EL, Siddiqui G, Creek DJ, Scott JW, Pearen MA, Fonseka P, Nicolazzo JA, Mathivanan S, Hannan AJ, Ramm GA, Whitham M, Febbraio MA. Extracellular vesicles contribute to the beneficial effects of exercise training in APP/PS1 mice. iScience 2025; 28:111752. [PMID: 39898049 PMCID: PMC11787611 DOI: 10.1016/j.isci.2025.111752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/21/2024] [Accepted: 01/03/2025] [Indexed: 02/04/2025] Open
Abstract
Exercise improves cognitive function in Alzheimer's disease (AD) via mechanism that are not fully clear. Here, we first examined the effect of voluntary exercise training (VET) on energy metabolism and cognitive function in the APP/PS1 transgenic mouse (Tg) model of familial AD. Next, we profiled extracellular vesicles (EVs) and examined whether they may play a role in the protective effects of VET via intranasal administration of EVs, purified from the blood of sedentary (sEV) and/or acutely exercised (eEV) donor wild-type mice into APP/PS1Tg mice. We show that VET reduced resting energy expenditure (REE) and improved cognition in APP/PS1 Tg mice. Administration of eEV, but not sEV, also reduced REE, but had no effect on cognition. Taken together, these data show that exercise is effective intervention to improve symptoms of AD in APP/PS1Tg mice. In addition, eEVs mediate some of these effects, implicating EVs in the treatment of age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Oliver K. Fuller
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Emma D. McLennan
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Casey L. Egan
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Nimna Perera
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Lauren V. Terry
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Jae Pyun
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Mariana de Mendonca
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | | | - Benoit Smeuninx
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Emma L. Burrows
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Ghizal Siddiqui
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Darren J. Creek
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - John W. Scott
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | | | - Pamali Fonseka
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Joseph A. Nicolazzo
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Suresh Mathivanan
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Anthony J. Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Grant A. Ramm
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Martin Whitham
- School of Sport, Exercise & Rehabilitation Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Mark A. Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
10
|
Huang SY, Wu MT, Sun CF, Yang FY. Volume Changes in Brain Subfields of Patients with Alzheimer's Disease After Transcranial Ultrasound Stimulation. Diagnostics (Basel) 2025; 15:359. [PMID: 39941289 PMCID: PMC11817765 DOI: 10.3390/diagnostics15030359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Alzheimer's disease (AD) is characterized by progressive brain atrophy marked by cognitive decline and memory loss, which significantly affect patients' quality of life. Transcranial ultrasound stimulation (TUS) is a potential physical treatment for AD patients. However, the specific brain regions stimulated by TUS and its therapeutic effects remain unclear. Methods: In this study, magnetic resonance imaging (MRI) and FreeSurfer segmentation were employed to assess alterations in the brain volume of AD patients after TUS. Results: Our findings revealed significant volume increases in the corpus callosum (CC) and lateral orbitofrontal cortex (lOFC) in the TUS group. Moreover, the volumetric changes in the CC were strongly correlated with improvements in the Mini-Mental State Examination score, which is a widely used measure of cognitive function of AD patients. Conclusions: TUS has the potential to alleviate disease progression and offers a non-invasive therapeutic approach to the improvement of cognitive function in AD patients.
Collapse
Affiliation(s)
- Sheng-Yao Huang
- Department of Mathematics, Soochow University, Taipei 111, Taiwan;
| | - Meng-Ting Wu
- Division of Neurosurgery, Cheng Hsin General Hospital, Taipei 111, Taiwan;
| | - Chung-Fu Sun
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 111, Taiwan;
| | - Feng-Yi Yang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 111, Taiwan;
| |
Collapse
|
11
|
Genius P, Calle ML, Rodríguez‐Fernández B, Minguillon C, Cacciaglia R, Garrido‐Martin D, Esteller M, Navarro A, Gispert JD, Vilor‐Tejedor N. Compositional brain scores capture Alzheimer's disease-specific structural brain patterns along the disease continuum. Alzheimers Dement 2025; 21:e14490. [PMID: 39868465 PMCID: PMC11848177 DOI: 10.1002/alz.14490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 01/28/2025]
Abstract
INTRODUCTION Traditional multivariate methods for neuroimaging studies overlook the interdependent relationship between brain features. This study addresses this gap by analyzing relative brain volumetric patterns to capture how Alzheimer's disease (AD) and genetics influence brain structure along the disease continuum. METHODS This study analyzed data from participants across the AD continuum from the Alzheimer's and Families (ALFA) and Alzheimer's Disease Neuroimaging Initiative (ADNI) studies. Compositional data analysis (CoDA) was exploited to examine relative brain volumetric variations that (1) were linked to different AD stages compared to cognitively unimpaired amyloid-β-negative (CU A-) individuals and (2) varied by AD genetic risk. RESULTS Disease stage-specific compositional brain scores were identified, differentiating CU A- individuals from those in more advanced stages. Genetic risk-stratified models revealed a broader genetic landscape affecting brain morphology in AD, beyond the well-known apolipoprotein E ε4 allele. DISCUSSION CoDA emerges as an alternative multivariate framework to deepen understanding of AD-related structural changes and support targeted interventions for those at higher genetic risk. HIGHLIGHTS Compositional data analysis (CoDA) revealed the relative variation of brain region volumes, captured in compositional brain scores, capable of discerning between cognitively unimpaired amyloid-β-negative individuals and subjects within other disease-stage groups along the Alzheimer's disease (AD) continuum. CoDA also uncovered the genetic vulnerability of specific brain regions at each stage of the disease along the continuum. CoDA is capable of integrating magnetic resonance imaging data from two different cohorts without stringent requirements for harmonization. This translates as an advantage, compared to traditional methods, and strengthens the reliability of cross-study comparisons by standardizing the data despite different labeling agreements, facilitating collaborative and large-scale research. The algorithm is sensitive to AD-specific effects, as the main compositional brain scores display little overlap with the age-specific compositional brain score. CoDA provides a more accurate analysis of brain imaging data addressing its compositional nature, which can influence the development of targeted approaches, opening new avenues for enhancing brain health.
Collapse
Affiliation(s)
- Patricia Genius
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research Institute, Barcelona, Spain.BarcelonaSpain
- Centre for Genomic Regulation (CRG)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
- Doctoral SchoolPhD programme in BioinformaticsUniversity of Vic–Central University of Catalonia (UVic‐UCC)VicSpain
| | - M. Luz Calle
- Biosciences DepartmentFaculty of SciencesTechnology and EngineeringUniversity of Vic–Central University of CataloniaVicSpain
| | - Blanca Rodríguez‐Fernández
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research Institute, Barcelona, Spain.BarcelonaSpain
- Centre for Genomic Regulation (CRG)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Carolina Minguillon
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research Institute, Barcelona, Spain.BarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
| | - Raffaele Cacciaglia
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research Institute, Barcelona, Spain.BarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
| | - Diego Garrido‐Martin
- Department of Genetics, Microbiology and StatisticsUniversity of Barcelona (UB)BarcelonaSpain
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC)Ctra de Can RutiCamí de les EscolesBarcelonaSpain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)BarcelonaSpain
- Physiological Sciences DepartmentSchool of Medicine and Health SciencesUniversity of Barcelona (UB)BarcelonaSpain
- Centro de Investigación Biomédica en Red Cancer (CIBERONC)MadridSpain
| | - Arcadi Navarro
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centre for Genomic Regulation (CRG)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)BarcelonaSpain
- Pompeu Fabra UniversityBarcelonaSpain
- Institute of Evolutionary Biology (CSIC‐UPF)Department of Experimental and Health SciencesUniversitat Pompeu FabraBarcelonaSpain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research Institute, Barcelona, Spain.BarcelonaSpain
- Centro de Investigación Biomédica en Red BioingenieríaBiomateriales y NanomedicinaInstituto de Salud Carlos IIIMadridSpain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | - Natalia Vilor‐Tejedor
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research Institute, Barcelona, Spain.BarcelonaSpain
- Centre for Genomic Regulation (CRG)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
- Department of Human GeneticsRadboud University Medical CenterNijmegenthe Netherlands
| | | | | |
Collapse
|
12
|
Taddei RN, E Duff K. Synapse vulnerability and resilience underlying Alzheimer's disease. EBioMedicine 2025; 112:105557. [PMID: 39891995 PMCID: PMC11833146 DOI: 10.1016/j.ebiom.2025.105557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 02/03/2025] Open
Abstract
Synapse preservation is key for healthy cognitive ageing, and synapse loss represents a critical anatomical basis of cognitive dysfunction in Alzheimer's disease (AD), predicting dementia onset, severity, and progression. Synapse loss is viewed as a primary pathologic event, preceding neuronal loss and brain atrophy in AD. Synapses may, therefore, represent one of the earliest and clinically most meaningful targets of the neuropathologic processes driving AD dementia. The synapse loss in AD is highly selective and targets particularly vulnerable synapses while leaving others, termed resilient, largely unaffected. Yet, the anatomic and molecular hallmarks of the vulnerable and resilient synapse populations and their association with AD neuropathologic changes (e.g. amyloid-β plaques and tau tangles) and memory dysfunction remain poorly understood. Characterising the selectively vulnerable and resilient synapses in AD may be key to understanding the mechanisms of cognitive preservation versus loss and enable the development of robust biomarkers and disease-modifying therapies for dementia.
Collapse
Affiliation(s)
- Raquel N Taddei
- Neurology Department, Massachusetts General Hospital, Harvard Medical School, Boston, USA; UK Dementia Research Institute at UCL, Institute of Neurology, University College London, UK.
| | - Karen E Duff
- UK Dementia Research Institute at UCL, Institute of Neurology, University College London, UK
| |
Collapse
|
13
|
McDowall S, Bagda V, Hodgetts S, Mastaglia F, Li D. Controversies and insights into PTBP1-related astrocyte-neuron transdifferentiation: neuronal regeneration strategies for Parkinson's and Alzheimer's disease. Transl Neurodegener 2024; 13:59. [PMID: 39627843 PMCID: PMC11613593 DOI: 10.1186/s40035-024-00450-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
Promising therapeutic strategies are being explored to replace or regenerate the neuronal populations that are lost in patients with neurodegenerative disorders. Several research groups have attempted direct reprogramming of astrocytes into neurons by manipulating the expression of polypyrimidine tract-binding protein 1 (PTBP1) and claimed putative converted neurons to be functional, which led to improved disease outcomes in animal models of several neurodegenerative disorders. However, a few other studies reported data that contradict these claims, raising doubt about whether PTBP1 suppression truly reprograms astrocytes into neurons and the therapeutic potential of this approach. This review discusses recent advances in regenerative therapeutics including stem cell transplantations for central nervous system disorders, with a particular focus on Parkinson's and Alzheimer's diseases. We also provide a perspective on this controversy by considering that astrocyte heterogeneity may be the key to understanding the discrepancy in published studies, and that certain subpopulations of these glial cells may be more readily converted into neurons.
Collapse
Affiliation(s)
- Simon McDowall
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
- School of Human Sciences, The University of Western Australia, Crawley, Perth, WA, Australia
- Department of Anatomy and Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Vaishali Bagda
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
| | - Stuart Hodgetts
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
- School of Human Sciences, The University of Western Australia, Crawley, Perth, WA, Australia
| | - Frank Mastaglia
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.
| | - Dunhui Li
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.
- Centre for Neuromuscular and Neurological Disorders, Nedlands, WA, Australia.
- Department of Neurology and Stephen and Denise Adams Center for Parkinson's Disease Research, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
14
|
Tang J, Cao Z, Lei M, Yu Q, Mai Y, Xu J, Liao W, Ruan Y, Shi L, Yang L, Liu J. Heterogeneity of cerebral atrophic rate in mild cognitive impairment and its interactive association with proteins related to microglia activity on longitudinal cognitive changes. Arch Gerontol Geriatr 2024; 127:105582. [PMID: 39079281 DOI: 10.1016/j.archger.2024.105582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Heterogeneity of cerebral atrophic rate commonly exists in mild cognitive impairment (MCI), which may be associated with microglia-involved neuropathology and have an influence on cognitive outcomes. OBJECTIVE We aim to explore the heterogeneity of cerebral atrophic rate among MCI and its association with plasma proteins related to microglia activity, with further investigation of their interaction effects on long-term cognition. SUBJECTS A total of 630 MCI subjects in the ADNI database were included, of which 260 subjects were available with baseline data on plasma proteins. METHODS Group-based multi-trajectory modeling (GBMT) was used to identify the latent classes with heterogeneous cerebral atrophic rates. Associations between latent classes and plasma proteins related to microglia activity were investigated with generalized linear models. Linear mixed effect models (LME) were implemented to explore the interaction effects between proteins related to microglia activity and identified latent classes on longitudinal cognitive changes. RESULTS Two latent classes were identified and labeled as the slow-atrophy class and the fast-atrophy class. Associations were found between such heterogeneity of atrophic rates and plasma proteins related to microglia activity, especially AXL receptor tyrosine kinase (AXL), CD40 antigen (CD40), and tumor necrosis factor receptor-like 2 (TNF-R2). Interaction effects on longitudinal cognitive changes showed that higher CD40 was associated with faster cognitive decline in the slow-atrophy class and higher AXL or TNF-R2 was associated with slower cognitive decline in the fast-atrophy class. CONCLUSIONS Heterogeneity of atrophic rates at the MCI stage is associated with several plasma proteins related to microglia activity, which show either protective or adverse effects on long-term cognition depending on the variability of atrophic rates.
Collapse
Affiliation(s)
- Jingyi Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China
| | - Zhiyu Cao
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Ming Lei
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China
| | - Qun Yu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Yingren Mai
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Jiaxin Xu
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China
| | - Wang Liao
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Yuting Ruan
- Department of Rehabilitation, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Lin Shi
- BrainNow Research Institute, Shenzhen City, Guangdong Province, MN 518000, China; Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, MN 999077, China
| | - Lianhong Yang
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China.
| | - Jun Liu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China.
| |
Collapse
|
15
|
Lee H, Lee H, Ma Y, Eskandarian L, Gaudet K, Tian Q, Krijnen EA, Russo AW, Salat DH, Klawiter EC, Huang SY. Age-related alterations in human cortical microstructure across the lifespan: Insights from high-gradient diffusion MRI. Aging Cell 2024; 23:e14267. [PMID: 39118344 PMCID: PMC11561659 DOI: 10.1111/acel.14267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/16/2024] [Accepted: 06/24/2024] [Indexed: 08/10/2024] Open
Abstract
The human brain undergoes age-related microstructural alterations across the lifespan. Soma and Neurite Density Imaging (SANDI), a novel biophysical model of diffusion MRI, provides estimates of cell body (soma) radius and density, and neurite density in gray matter. The goal of this cross-sectional study was to assess the sensitivity of high-gradient diffusion MRI toward age-related alterations in cortical microstructure across the adult lifespan using SANDI. Seventy-two cognitively unimpaired healthy subjects (ages 19-85 years; 40 females) were scanned on the 3T Connectome MRI scanner with a maximum gradient strength of 300mT/m using a multi-shell diffusion MRI protocol incorporating 8 b-values and diffusion time of 19 ms. Intra-soma signal fraction obtained from SANDI model-fitting to the data was strongly correlated with age in all major cortical lobes (r = -0.69 to -0.60, FDR-p < 0.001). Intra-soma signal fraction (r = 0.48-0.63, FDR-p < 0.001) and soma radius (r = 0.28-0.40, FDR-p < 0.04) were significantly correlated with cortical volume in the prefrontal cortex, frontal, parietal, and temporal lobes. The strength of the relationship between SANDI metrics and age was greater than or comparable to the relationship between cortical volume and age across the cortical regions, particularly in the occipital lobe and anterior cingulate gyrus. In contrast to the SANDI metrics, all associations between diffusion tensor imaging (DTI) and diffusion kurtosis imaging metrics and age were low to moderate. These results suggest that high-gradient diffusion MRI may be more sensitive to underlying substrates of neurodegeneration in the aging brain than DTI and traditional macroscopic measures of neurodegeneration such as cortical volume and thickness.
Collapse
Affiliation(s)
- Hansol Lee
- Department of Radiology, Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Hong‐Hsi Lee
- Department of Radiology, Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Yixin Ma
- Department of Radiology, Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Laleh Eskandarian
- Department of Radiology, Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Kyla Gaudet
- Department of Radiology, Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Qiyuan Tian
- Department of Radiology, Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Eva A. Krijnen
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- MS Center Amsterdam, Anatomy and Neurosciences, Amsterdam NeuroscienceAmsterdam UMC Location VUmcAmsterdamThe Netherlands
| | - Andrew W. Russo
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - David H. Salat
- Department of Radiology, Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Eric C. Klawiter
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Susie Y. Huang
- Department of Radiology, Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalCharlestownMassachusettsUSA
- Harvard‐MIT Division of Health Sciences and TechnologyMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| |
Collapse
|
16
|
Lorin C, Guiet R, Chiaruttini N, Ambrosini G, Boci E, Abdellah M, Markram H, Keller D. Structural and molecular characterization of astrocyte and vasculature connectivity in the mouse hippocampus and cortex. Glia 2024; 72:2001-2021. [PMID: 39007459 DOI: 10.1002/glia.24594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024]
Abstract
The relation of astrocytic endfeet to the vasculature plays a key functional role in the neuro-glia-vasculature unit. We characterize the spatial organization of astrocytes and the structural aspects that facilitate their involvement in molecular exchanges. Using double transgenic mice, we performed co-immunostaining, confocal microscopy, and three-dimensional digital segmentation to investigate the biophysical and molecular organization of astrocytes and their intricate endfoot network at the micrometer level in the isocortex and hippocampus. The results showed that hippocampal astrocytes had smaller territories, reduced endfoot dimensions, and fewer contacts with blood vessels compared with those in the isocortex. Additionally, we found that both connexins 43 and 30 have a higher density in the endfoot and the former is overexpressed relative to the latter. However, due to the limitations of the method, further studies are needed to determine the exact localization on the endfoot. The quantitative information obtained in this study will be useful for modeling the interactions of astrocytes with the vasculature.
Collapse
Affiliation(s)
- Charlotte Lorin
- Blue Brain Project, Swiss Federal Institute of Technology Lausanne (EPFL), Campus Biotech, Geneva, Switzerland
| | - Romain Guiet
- Bioimaging and Optics Platform, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Nicolas Chiaruttini
- Bioimaging and Optics Platform, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Giovanna Ambrosini
- Bioinformatics Competence Center, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
- Bioinformatics Competence Center, University of Lausanne, Lausanne, Switzerland
| | - Elvis Boci
- Blue Brain Project, Swiss Federal Institute of Technology Lausanne (EPFL), Campus Biotech, Geneva, Switzerland
| | - Marwan Abdellah
- Blue Brain Project, Swiss Federal Institute of Technology Lausanne (EPFL), Campus Biotech, Geneva, Switzerland
| | - Henry Markram
- Blue Brain Project, Swiss Federal Institute of Technology Lausanne (EPFL), Campus Biotech, Geneva, Switzerland
| | - Daniel Keller
- Blue Brain Project, Swiss Federal Institute of Technology Lausanne (EPFL), Campus Biotech, Geneva, Switzerland
| |
Collapse
|
17
|
Alves VC, Carro E, Figueiro-Silva J. Unveiling DNA methylation in Alzheimer's disease: a review of array-based human brain studies. Neural Regen Res 2024; 19:2365-2376. [PMID: 38526273 PMCID: PMC11090417 DOI: 10.4103/1673-5374.393106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/05/2023] [Indexed: 03/26/2024] Open
Abstract
The intricacies of Alzheimer's disease pathogenesis are being increasingly illuminated by the exploration of epigenetic mechanisms, particularly DNA methylation. This review comprehensively surveys recent human-centered studies that investigate whole genome DNA methylation in Alzheimer's disease neuropathology. The examination of various brain regions reveals distinctive DNA methylation patterns that associate with the Braak stage and Alzheimer's disease progression. The entorhinal cortex emerges as a focal point due to its early histological alterations and subsequent impact on downstream regions like the hippocampus. Notably, ANK1 hypermethylation, a protein implicated in neurofibrillary tangle formation, was recurrently identified in the entorhinal cortex. Further, the middle temporal gyrus and prefrontal cortex were shown to exhibit significant hypermethylation of genes like HOXA3, RHBDF2, and MCF2L, potentially influencing neuroinflammatory processes. The complex role of BIN1 in late-onset Alzheimer's disease is underscored by its association with altered methylation patterns. Despite the disparities across studies, these findings highlight the intricate interplay between epigenetic modifications and Alzheimer's disease pathology. Future research efforts should address methodological variations, incorporate diverse cohorts, and consider environmental factors to unravel the nuanced epigenetic landscape underlying Alzheimer's disease progression.
Collapse
Affiliation(s)
- Victoria Cunha Alves
- Neurodegenerative Diseases Group, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain
- Network Center for Biomedical Research, Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- PhD Program in Neuroscience, Autonoma de Madrid University, Madrid, Spain
- Neurotraumatology and Subarachnoid Hemorrhage Group, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain
| | - Eva Carro
- Network Center for Biomedical Research, Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research Into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Joana Figueiro-Silva
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Science, University of Zurich, Zurich, Switzerland
| |
Collapse
|
18
|
Yang C, Sun ZP, Jiang J, Cai XL, Wang Y, Wang H, Che C, Tu E, Pan AH, Zhang Y, Wang XP, Cui MZ, Xu XM, Yan XX, Zhang QL. Increased expression of the proapoptotic presenilin associated protein is involved in neuronal tangle formation in human brain. Sci Rep 2024; 14:25274. [PMID: 39455681 PMCID: PMC11512019 DOI: 10.1038/s41598-024-77026-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Presenilin-associated protein (PSAP) is a mitochondrial proapoptotic protein as established in cell biology studies. It remains unknown whether it involves in neurodegenerative diseases. Here, we explored PASP expression in adult and aged human brains and its alteration relative to Alzheimer-disease (AD)-type neuropathology. In pathology-free brains, light PASP immunoreactivity (IR) occurred among largely principal neurons in the cerebrum and subcortical structures. In the brains with AD pathology, enhanced PSAP IR occurred in neuronal and neuritic profiles with a tangle-like appearance, with PSAP and pTau protein levels elevated in neocortical lysates relative to control. Neuronal/neuritic profiles with enhanced PSAP IR partially colocalized with pTau, but invariably with Amylo-Glo labelled tangles. The neuronal somata with enhanced PASP IR also showed diminished IR for casein kinase 1 delta (Ck1δ), a marker of granulovacuolar degeneration; and diminished IR for sortilin, which is normally expressed in membrane and intracellular protein sorting/trafficking organelles. In old 3xTg-AD mice with β-amyloid and pTau pathologies developed in the brain, PSAP IR in the cerebral sections exhibited no difference relative to wildtype mice. These findings indicate that PSAP upregulation is involved in the course of tangle formation especially in the human brain during aging and in AD pathogenesis.
Collapse
Affiliation(s)
- Chen Yang
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Zhong-Ping Sun
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
- Department of Biology, College of Arts and Sciences, University of Texas of the Permian Basin, Odessa, TX, USA
| | - Juan Jiang
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
- Department of Biology, College of Arts and Sciences, University of Texas of the Permian Basin, Odessa, TX, USA
| | - Xiao-Lu Cai
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
- Department of Biology, College of Arts and Sciences, University of Texas of the Permian Basin, Odessa, TX, USA
| | - Yan Wang
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Hui Wang
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Chong Che
- GeneScience Pharmaceuticals Co., Ltd, Changchun High-Tech Development Zone, Changchun, Jilin Province, China
| | - Ewen Tu
- Department of Neurology, Brain Hospital of Hunan Province, Changsha, Hunan Province, China
| | - Ai-Hua Pan
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Yan Zhang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, Second Xiangya Hospital Central South University, Changsha, Hunan Province, China
| | - Xiao-Ping Wang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, Second Xiangya Hospital Central South University, Changsha, Hunan Province, China
| | - Mei-Zhen Cui
- Department of Biology, College of Arts and Sciences, University of Texas of the Permian Basin, Odessa, TX, USA
| | - Xue-Min Xu
- Department of Biology, College of Arts and Sciences, University of Texas of the Permian Basin, Odessa, TX, USA
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Qi-Lei Zhang
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China.
| |
Collapse
|
19
|
Ly M, Yu G, Son SJ, Pascoal T, Karim HT. Longitudinal accelerated brain age in mild cognitive impairment and Alzheimer's disease. Front Aging Neurosci 2024; 16:1433426. [PMID: 39503045 PMCID: PMC11534682 DOI: 10.3389/fnagi.2024.1433426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Brain age is a machine learning-derived estimate that captures lower brain volume. Previous studies have found that brain age is significantly higher in mild cognitive impairment and Alzheimer's disease (AD) compared to healthy controls. Few studies have investigated changes in brain age longitudinally in MCI and AD. We hypothesized that individuals with MCI and AD would show heightened brain age over time and across the lifespan. We also hypothesized that both MCI and AD would show faster rates of brain aging (higher slopes) over time compared to healthy controls. Methods We utilized data from an archival dataset, mainly Alzheimer's disease Neuroimaging Initiative (ADNI) 1 with 3Tesla (3 T) data which totaled 677 scans from 183 participants. This constitutes a secondary data analysis on existing data. We included control participants (healthy controls or HC), individuals with MCI, and individuals with AD. We predicted brain age using a pre-trained model and tested for accuracy. We investigated cross-sectional differences in brain age by group [healthy controls or HC, mild cognitive impairment (MCI), and AD]. We conducted longitudinal modeling of age and brain age by group using time from baseline in one model and chronological age in another model. Results We predicted brain age with a mean absolute error (MAE) < 5 years. Brain age was associated with age across the study and individuals with MCI and AD had greater brain age on average. We found that the MCI group had significantly higher rates of change in brain age over time compared to the HC group regardless of individual chronologic age, while the AD group did not differ in rate of brain age change. Discussion We replicated past studies that showed that MCI and AD had greater brain age than HC. We additionally found that this was true over time, both groups showed higher brain age longitudinally. Contrary to our hypothesis, we found that the MCI, but not the AD group, showed faster rates of brain aging. We essentially found that while the MCI group was actively experiencing faster rates of brain aging, the AD group may have already experienced this acceleration (as they show higher brain age). Individuals with MCI may experience higher rates of brain aging than AD and controls. AD may represent a homeostatic endpoint after significant neurodegeneration. Future work may focus on individuals with MCI as one potential therapeutic option is to alter rates of brain aging, which ultimately may slow cognitive decline in the long-term.
Collapse
Affiliation(s)
- Maria Ly
- Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA, United States
| | - Gary Yu
- Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA, United States
| | - Sang Joon Son
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Tharick Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Helmet T. Karim
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | | |
Collapse
|
20
|
Mugotitsa B, Bhattacharjee T, Ochola M, Mailosi D, Amadi D, Andeso P, Kuria J, Momanyi R, Omondi E, Kajungu D, Todd J, Kiragga A, Greenfield J. Integrating longitudinal mental health data into a staging database: harnessing DDI-lifecycle and OMOP vocabularies within the INSPIRE Network Datahub. Front Big Data 2024; 7:1435510. [PMID: 39463847 PMCID: PMC11502395 DOI: 10.3389/fdata.2024.1435510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/09/2024] [Indexed: 10/29/2024] Open
Abstract
Background Longitudinal studies are essential for understanding the progression of mental health disorders over time, but combining data collected through different methods to assess conditions like depression, anxiety, and psychosis presents significant challenges. This study presents a mapping technique allowing for the conversion of diverse longitudinal data into a standardized staging database, leveraging the Data Documentation Initiative (DDI) Lifecycle and the Observational Medical Outcomes Partnership (OMOP) Common Data Model (CDM) standards to ensure consistency and compatibility across datasets. Methods The "INSPIRE" project integrates longitudinal data from African studies into a staging database using metadata documentation standards structured with a snowflake schema. This facilitates the development of Extraction, Transformation, and Loading (ETL) scripts for integrating data into OMOP CDM. The staging database schema is designed to capture the dynamic nature of longitudinal studies, including changes in research protocols and the use of different instruments across data collection waves. Results Utilizing this mapping method, we streamlined the data migration process to the staging database, enabling subsequent integration into the OMOP CDM. Adherence to metadata standards ensures data quality, promotes interoperability, and expands opportunities for data sharing in mental health research. Conclusion The staging database serves as an innovative tool in managing longitudinal mental health data, going beyond simple data hosting to act as a comprehensive study descriptor. It provides detailed insights into each study stage and establishes a data science foundation for standardizing and integrating the data into OMOP CDM.
Collapse
Affiliation(s)
- Bylhah Mugotitsa
- African Population and Health Research Center (APHRC), Nairobi, Kenya
- Strathmore University Business School, Strathmore University, Nairobi, Kenya
| | - Tathagata Bhattacharjee
- Department of Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Michael Ochola
- African Population and Health Research Center (APHRC), Nairobi, Kenya
| | - Dorothy Mailosi
- Artificial Intelligence and Machine Learning (AI and ML), CODATA-Committee on Data of the International Science Council, Paris, France
| | - David Amadi
- Department of Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Pauline Andeso
- African Population and Health Research Center (APHRC), Nairobi, Kenya
| | - Joseph Kuria
- African Population and Health Research Center (APHRC), Nairobi, Kenya
| | - Reinpeter Momanyi
- African Population and Health Research Center (APHRC), Nairobi, Kenya
| | - Evans Omondi
- African Population and Health Research Center (APHRC), Nairobi, Kenya
- Institute of Mathematical Sciences, Strathmore University, Nairobi, Kenya
| | - Dan Kajungu
- Iganga Mayuge Health and Demographic Surveillance Site (IMHDSS), Makerere University Centre for Health and Population Research (MUCHAP), Kampala, Uganda
| | - Jim Todd
- Department of Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Agnes Kiragga
- African Population and Health Research Center (APHRC), Nairobi, Kenya
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Jay Greenfield
- Artificial Intelligence and Machine Learning (AI and ML), CODATA-Committee on Data of the International Science Council, Paris, France
| |
Collapse
|
21
|
Woods C, Xing X, Khanal S, Lin AL. Machine Learning-Driven Prediction of Brain Age for Alzheimer's Risk: APOE4 Genotype and Gender Effects. Bioengineering (Basel) 2024; 11:943. [PMID: 39329685 PMCID: PMC11429338 DOI: 10.3390/bioengineering11090943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/11/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024] Open
Abstract
Background: Alzheimer's disease (AD) is a leading cause of dementia, and it is significantly influenced by the apolipoprotein E4 (APOE4) gene and gender. This study aimed to use machine learning (ML) algorithms to predict brain age and assess AD risk by considering the effects of the APOE4 genotype and gender. Methods: We collected brain volumetric MRI data and medical records from 1100 cognitively unimpaired individuals and 602 patients with AD. We applied three ML regression models-XGBoost, random forest (RF), and linear regression (LR)-to predict brain age. Additionally, we introduced two novel metrics, brain age difference (BAD) and integrated difference (ID), to evaluate the models' performances and analyze the influences of the APOE4 genotype and gender on brain aging. Results: Patients with AD displayed significantly older brain ages compared to their chronological ages, with BADs ranging from 6.5 to 10 years. The RF model outperformed both XGBoost and LR in terms of accuracy, delivering higher ID values and more precise predictions. Comparing the APOE4 carriers with noncarriers, the models showed enhanced ID values and consistent brain age predictions, improving the overall performance. Gender-specific analyses indicated slight enhancements, with the models performing equally well for both genders. Conclusions: This study demonstrates that robust ML models for brain age prediction can play a crucial role in the early detection of AD risk through MRI brain structural imaging. The significant impact of the APOE4 genotype on brain aging and AD risk is also emphasized. These findings highlight the potential of ML models in assessing AD risk and suggest that utilizing AI for AD identification could enable earlier preventative interventions.
Collapse
Affiliation(s)
- Carter Woods
- Department of Physics, University of Missouri, Columbia, MO 65211, USA
| | - Xin Xing
- Department of Computer Science, University of Nebraska Omaha, Omaha, NE 68182, USA;
| | - Subash Khanal
- Department of Computer Science & Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Ai-Ling Lin
- Department of Radiology, Biology and Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65211, USA
- Sanders-Brown Center on Aging, Department for Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
22
|
Soelter TM, Howton TC, Wilk EJ, Whitlock JH, Clark AD, Birnbaum A, Patterson DC, Cortes CJ, Lasseigne BN. Evaluation of altered cell-cell communication between glia and neurons in the hippocampus of 3xTg-AD mice at two time points. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595199. [PMID: 38826305 PMCID: PMC11142088 DOI: 10.1101/2024.05.21.595199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by progressive memory loss and cognitive decline, affecting behavior, speech, and motor abilities. The neuropathology of AD includes the formation of extracellular amyloid-β plaque and intracellular neurofibrillary tangles of phosphorylated tau, along with neuronal loss. While neuronal loss is an AD hallmark, cell-cell communication between neuronal and non-neuronal cell populations maintains neuronal health and brain homeostasis. To study changes in cellcell communication during disease progression, we performed snRNA-sequencing of the hippocampus from female 3xTg-AD and wild-type littermates at 6 and 12 months. We inferred differential cell-cell communication between 3xTg-AD and wild-type mice across time points and between senders (astrocytes, microglia, oligodendrocytes, and OPCs) and receivers (excitatory and inhibitory neurons) of interest. We also assessed the downstream effects of altered glia-neuron communication using pseudobulk differential gene expression, functional enrichment, and gene regulatory analyses. We found that glia-neuron communication is increasingly dysregulated in 12-month 3xTg-AD mice. We also identified 23 AD-associated ligand-receptor pairs that are upregulated in the 12-month-old 3xTg-AD hippocampus. Our results suggest increased AD association of interactions originating from microglia. Signaling mediators were not significantly differentially expressed but showed altered gene regulation and TF activity. Our findings indicate that altered glia-neuron communication is increasingly dysregulated and affects the gene regulatory mechanisms in neurons of 12-month-old 3xTg-AD mice.
Collapse
Affiliation(s)
- Tabea M. Soelter
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Timothy C. Howton
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Elizabeth J. Wilk
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jordan H. Whitlock
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Amanda D. Clark
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Allison Birnbaum
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Dalton C. Patterson
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Constanza J. Cortes
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - Brittany N. Lasseigne
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
23
|
Liao W, Wang Y, Wang L, Li J, Huang D, Cheng W, Luan P. The current status and challenges of olfactory dysfunction study in Alzheimer's Disease. Ageing Res Rev 2024; 100:102453. [PMID: 39127444 DOI: 10.1016/j.arr.2024.102453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Olfactory functioning involves multiple cognitive processes and the coordinated actions of various neural systems. Any disruption at any stage of this process may result in olfactory dysfunction, which is consequently widely used to predict the onset and progression of diseases, such as Alzheimer's Disease (AD). Although the underlying mechanisms have not yet been fully unraveled, apparent changes were observed in olfactory brain areas form patients who suffer from AD by means of medical imaging and electroencephalography (EEG). Olfactory dysfunction holds significant promise in detecting AD during the preclinical stage preceding mild cognitive impairment (MCI). Owing to the strong specificity, olfactory tests are prevalently applied for screening in community cohorts. And combining olfactory tests with other biomarkers may further establish an optimal model for AD prediction in studies of specific olfactory dysfunctions and improve the sensitivity and specificity of early AD diagnosis.
Collapse
Affiliation(s)
- Wanchen Liao
- Department of Alzheimer's Disease Clinical Research Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Yulin Wang
- Department of Alzheimer's Disease Clinical Research Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Lei Wang
- Department of Alzheimer's Disease Clinical Research Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Jun Li
- Department of Alzheimer's Disease Clinical Research Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Dongqing Huang
- Department of Alzheimer's Disease Clinical Research Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Weibin Cheng
- Department of Alzheimer's Disease Clinical Research Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China.
| | - Ping Luan
- Department of Alzheimer's Disease Clinical Research Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China.
| |
Collapse
|
24
|
Whitman ET, Elliott ML, Knodt AR, Abraham WC, Anderson TJ, Cutfield N, Hogan S, Ireland D, Melzer TR, Ramrakha S, Sugden K, Theodore R, Williams BS, Caspi A, Moffitt TE, Hariri AR. An estimate of the longitudinal pace of aging from a single brain scan predicts dementia conversion, morbidity, and mortality. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608305. [PMID: 39229058 PMCID: PMC11370321 DOI: 10.1101/2024.08.19.608305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
To understand how aging affects functional decline and increases disease risk, it is necessary to develop accurate and reliable measures of how fast a person is aging. Epigenetic clocks measure aging but require DNA methylation data, which many studies lack. Using data from the Dunedin Study, we introduce an accurate and reliable measure for the rate of longitudinal aging derived from cross-sectional brain MRI: the Dunedin Pace of Aging Calculated from NeuroImaging or DunedinPACNI. Exporting this measure to the Alzheimer's Disease Neuroimaging Initiative and UK Biobank neuroimaging datasets revealed that faster DunedinPACNI predicted participants' cognitive impairment, accelerated brain atrophy, and conversion to diagnosed dementia. Underscoring close links between longitudinal aging of the body and brain, faster DunedinPACNI also predicted physical frailty, poor health, future chronic diseases, and mortality in older adults. Furthermore, DunedinPACNI followed the expected socioeconomic health gradient. When compared to brain age gap, an existing MRI aging biomarker, DunedinPACNI was similarly or more strongly related to clinical outcomes. DunedinPACNI is a "next generation" MRI measure that will be made publicly available to the research community to help accelerate aging research and evaluate the effectiveness of dementia prevention and anti-aging strategies.
Collapse
Affiliation(s)
- Ethan T Whitman
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Maxwell L Elliott
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Annchen R Knodt
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | | | - Tim J Anderson
- Department of Medicine, University of Otago, Christchurch, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Department of Neurology, Canterbury District Health Board, Christchurch, New Zealand
| | - Nick Cutfield
- Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Sean Hogan
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - David Ireland
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Tracy R Melzer
- Brain Research New Zealand-Rangahau Roro Aotearoa, Centre of Research Excellence, Universities of Auckland and Otago, New Zealand
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Sandhya Ramrakha
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Reremoana Theodore
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | | | - Avshalom Caspi
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
- King's College London, Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, & Neuroscience, London, UK
- PROMENTA, Department of Psychology, University of Oslo, Norway
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Terrie E Moffitt
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
- King's College London, Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, & Neuroscience, London, UK
- PROMENTA, Department of Psychology, University of Oslo, Norway
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Ahmad R Hariri
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| |
Collapse
|
25
|
Sacchi L, D'Agata F, Campisi C, Arcaro M, Carandini T, Örzsik B, Dal Maschio VP, Fenoglio C, Pietroboni AM, Ghezzi L, Serpente M, Pintus M, Conte G, Triulzi F, Lopiano L, Galimberti D, Cercignani M, Bozzali M, Arighi A. A "glympse" into neurodegeneration: Diffusion MRI and cerebrospinal fluid aquaporin-4 for the assessment of glymphatic system in Alzheimer's disease and other dementias. Hum Brain Mapp 2024; 45:e26805. [PMID: 39185685 PMCID: PMC11345637 DOI: 10.1002/hbm.26805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 06/17/2024] [Accepted: 07/17/2024] [Indexed: 08/27/2024] Open
Abstract
The glymphatic system (GS) is a whole-brain perivascular network, consisting of three compartments: the periarterial and perivenous spaces and the interposed brain parenchyma. GS dysfunction has been implicated in neurodegenerative diseases, particularly Alzheimer's disease (AD). So far, comprehensive research on GS in humans has been limited by the absence of easily accessible biomarkers. Recently, promising non-invasive methods based on magnetic resonance imaging (MRI) along with aquaporin-4 (AQP4) quantification in the cerebrospinal fluid (CSF) were introduced for an indirect assessment of each of the three GS compartments. We recruited 111 consecutive subjects presenting with symptoms suggestive of degenerative cognitive decline, who underwent 3 T MRI scanning including multi-shell diffusion-weighted images. Forty nine out of 111 also underwent CSF examination with quantification of CSF-AQP4. CSF-AQP4 levels and MRI measures-including perivascular spaces (PVS) counts and volume fraction (PVSVF), white matter free water fraction (FW-WM) and mean kurtosis (MK-WM), diffusion tensor imaging analysis along the perivascular spaces (DTI-ALPS) (mean, left and right)-were compared among patients with AD (n = 47) and other neurodegenerative diseases (nAD = 24), patients with stable mild cognitive impairment (MCI = 17) and cognitively unimpaired (CU = 23) elderly people. Two runs of analysis were conducted, the first including all patients; the second after dividing both nAD and AD patients into two subgroups based on gray matter atrophy as a proxy of disease stage. Age, sex, years of education, and scanning time were included as confounding factors in the analyses. Considering the whole cohort, patients with AD showed significantly higher levels of CSF-AQP4 (exp(b) = 2.05, p = .005) and FW-WM FW-WM (exp(b) = 1.06, p = .043) than CU. AQP4 levels were also significantly higher in nAD in respect to CU (exp(b) = 2.98, p < .001). CSF-AQP4 and FW-WM were significantly higher in both less atrophic AD (exp(b) = 2.20, p = .006; exp(b) = 1.08, p = .019, respectively) and nAD patients (exp(b) = 2.66, p = .002; exp(b) = 1.10, p = .019, respectively) compared to CU subjects. Higher total (exp(b) = 1.59, p = .013) and centrum semiovale PVS counts (exp(b) = 1.89, p = .016), total (exp(b) = 1.50, p = .036) and WM PVSVF (exp(b) = 1.89, p = .005) together with lower MK-WM (exp(b) = 0.94, p = .006), mean and left ALPS (exp(b) = 0.91, p = .043; exp(b) = 0.88, p = .010 respectively) were observed in more atrophic AD patients in respect to CU. In addition, more atrophic nAD patients exhibited higher levels of AQP4 (exp(b) = 3.39, p = .002) than CU. Our results indicate significant changes in putative MRI biomarkers of GS and CSF-AQP4 levels in AD and in other neurodegenerative dementias, suggesting a close interaction between glymphatic dysfunction and neurodegeneration, particularly in the case of AD. However, the usefulness of some of these biomarkers as indirect and standalone indices of glymphatic activity may be hindered by their dependence on disease stage and structural brain damage.
Collapse
Affiliation(s)
- Luca Sacchi
- Department of Biomedical, Surgical and Dental SciencesUniversity of MilanMilanItaly
| | - Federico D'Agata
- Department of Neurosciences “Rita Levi Montalcini”University of TurinTurinItaly
| | - Corrado Campisi
- Department of Neurosciences “Rita Levi Montalcini”University of TurinTurinItaly
| | - Marina Arcaro
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore PoliclinicoMilanItaly
| | - Tiziana Carandini
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore PoliclinicoMilanItaly
| | - Balázs Örzsik
- Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Vera Pacoova Dal Maschio
- Department of Neurosciences “Rita Levi Montalcini”University of TurinTurinItaly
- Neurology 2 Unit, A.O.U. Città della Salute e Della Scienza di TorinoTurinItaly
| | - Chiara Fenoglio
- Department of Biomedical, Surgical and Dental SciencesUniversity of MilanMilanItaly
| | | | - Laura Ghezzi
- Department of Biomedical, Surgical and Dental SciencesUniversity of MilanMilanItaly
| | - Maria Serpente
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore PoliclinicoMilanItaly
| | - Manuela Pintus
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore PoliclinicoMilanItaly
| | - Giorgio Conte
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore PoliclinicoMilanItaly
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Fabio Triulzi
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore PoliclinicoMilanItaly
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Leonardo Lopiano
- Department of Neurosciences “Rita Levi Montalcini”University of TurinTurinItaly
- Neurology 2 Unit, A.O.U. Città della Salute e Della Scienza di TorinoTurinItaly
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental SciencesUniversity of MilanMilanItaly
| | | | - Marco Bozzali
- Department of Neurosciences “Rita Levi Montalcini”University of TurinTurinItaly
- Neurology 2 Unit, A.O.U. Città della Salute e Della Scienza di TorinoTurinItaly
| | - Andrea Arighi
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore PoliclinicoMilanItaly
| |
Collapse
|
26
|
Sen S, Mukhopadhyay D. A Holistic Analysis of Alzheimer's Disease-Associated lncRNA Communities Reveals Enhanced lncRNA-miRNA-RBP Regulatory Triad Formation Within Functionally Segregated Clusters. J Mol Neurosci 2024; 74:77. [PMID: 39143264 PMCID: PMC11324768 DOI: 10.1007/s12031-024-02244-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/04/2024] [Indexed: 08/16/2024]
Abstract
Recent studies on the regulatory networks implicated in Alzheimer's disease (AD) evince long non-coding RNAs (lncRNAs) as crucial regulatory players, albeit a poor understanding of the mechanism. Analyzing differential gene expression in the RNA-seq data from the post-mortem AD brain hippocampus, we categorized a list of AD-dysregulated lncRNA transcripts into functionally similar communities based on their k-mer profiles. Using machine-learning-based algorithms, their subcellular localizations were mapped. We further explored the functional relevance of each community through AD-dysregulated miRNA, RNA-binding protein (RBP) interactors, and pathway enrichment analyses. Further investigation of the miRNA-lncRNA and RBP-lncRNA networks from each community revealed the top RBPs, miRNAs, and lncRNAs for each cluster. The experimental validation community yielded ELAVL4 and miR-16-5p as the predominant RBP and miRNA, respectively. Five lncRNAs emerged as the top-ranking candidates from the RBP/miRNA-lncRNA networks. Further analyses of these networks revealed the presence of multiple regulatory triads where the RBP-lncRNA interactions could be augmented by the enhanced miRNA-lncRNA interactions. Our results advance the understanding of the mechanism of lncRNA-mediated AD regulation through their interacting partners and demonstrate how these functionally segregated but overlapping regulatory networks can modulate the disease holistically.
Collapse
Affiliation(s)
- Somenath Sen
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, A CI of Homi Bhabha National Institute, Kolkata, 700 064, India
| | - Debashis Mukhopadhyay
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, A CI of Homi Bhabha National Institute, Kolkata, 700 064, India.
| |
Collapse
|
27
|
Singh S, Malo PK, Stezin A, Mensegere AL, Issac TG. Alteration in amygdala subfield volumes and their association with cognition in mild cognitive impairment. J Neurol 2024; 271:5460-5467. [PMID: 38879703 DOI: 10.1007/s00415-024-12500-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/31/2024] [Accepted: 06/07/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND The amygdala has an important role in cognitive and affective functions. The involvement of amygdala and related limbic structures is implicated in many aspects of memory and emotion in mild cognitive impairment (MCI). In the present study, we aimed to compare the volumetric measurements of amygdala and its subfields as well as their association with cognitive functions in stable MCI (sMCI). METHODS We performed Addenbrooke's cognitive examination III (ACE-III) test, as well as high-resolution T1-weighted images from 31 participants with sMCI and 31 age-matched healthy controls. The amygdala subfield volumes were extracted using Freesurfer software, and group differences were assessed using general linear model (GLM) with age, gender, education and estimated intracranial volume (ICV) as covariates. Partial correlation was also calculated between cognitive scores and volumes of amygdala subfields in healthy controls and sMCI participants controlling for estimated ICV. RESULTS sMCI participants exhibited significantly reduced volumes in most of the right amygdala subfields, including basal nucleus, accessory basal nucleus, central nucleus, medial nucleus, corticoamygdaloid transition area, and whole amygdala, as well as significantly reduced right amygdala/hippocampus ratio compared to healthy controls. In addition, our results revealed statistically significant positive correlations between ACE memory scores and the volumes of right central nucleus, right medial nucleus, right cortical nucleus, and the right whole amygdala, in sMCI. CONCLUSIONS Our findings revealed volumetric reductions in most of the right amygdala subfields along with its association with the memory functions in sMCI. These findings provide valuable insights into the underlying anatomical factors contributing to neurocognitive symptoms in MCI.
Collapse
Affiliation(s)
- Sadhana Singh
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India
| | - Palash Kumar Malo
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India
| | - Albert Stezin
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India
| | - Abhishek L Mensegere
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India
| | - Thomas Gregor Issac
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
28
|
Ma Z, Zhang Z, Lv X, Zhang H, Lu K, Su G, Huang B, Chen H. Dual sensitivity-enhanced microring resonance-based integrated microfluidic biosensor for Aβ 42 detection. Talanta 2024; 275:126111. [PMID: 38657362 DOI: 10.1016/j.talanta.2024.126111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 04/07/2024] [Accepted: 04/13/2024] [Indexed: 04/26/2024]
Abstract
Sensitive, accurate, and straightforward biosensors are pivotal in the battle against Alzheimer's disease, particularly in light of the escalating patient population. These biosensors enable early adjunctive diagnosis, thereby facilitating prompt intervention, alleviating socioeconomic burdens, and preserving individual well-being. In this study, we introduce the development of a highly sensitive add-drop dual-microring resonant microfluidic sensing chip boasting a sensitivity of 188.11 nm/RIU, marking a significant 20.7% enhancement over single microring systems. Leveraging ultra-thin Parylene C for streamlined antibody immobilization and non-destructive removal, this platform facilitates the precise quantification of the Alzheimer's disease biomarker Aβ42. Employing an immune sensing strategy that amplifies and captures antigen signals using Au-labeled antibodies, we achieve an exceptional limit of detection of 9.02 pg/mL. The designed microring-based microfluidic biosensor chip exhibits outstanding specificity and sensitivity for Aβ42 in serum samples, offering a promising avenue for the early adjunctive diagnosis of Alzheimer's disease.
Collapse
Affiliation(s)
- Zhengtai Ma
- Key Laboratory of Optoelectronic Materials and Devices, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, China; College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Zan Zhang
- School of Electronic and Control Engineering, Chang'an University, Xi'an, China
| | - Xiaoqing Lv
- Key Laboratory of Optoelectronic Materials and Devices, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, China; College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Huan Zhang
- Key Laboratory of Optoelectronics Technology, Ministry of Education, Beijing University of Technology, Beijing, China
| | - Kaiwei Lu
- Key Laboratory of Optoelectronic Materials and Devices, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, China; College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Guoshuai Su
- Suzhou Institute of Microelectronics and Optoelectronics Integration, Suzhou, China; Suzhou Jiwei Photoelectric Co., Ltd, Suzhou, China
| | - Beiju Huang
- Key Laboratory of Optoelectronic Materials and Devices, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, China; College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, China.
| | - Hongda Chen
- Key Laboratory of Optoelectronic Materials and Devices, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, China; College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
29
|
Liu YT, Yang YT, Tang CX, Ma JQ, Kong X, Li JH, Li YM, Liu SY, Zhou CS, Wang YF, Zhang LJ. Aberrant cortical morphology patterns are associated with cognitive impairment in patients with chronic heart failure. Eur J Neurosci 2024; 60:3973-3983. [PMID: 38711292 DOI: 10.1111/ejn.16382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024]
Abstract
A mounting body of evidences suggests that patients with chronic heart failure (HF) frequently experience cognitive impairments, but the neuroanatomical mechanism underlying these impairments remains elusive. In this retrospective study, 49 chronic HF patients and 49 healthy controls (HCs) underwent brain structural MRI scans and cognitive assessments. Cortical morphology index (cortical thickness, complexity, sulcal depth and gyrification) were evaluated. Correlations between cortical morphology and cognitive scores and clinical variables were explored. Logistic regression analysis was employed to identify risk factors for predicting 3-year major adverse cardiovascular events. Compared with HCs, patients with chronic HF exhibited decreased cognitive scores (p < .001) and decreased cortical thickness, sulcal depth and gyrification in brain regions involved cognition, sensorimotor, autonomic nervous system (family-wise error correction, all p values <.05). Notably, HF duration and New York Heart Association (NYHA) demonstrated negative correlations with abnormal cortex morphology, particularly HF duration and thickness in left precentral gyrus (r = -.387, p = .006). Cortical morphology characteristics exhibited positive associations with global cognition, particularly cortical thickness in left pars opercularis (r = .476, p < .001). NYHA class is an independent risk factor for adverse outcome (p = .001). The observed correlation between abnormal cortical morphology and global cognition suggested that cortical morphology may serve as a promising imaging biomarker and provide insights into neuroanatomical underpinnings of cognitive impairment in patients with chronic HF.
Collapse
Affiliation(s)
- Yu Ting Liu
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yu Ting Yang
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chun Xiang Tang
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jun Qing Ma
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiang Kong
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jian Hua Li
- Department of Cardiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yan Ming Li
- Department of Cardiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shu Yu Liu
- Department of Cardiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chang Sheng Zhou
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yun Fei Wang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Long Jiang Zhang
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
30
|
Munsch F, Planes D, Fukutomi H, Marnat G, Courret T, Micard E, Chen B, Seners P, Dubos J, Planche V, Coupé P, Dousset V, Lapergue B, Olivot JM, Sibon I, Thiebaut De Schotten M, Tourdias T. Dynamic Evolution of Infarct Volumes at MRI in Ischemic Stroke Due to Large Vessel Occlusion. Neurology 2024; 102:e209427. [PMID: 38815232 DOI: 10.1212/wnl.0000000000209427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND AND OBJECTIVES The typical infarct volume trajectories in stroke patients, categorized as slow or fast progressors, remain largely unknown. This study aimed to reveal the characteristic spatiotemporal evolutions of infarct volumes caused by large vessel occlusion (LVO) and show that such growth charts help anticipate clinical outcomes. METHODS We conducted a secondary analysis from prospectively collected databases (FRAME, 2017-2019; ETIS, 2015-2022). We selected acute MRI data from anterior LVO stroke patients with witnessed onset, which were divided into training and independent validation datasets. In the training dataset, using Gaussian mixture analysis, we classified the patients into 3 growth groups based on their rate of infarct growth (diffusion volume/time-to-imaging). Subsequently, we extrapolated pseudo-longitudinal models of infarct growth for each group and generated sequential frequency maps to highlight the spatial distribution of infarct growth. We used these charts to attribute a growth group to the independent patients from the validation dataset. We compared their 3-month modified Rankin scale (mRS) with the predicted values based on a multivariable regression model from the training dataset that used growth group as an independent variable. RESULTS We included 804 patients (median age 73.0 years [interquartile range 61.2-82.0 years]; 409 men). The training dataset revealed nonsupervised clustering into 11% (74/703) slow, 62% (437/703) intermediate, and 27% (192/703) fast progressors. Infarct volume evolutions were best fitted with a linear (r = 0.809; p < 0.001), cubic (r = 0.471; p < 0.001), and power (r = 0.63; p < 0.001) function for the slow, intermediate, and fast progressors, respectively. Notably, the deep nuclei and insular cortex were rapidly affected in the intermediate and fast groups with further cortical involvement in the fast group. The variable growth group significantly predicted the 3-month mRS (multivariate odds ratio 0.51; 95% CI 0.37-0.72, p < 0.0001) in the training dataset, yielding a mean area under the receiver operating characteristic curve of 0.78 (95% CI 0.66-0.88) in the independent validation dataset. DISCUSSION We revealed spatiotemporal archetype dynamic evolutions following LVO stroke according to 3 growth phenotypes called slow, intermediate, and fast progressors, providing insight into anticipating clinical outcome. We expect this could help in designing neuroprotective trials aiming at modulating infarct growth before EVT.
Collapse
Affiliation(s)
- Fanny Munsch
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - David Planes
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Hikaru Fukutomi
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Gaultier Marnat
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Thomas Courret
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Emilien Micard
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Bailiang Chen
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Pierre Seners
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Johanna Dubos
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Vincent Planche
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Pierrick Coupé
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Vincent Dousset
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Bertrand Lapergue
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Jean Marc Olivot
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Igor Sibon
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Michel Thiebaut De Schotten
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| | - Thomas Tourdias
- From the Institut de Bio-imagerie IBIO (F.M., J.D., V.D., T.T.), University Bordeaux; Neuroimagerie Diagnostique et Thérapeutique (D.P., G.M., T.C., V.D., T.T.), CHU de Bordeaux, France; Kansai Electric Power Hospital (H.F.), Osaka, Japan; Inserm CIC-IT U1433 (E.M., B.C.), CHRU Nancy; Institut de Psychiatrie et Neurosciences de Paris (IPNP) (P.S.), INSERM U1266; Département de Neurologie (P.S.), Hopital Fondation Rothschild, Paris; Institut des Maladies Neurodégénératives (V.P.), CNRS, UMR 5293, Bordeaux INP (P.C.), LABRI, CNRS, UMR5800, and Neurocentre Magendie (V.D., T.T.), INSERM U1215, Univ. Bordeaux; Service de Neurologie et Unité de Neuro Vasculaire (B.L.), Hôpital FOCH, Suresnes; Unité Neurovasculaire (J.M.O.), CHU de Toulouse; Unité Neurovasculaire (I.S.), CHU de Bordeaux; CNRS (M.T.D.S.), UMR-5293, Univ. Bordeaux; and Brain Connectivity and Behaviour Laboratory (M.T.D.S.), Paris, France
| |
Collapse
|
31
|
Schneider C, Prokopiou PC, Papp KV, Engels‐Domínguez N, Hsieh S, Juneau TA, Schultz AP, Rentz DM, Sperling RA, Johnson KA, Jacobs HIL. Atrophy links lower novelty-related locus coeruleus connectivity to cognitive decline in preclinical AD. Alzheimers Dement 2024; 20:3958-3971. [PMID: 38676563 PMCID: PMC11180940 DOI: 10.1002/alz.13839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/29/2024] [Accepted: 03/08/2024] [Indexed: 04/29/2024]
Abstract
INTRODUCTION Animal research has shown that tau pathology in the locus coeruleus (LC) is associated with reduced norepinephrine signaling, lower projection density to the medial temporal lobe (MTL), atrophy, and cognitive impairment. We investigated the contribution of LC-MTL functional connectivity (FCLC-MTL) on cortical atrophy across Braak stage regions and its impact on cognition. METHODS We analyzed functional magnetic resonance imaging and amyloid beta (Aβ) positron emission tomography data from 128 cognitively normal participants, associating novelty-related FCLC-MTL with longitudinal atrophy and cognition with and without Aβ moderation. RESULTS Cross-sectionally, lower FCLC-MTL was associated with atrophy in Braak stage II regions. Longitudinally, atrophy in Braak stage 2 to 4 regions related to lower baseline FCLC-MTL at elevated levels of Aβ, but not to other regions. Atrophy in Braak stage 2 regions mediated the relation between FCLC-MTL and subsequent cognitive decline. DISCUSSION FCLC-MTL is implicated in Aβ-related cortical atrophy, suggesting that LC-MTL connectivity could confer neuroprotective effects in preclinical AD. HIGHLIGHTS Novelty-related functional magnetic resonance imaging (fMRI) LC-medial temporal lobe (MTL) connectivity links to longitudinal Aβ-dependent atrophy. This relationship extended to higher Braak stage regions with increasing Aβ burden. Longitudinal MTL atrophy mediated the LC-MTL connectivity-cognition relationship. Our findings mirror the animal data on MTL atrophy following NE signal dysfunction.
Collapse
Affiliation(s)
- Christoph Schneider
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Prokopis C. Prokopiou
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Kathryn V. Papp
- Harvard Medical SchoolBostonMassachusettsUSA
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Nina Engels‐Domínguez
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General HospitalBostonMassachusettsUSA
- Faculty of HealthMedicine and Life SciencesSchool for Mental Health and NeuroscienceAlzheimer Centre LimburgMaastricht University, MDMaastrichtThe Netherlands
| | - Stephanie Hsieh
- The Athinoula A. Martinos Center for Biomedical ImagingDepartment of RadiologyMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Truley A. Juneau
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Aaron P. Schultz
- The Athinoula A. Martinos Center for Biomedical ImagingDepartment of RadiologyMassachusetts General HospitalCharlestownMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Dorene M. Rentz
- Harvard Medical SchoolBostonMassachusettsUSA
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Reisa A. Sperling
- Harvard Medical SchoolBostonMassachusettsUSA
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Keith A. Johnson
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Heidi I. L. Jacobs
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
- Faculty of HealthMedicine and Life SciencesSchool for Mental Health and NeuroscienceAlzheimer Centre LimburgMaastricht University, MDMaastrichtThe Netherlands
- The Athinoula A. Martinos Center for Biomedical ImagingDepartment of RadiologyMassachusetts General HospitalCharlestownMassachusettsUSA
| |
Collapse
|
32
|
Romascano D, Rebsamen M, Radojewski P, Blattner T, McKinley R, Wiest R, Rummel C. Cortical thickness and grey-matter volume anomaly detection in individual MRI scans: Comparison of two methods. Neuroimage Clin 2024; 43:103624. [PMID: 38823248 PMCID: PMC11168488 DOI: 10.1016/j.nicl.2024.103624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/21/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024]
Abstract
Over the past decades, morphometric analysis of brain MRI has contributed substantially to the understanding of healthy brain structure, development and aging as well as to improved characterisation of disease related pathologies. Certified commercial tools based on normative modeling of these metrics are meanwhile available for diagnostic purposes, but they are cost intensive and their clinical evaluation is still in its infancy. Here we have compared the performance of "ScanOMetrics", an open-source research-level tool for detection of statistical anomalies in individual MRI scans, depending on whether it is operated on the output of FreeSurfer or of the deep learning based brain morphometry tool DL + DiReCT. When applied to the public OASIS3 dataset, containing patients with Alzheimer's disease (AD) and healthy controls (HC), cortical thickness anomalies in patient scans were mainly detected in regions that are known as predilection areas of cortical atrophy in AD, regardless of the software used for extraction of the metrics. By contrast, anomaly detections in HCs were up to twenty-fold reduced and spatially unspecific using both DL + DiReCT and FreeSurfer. Progression of the atrophy pattern with clinical dementia rating (CDR) was clearly observable with both methods. DL + DiReCT provided results in less than 25 min, more than 15 times faster than FreeSurfer. This difference in computation time might be relevant when considering application of this or similar methodology as diagnostic decision support for neuroradiologists.
Collapse
Affiliation(s)
- David Romascano
- Support Center for Advanced Neuroimaging (SCAN), University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, University Hospital Bern, CH-3010 Bern, Switzerland; Danish Research Center for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
| | - Michael Rebsamen
- Support Center for Advanced Neuroimaging (SCAN), University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, University Hospital Bern, CH-3010 Bern, Switzerland
| | - Piotr Radojewski
- Support Center for Advanced Neuroimaging (SCAN), University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, University Hospital Bern, CH-3010 Bern, Switzerland; Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, sitem-insel, Bern, Switzerland
| | - Timo Blattner
- Support Center for Advanced Neuroimaging (SCAN), University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, University Hospital Bern, CH-3010 Bern, Switzerland
| | - Richard McKinley
- Support Center for Advanced Neuroimaging (SCAN), University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, University Hospital Bern, CH-3010 Bern, Switzerland
| | - Roland Wiest
- Support Center for Advanced Neuroimaging (SCAN), University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, University Hospital Bern, CH-3010 Bern, Switzerland; Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, sitem-insel, Bern, Switzerland
| | - Christian Rummel
- Support Center for Advanced Neuroimaging (SCAN), University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, University Hospital Bern, CH-3010 Bern, Switzerland; European Campus Rottal-Inn, Technische Hochschule Deggendorf, Max-Breiherr-Straße 32, D-84347 Pfarrkirchen, Germany.
| |
Collapse
|
33
|
Wang B. Exploring intricate connectivity patterns for cognitive functioning and neurological disorders: incorporating frequency-domain NC method into fMRI analysis. Cereb Cortex 2024; 34:bhae195. [PMID: 38741270 DOI: 10.1093/cercor/bhae195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
This study extends the application of the frequency-domain new causality method to functional magnetic resonance imaging analysis. Strong causality, weak causality, balanced causality, cyclic causality, and transitivity causality were constructed to simulate varying degrees of causal associations among multivariate functional-magnetic-resonance-imaging blood-oxygen-level-dependent signals. Data from 1,252 groups of individuals with different degrees of cognitive impairment were collected. The frequency-domain new causality method was employed to construct directed efficient connectivity networks of the brain, analyze the statistical characteristics of topological variations in brain regions related to cognitive impairment, and utilize these characteristics as features for training a deep learning model. The results demonstrated that the frequency-domain new causality method accurately detected causal associations among simulated signals of different degrees. The deep learning tests also confirmed the superior performance of new causality, surpassing the other three methods in terms of accuracy, precision, and recall rates. Furthermore, consistent significant differences were observed in the brain efficiency networks, where several subregions defined by the multimodal parcellation method of Human Connectome Project simultaneously appeared in the topological statistical results of different patient groups. This suggests a significant association between these fine-grained cortical subregions, driven by multimodal data segmentation, and human cognitive function, making them potential biomarkers for further analysis of Alzheimer's disease.
Collapse
Affiliation(s)
- Bocheng Wang
- College of Media Engineering, Communication University of Zhejiang, 998 Xue Yuan Street, Qiantang District, Hangzhou, Zhejiang 310018, China
| |
Collapse
|
34
|
Kim HH, Kwon MJ, Jo S, Park JE, Kim JW, Kim JH, Kim SE, Kim KW, Han JW. Exploration of neuroanatomical characteristics to differentiate prodromal Alzheimer's disease from cognitively unimpaired amyloid-positive individuals. Sci Rep 2024; 14:10083. [PMID: 38698190 PMCID: PMC11066072 DOI: 10.1038/s41598-024-60843-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 04/28/2024] [Indexed: 05/05/2024] Open
Abstract
Differentiating clinical stages based solely on positive findings from amyloid PET is challenging. We aimed to investigate the neuroanatomical characteristics at the whole-brain level that differentiate prodromal Alzheimer's disease (AD) from cognitively unimpaired amyloid-positive individuals (CU A+) in relation to amyloid deposition and regional atrophy. We included 45 CU A+ participants and 135 participants with amyloid-positive prodromal AD matched 1:3 by age, sex, and education. All participants underwent 18F-florbetaben positron emission tomography and 3D structural T1-weighted magnetic resonance imaging. We compared the standardized uptake value ratios (SUVRs) and volumes in 80 regions of interest (ROIs) between CU A+ and prodromal AD groups using independent t-tests, and employed the least absolute selection and shrinkage operator (LASSO) logistic regression model to identify ROIs associated with prodromal AD in relation to amyloid deposition, regional atrophy, and their interaction. After applying False Discovery Rate correction at < 0.1, there were no differences in global and regional SUVR between CU A+ and prodromal AD groups. Regional volume differences between the two groups were observed in the amygdala, hippocampus, entorhinal cortex, insula, parahippocampal gyrus, and inferior temporal and parietal cortices. LASSO logistic regression model showed significant associations between prodromal AD and atrophy in the entorhinal cortex, inferior parietal cortex, both amygdalae, and left hippocampus. The mean SUVR in the right superior parietal cortex (beta coefficient = 0.0172) and its interaction with the regional volume (0.0672) were also selected in the LASSO model. The mean SUVR in the right superior parietal cortex was associated with an increased likelihood of prodromal AD (Odds ratio [OR] 1.602, p = 0.014), particularly in participants with lower regional volume (OR 3.389, p < 0.001). Only regional volume differences, not amyloid deposition, were observed between CU A+ and prodromal AD. The reduced volume in the superior parietal cortex may play a significant role in the progression to prodromal AD through its interaction with amyloid deposition in that region.
Collapse
Affiliation(s)
- Hak Hyeon Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
- Department of Psychiatry, College of Medicine, Seoul National University, Seoul, South Korea
| | - Min Jeong Kwon
- Department of Brain and Cognitive Science, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Sungman Jo
- Department of Health Science and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Ji Eun Park
- Department of Brain and Cognitive Science, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Ji Won Kim
- Department of Brain and Cognitive Science, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Jae Hyoung Kim
- Department of Radiology, Seoul National University Bundang Hospital, College of Medicine, Seoul National University, Seongnam-si, Gyeonggi-do, South Korea
| | - Sang Eun Kim
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, College of Medicine, Seoul National University, Seongnam-si, Gyeonggi-do, Korea
- Center for Nanomolecular Imaging and Innovative Drug Development, Advanced Institutes of Convergence Technology, Suwon, Republic of Korea
| | - Ki Woong Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
- Department of Psychiatry, College of Medicine, Seoul National University, Seoul, South Korea
- Department of Brain and Cognitive Science, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Department of Health Science and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Ji Won Han
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea.
- Department of Psychiatry, College of Medicine, Seoul National University, Seoul, South Korea.
| |
Collapse
|
35
|
Koubiyr I, Yamamoto T, Blyau S, Kamroui RA, Mansencal B, Planche V, Petit L, Saranathan M, Casey R, Ruet A, Brochet B, Manjón JV, Dousset V, Coupé P, Tourdias T. Vulnerability of Thalamic Nuclei at CSF Interface During the Entire Course of Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200222. [PMID: 38635941 PMCID: PMC11087027 DOI: 10.1212/nxi.0000000000200222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 01/19/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND AND OBJECTIVES Thalamic atrophy can be used as a proxy for neurodegeneration in multiple sclerosis (MS). Some data point toward thalamic nuclei that could be affected more than others. However, the dynamic of their changes during MS evolution and the mechanisms driving their differential alterations are still uncertain. METHODS We paired a large cohort of 1,123 patients with MS with the same number of healthy controls, all scanned with conventional 3D-T1 MRI. To highlight the main atrophic regions at the thalamic nuclei level, we validated a segmentation strategy consisting of deep learning-based synthesis of sequences, which were used for automatic multiatlas segmentation. Then, through a lifespan-based approach, we could model the dynamics of the 4 main thalamic nuclei groups. RESULTS All analyses converged toward a higher rate of atrophy for the posterior and medial groups compared with the anterior and lateral groups. We also demonstrated that focal MS white matter lesions were associated with atrophy of groups of nuclei when specifically located within the associated thalamocortical projections. The volumes of the most affected posterior group, but also of the anterior group, were better associated with clinical disability than the volume of the whole thalamus. DISCUSSION These findings point toward the thalamic nuclei adjacent to the third ventricle as more susceptible to neurodegeneration during the entire course of MS through potentiation of disconnection effects by regional factors. Because this information can be obtained even from standard T1-weighted MRI, this paves the way toward such an approach for future monitoring of patients with MS.
Collapse
Affiliation(s)
- Ismail Koubiyr
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| | - Takayuki Yamamoto
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| | - Simon Blyau
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| | - Reda A Kamroui
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| | - Boris Mansencal
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| | - Vincent Planche
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| | - Laurent Petit
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| | - Manojkumar Saranathan
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| | - Romain Casey
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| | - Aurélie Ruet
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| | - Bruno Brochet
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| | - José V Manjón
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| | - Vincent Dousset
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| | - Pierrick Coupé
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| | - Thomas Tourdias
- From the University of Bordeaux (I.K., T.Y., A.R., B.B., V.D., T.T.), INSERM, Neurocentre Magendie, U1215; Neuroimagerie diagnostique et thérapeutique (S.B.), CHU de Bordeaux; University of Bordeaux (R.A.K., B.M., P.C.), CNRS, Bordeaux INP, LABRI, UMR5800, Talence; Univ. Bordeaux (V.P.), CNRS, IMN, UMR 5293; Groupe d'Imagerie Neurofonctionnelle (L.P.), Institut des Maladies Neurodégénératives CNRS UMR 5293, Bordeaux, France; Department of Medical Imaging (M.S.), The University of Arizona, Tucson; Université de Lyon (R.C.), Université Claude Bernard Lyon 1, France; and Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA) (J.V.M.), Universitat Politècnica de València, Spain
| |
Collapse
|
36
|
Kozberg MG, Munting LP, Maresco LH, Auger CA, van den Berg ML, Denis de Senneville B, Hirschler L, Warnking JM, Barbier EL, Farrar CT, Greenberg SM, Bacskai BJ, van Veluw SJ. Loss of spontaneous vasomotion precedes impaired cerebrovascular reactivity and microbleeds in a mouse model of cerebral amyloid angiopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.591414. [PMID: 38746419 PMCID: PMC11092483 DOI: 10.1101/2024.04.26.591414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background Cerebral amyloid angiopathy (CAA) is a cerebral small vessel disease in which amyloid-β accumulates in vessel walls. CAA is a leading cause of symptomatic lobar intracerebral hemorrhage and an important contributor to age-related cognitive decline. Recent work has suggested that vascular dysfunction may precede symptomatic stages of CAA, and that spontaneous slow oscillations in arteriolar diameter (termed vasomotion), important for amyloid-β clearance, may be impaired in CAA. Methods To systematically study the progression of vascular dysfunction in CAA, we used the APP23 mouse model of amyloidosis, which is known to develop spontaneous cerebral microbleeds mimicking human CAA. Using in vivo 2-photon microscopy, we longitudinally imaged unanesthetized APP23 transgenic mice and wildtype littermates from 7 to 14 months of age, tracking amyloid-β accumulation and vasomotion in individual pial arterioles over time. MRI was used in separate groups of 12-, 18-, and 24-month-old APP23 transgenic mice and wildtype littermates to detect microbleeds and to assess cerebral blood flow and cerebrovascular reactivity with pseudo-continuous arterial spin labeling. Results We observed a significant decline in vasomotion with age in APP23 mice, while vasomotion remained unchanged in wildtype mice with age. This decline corresponded in timing to initial vascular amyloid-β deposition (∼8-10 months of age), although was more strongly correlated with age than with vascular amyloid-β burden in individual arterioles. Declines in vasomotion preceded the development of MRI-visible microbleeds and the loss of smooth muscle actin in arterioles, both of which were observed in APP23 mice by 18 months of age. Additionally, evoked cerebrovascular reactivity was intact in APP23 mice at 12 months of age, but significantly lower in APP23 mice by 24 months of age. Conclusions Our findings suggest that a decline in spontaneous vasomotion is an early, potentially pre-symptomatic, manifestation of CAA and vascular dysfunction, and a possible future treatment target.
Collapse
|
37
|
Planche V, Mansencal B, Manjon JV, Meissner WG, Tourdias T, Coupé P. Staging of progressive supranuclear palsy-Richardson syndrome using MRI brain charts for the human lifespan. Brain Commun 2024; 6:fcae055. [PMID: 38444913 PMCID: PMC10914441 DOI: 10.1093/braincomms/fcae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/22/2023] [Accepted: 02/19/2024] [Indexed: 03/07/2024] Open
Abstract
Brain charts for the human lifespan have been recently proposed to build dynamic models of brain anatomy in normal aging and various neurological conditions. They offer new possibilities to quantify neuroanatomical changes from preclinical stages to death, where longitudinal MRI data are not available. In this study, we used brain charts to model the progression of brain atrophy in progressive supranuclear palsy-Richardson syndrome. We combined multiple datasets (n = 8170 quality controlled MRI of healthy subjects from 22 cohorts covering the entire lifespan, and n = 62 MRI of progressive supranuclear palsy-Richardson syndrome patients from the Four Repeat Tauopathy Neuroimaging Initiative (4RTNI)) to extrapolate lifetime volumetric models of healthy and progressive supranuclear palsy-Richardson syndrome brain structures. We then mapped in time and space the sequential divergence between healthy and progressive supranuclear palsy-Richardson syndrome charts. We found six major consecutive stages of atrophy progression: (i) ventral diencephalon (including subthalamic nuclei, substantia nigra, and red nuclei), (ii) pallidum, (iii) brainstem, striatum and amygdala, (iv) thalamus, (v) frontal lobe, and (vi) occipital lobe. The three structures with the most severe atrophy over time were the thalamus, followed by the pallidum and the brainstem. These results match the neuropathological staging of tauopathy progression in progressive supranuclear palsy-Richardson syndrome, where the pathology is supposed to start in the pallido-nigro-luysian system and spreads rostrally via the striatum and the amygdala to the cerebral cortex, and caudally to the brainstem. This study supports the use of brain charts for the human lifespan to study the progression of neurodegenerative diseases, especially in the absence of specific biomarkers as in PSP.
Collapse
Affiliation(s)
- Vincent Planche
- Institut des Maladies Neurodégénératives, Univ. Bordeaux, CNRS, UMR 5293, F-33000 Bordeaux, France
- Centre Mémoire Ressources Recherches, Service de Neurologie des Maladies Neurodégénératives, Pôle de Neurosciences Cliniques, CHU de Bordeaux, F-33000 Bordeaux, France
| | - Boris Mansencal
- CNRS, Univ. Bordeaux, Bordeaux INP, Laboratoire Bordelais de Recherche en Informatique (LABRI), UMR5800, F-33400 Talence, France
| | - Jose V Manjon
- Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA), Universitat Politècnica de València, Camino de Vera s/n, 46022, Valencia, Spain
| | - Wassilios G Meissner
- Institut des Maladies Neurodégénératives, Univ. Bordeaux, CNRS, UMR 5293, F-33000 Bordeaux, France
- Service de Neurologie des Maladies Neurodégénératives, Réseau NS-Park/FCRIN, CHU Bordeaux, F-33000, Bordeaux, France
- Department of Medicine, Christchurch, and New Zealand Brain Research Institute, Christchurch, 8011, New Zealand
| | - Thomas Tourdias
- Inserm U1215—Neurocentre Magendie, Bordeaux F-33000, France
- Service de Neuroimagerie diagnostique et thérapeutique, CHU de Bordeaux, F-33000 Bordeaux, France
| | - Pierrick Coupé
- CNRS, Univ. Bordeaux, Bordeaux INP, Laboratoire Bordelais de Recherche en Informatique (LABRI), UMR5800, F-33400 Talence, France
| |
Collapse
|
38
|
Wrzesień A, Andrzejewski K, Jampolska M, Kaczyńska K. Respiratory Dysfunction in Alzheimer's Disease-Consequence or Underlying Cause? Applying Animal Models to the Study of Respiratory Malfunctions. Int J Mol Sci 2024; 25:2327. [PMID: 38397004 PMCID: PMC10888758 DOI: 10.3390/ijms25042327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative brain disease that is the most common cause of dementia among the elderly. In addition to dementia, which is the loss of cognitive function, including thinking, remembering, and reasoning, and behavioral abilities, AD patients also experience respiratory disturbances. The most common respiratory problems observed in AD patients are pneumonia, shortness of breath, respiratory muscle weakness, and obstructive sleep apnea (OSA). The latter is considered an outcome of Alzheimer's disease and is suggested to be a causative factor. While this narrative review addresses the bidirectional relationship between obstructive sleep apnea and Alzheimer's disease and reports on existing studies describing the most common respiratory disorders found in patients with Alzheimer's disease, its main purpose is to review all currently available studies using animal models of Alzheimer's disease to study respiratory impairments. These studies on animal models of AD are few in number but are crucial for establishing mechanisms, causation, implementing potential therapies for respiratory disorders, and ultimately applying these findings to clinical practice. This review summarizes what is already known in the context of research on respiratory disorders in animal models, while pointing out directions for future research.
Collapse
Affiliation(s)
| | | | | | - Katarzyna Kaczyńska
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.W.); (K.A.); (M.J.)
| |
Collapse
|
39
|
Davidson TL, Stevenson RJ. Vulnerability of the Hippocampus to Insults: Links to Blood-Brain Barrier Dysfunction. Int J Mol Sci 2024; 25:1991. [PMID: 38396670 PMCID: PMC10888241 DOI: 10.3390/ijms25041991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
The hippocampus is a critical brain substrate for learning and memory; events that harm the hippocampus can seriously impair mental and behavioral functioning. Hippocampal pathophysiologies have been identified as potential causes and effects of a remarkably diverse array of medical diseases, psychological disorders, and environmental sources of damage. It may be that the hippocampus is more vulnerable than other brain areas to insults that are related to these conditions. One purpose of this review is to assess the vulnerability of the hippocampus to the most prevalent types of insults in multiple biomedical domains (i.e., neuroactive pathogens, neurotoxins, neurological conditions, trauma, aging, neurodegenerative disease, acquired brain injury, mental health conditions, endocrine disorders, developmental disabilities, nutrition) and to evaluate whether these insults affect the hippocampus first and more prominently compared to other brain loci. A second purpose is to consider the role of hippocampal blood-brain barrier (BBB) breakdown in either causing or worsening the harmful effects of each insult. Recent research suggests that the hippocampal BBB is more fragile compared to other brain areas and may also be more prone to the disruption of the transport mechanisms that act to maintain the internal milieu. Moreover, a compromised BBB could be a factor that is common to many different types of insults. Our analysis indicates that the hippocampus is more vulnerable to insults compared to other parts of the brain, and that developing interventions that protect the hippocampal BBB may help to prevent or ameliorate the harmful effects of many insults on memory and cognition.
Collapse
Affiliation(s)
- Terry L. Davidson
- Department of Neuroscience, Center for Neuroscience and Behavior, American University, 4400 Massachusetts Avenue, NW, Washington, DC 20016, USA
| | | |
Collapse
|
40
|
Sebro R. Advancing Diagnostics and Patient Care: The Role of Biomarkers in Radiology. Semin Musculoskelet Radiol 2024; 28:3-13. [PMID: 38330966 DOI: 10.1055/s-0043-1776426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
The integration of biomarkers into medical practice has revolutionized the field of radiology, allowing for enhanced diagnostic accuracy, personalized treatment strategies, and improved patient care outcomes. This review offers radiologists a comprehensive understanding of the diverse applications of biomarkers in medicine. By elucidating the fundamental concepts, challenges, and recent advancements in biomarker utilization, it will serve as a bridge between the disciplines of radiology and epidemiology. Through an exploration of various biomarker types, such as imaging biomarkers, molecular biomarkers, and genetic markers, I outline their roles in disease detection, prognosis prediction, and therapeutic monitoring. I also discuss the significance of robust study designs, blinding, power and sample size calculations, performance metrics, and statistical methodologies in biomarker research. By fostering collaboration between radiologists, statisticians, and epidemiologists, I hope to accelerate the translation of biomarker discoveries into clinical practice, ultimately leading to improved patient care.
Collapse
Affiliation(s)
- Ronnie Sebro
- Department of Radiology, Center for Augmented Intelligence, Mayo Clinic, Jacksonville, Florida
- Department of Biostatistics, Center for Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
- Department of Orthopedic Surgery, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
41
|
Ambikairajah A, Khondoker M, Morris E, de Lange AG, Saleh RNM, Minihane AM, Hornberger M. Investigating the synergistic effects of hormone replacement therapy, apolipoprotein E and age on brain health in the UK Biobank. Hum Brain Mapp 2024; 45:e26612. [PMID: 38339898 PMCID: PMC10836173 DOI: 10.1002/hbm.26612] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 02/12/2024] Open
Abstract
Global prevalence of Alzheimer's Disease has a strong sex bias, with women representing approximately two-thirds of the patients. Yet, the role of sex-specific risk factors during midlife, including hormone replacement therapy (HRT) and their interaction with other major risk factors for Alzheimer's Disease, such as apolipoprotein E (APOE)-e4 genotype and age, on brain health remains unclear. We investigated the relationship between HRT (i.e., use, age of initiation and duration of use) and brain health (i.e., cognition and regional brain volumes). We then consider the multiplicative effects of HRT and APOE status (i.e., e2/e2, e2/e3, e3/e3, e3/e4 and e4/e4) via a two-way interaction and subsequently age of participants via a three-way interaction. Women from the UK Biobank with no self-reported neurological conditions were included (N = 207,595 women, mean age = 56.25 years, standard deviation = 8.01 years). Generalised linear regression models were computed to quantify the cross-sectional association between HRT and brain health, while controlling for APOE status, age, time since attending centre for completing brain health measure, surgical menopause status, smoking history, body mass index, education, physical activity, alcohol use, ethnicity, socioeconomic status, vascular/heart problems and diabetes diagnosed by doctor. Analyses of structural brain regions further controlled for scanner site. All brain volumes were normalised for head size. Two-way interactions between HRT and APOE status were modelled, in addition to three-way interactions including age. Results showed that women with the e4/e4 genotype who have used HRT had 1.82% lower hippocampal, 2.4% lower parahippocampal and 1.24% lower thalamus volumes than those with the e3/e3 genotype who had never used HRT. However, this interaction was not detected for measures of cognition. No clinically meaningful three-way interaction between APOE, HRT and age was detected when interpreted relative to the scales of the cognitive measures used and normative models of ageing for brain volumes in this sample. Differences in hippocampal volume between women with the e4/e4 genotype who have used HRT and those with the e3/e3 genotype who had never used HRT are equivalent to approximately 1-2 years of hippocampal atrophy observed in typical health ageing trajectories in midlife (i.e., 0.98%-1.41% per year). Effect sizes were consistent within APOE e4/e4 group post hoc sensitivity analyses, suggesting observed effects were not solely driven by APOE status and may, in part, be attributed to HRT use. Although, the design of this study means we cannot exclude the possibility that women who have used HRT may have a predisposition for poorer brain health.
Collapse
Affiliation(s)
- Ananthan Ambikairajah
- Discipline of Psychology, Faculty of HealthUniversity of CanberraCanberraAustralian Capital TerritoryAustralia
- Centre for Ageing Research and Translation, Faculty of HealthUniversity of CanberraCanberraAustralian Capital TerritoryAustralia
- National Centre for Epidemiology and Population HealthAustralian National UniversityCanberraAustralian Capital TerritoryAustralia
| | | | | | - Ann‐Marie G. de Lange
- Department of Clinical NeurosciencesLausanne University Hospital (CHUV) and University of LausanneLausanneSwitzerland
- Department of PsychologyUniversity of OsloOsloNorway
- Department of PsychiatryUniversity of OxfordOxfordUK
| | - Rasha N. M. Saleh
- Norwich Medical SchoolUniversity of East AngliaNorwichUK
- Department of Clinical and Chemical Pathology, Faculty of MedicineAlexandria UniversityAlexandriaEgypt
| | - Anne Marie Minihane
- Norwich Medical SchoolUniversity of East AngliaNorwichUK
- Norwich Institute of Healthy AgeingNorwichUK
| | | |
Collapse
|
42
|
DiProspero N, Sathishkumar M, Janecek J, Smith A, McMillan L, Petersen M, Tustison N, Keator DB, Doran E, Hom CL, Nguyen D, Andrews H, Krinsky‐McHale S, Brickman AM, Rosas HD, Lai F, Head E, Mapstone M, Silverman W, Lott IT, O'Bryant S, Yassa MA. Neurofilament light chain concentration mediates the association between regional medial temporal lobe structure and memory in adults with Down syndrome. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12542. [PMID: 38348178 PMCID: PMC10859879 DOI: 10.1002/dad2.12542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/03/2023] [Accepted: 09/26/2023] [Indexed: 02/15/2024]
Abstract
INTRODUCTION Virtually all people with Down syndrome (DS) develop neuropathology associated with Alzheimer's disease (AD). Atrophy of the hippocampus and entorhinal cortex (EC), as well as elevated plasma concentrations of neurofilament light chain (NfL) protein, are markers of neurodegeneration associated with late-onset AD. We hypothesized that hippocampus and EC gray matter loss and increased plasma NfL concentrations are associated with memory in adults with DS. METHODS T1-weighted structural magnetic resonance imaging (MRI) data were collected from 101 participants with DS. Hippocampus and EC volume, as well as EC subregional cortical thickness, were derived. In a subset of participants, plasma NfL concentrations and modified Cued Recall Test scores were obtained. Partial correlation and mediation were used to test relationships between medial temporal lobe (MTL) atrophy, plasma NfL, and episodic memory. RESULTS Hippocampus volume, left anterolateral EC (alEC) thickness, and plasma NfL were correlated with each other and were associated with memory. Plasma NfL mediated the relationship between left alEC thickness and memory as well as hippocampus volume and memory. DISCUSSION The relationship between MTL gray matter and memory is mediated by plasma NfL levels, suggesting a link between neurodegenerative processes underlying axonal injury and frank gray matter loss in key structures supporting episodic memory in people with DS.
Collapse
Affiliation(s)
- Natalie DiProspero
- Department of Neurobiology and BehaviorSchool of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
- Center for the Neurobiology of Learning and MemoryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Mithra Sathishkumar
- Department of Neurobiology and BehaviorSchool of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
- Center for the Neurobiology of Learning and MemoryUniversity of CaliforniaIrvineCaliforniaUSA
| | - John Janecek
- Department of Neurobiology and BehaviorSchool of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
- Center for the Neurobiology of Learning and MemoryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Anna Smith
- Department of Neurobiology and BehaviorSchool of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
- Center for the Neurobiology of Learning and MemoryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Liv McMillan
- Department of Neurobiology and BehaviorSchool of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
- Center for the Neurobiology of Learning and MemoryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Melissa Petersen
- Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Nicholas Tustison
- Department of Neurobiology and BehaviorSchool of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
- Department of RadiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - David B. Keator
- Department of Psychiatry and Behavioral SciencesSchool of MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Eric Doran
- Department of PediatricsSchool of MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Christy L. Hom
- Department of PediatricsSchool of MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Dana Nguyen
- Department of PediatricsSchool of MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Howard Andrews
- Department of PsychiatryCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Sharon Krinsky‐McHale
- Department of NeurologyCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Adam M. Brickman
- Department of NeurologyCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - H. Diana Rosas
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Florence Lai
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Elizabeth Head
- Center for the Neurobiology of Learning and MemoryUniversity of CaliforniaIrvineCaliforniaUSA
- Department of PathologySchool of MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Mark Mapstone
- Center for the Neurobiology of Learning and MemoryUniversity of CaliforniaIrvineCaliforniaUSA
- Department of NeurologySchool of MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Wayne Silverman
- Department of PediatricsSchool of MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Ira T. Lott
- Department of PediatricsSchool of MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Sid O'Bryant
- Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Michael A. Yassa
- Department of Neurobiology and BehaviorSchool of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
- Center for the Neurobiology of Learning and MemoryUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesSchool of MedicineUniversity of CaliforniaIrvineCaliforniaUSA
- Department of NeurologySchool of MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| |
Collapse
|
43
|
Bunzeck N, Steiger TK, Krämer UM, Luedtke K, Marshall L, Obleser J, Tune S. Trajectories and contributing factors of neural compensation in healthy and pathological aging. Neurosci Biobehav Rev 2024; 156:105489. [PMID: 38040075 DOI: 10.1016/j.neubiorev.2023.105489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/07/2023] [Accepted: 11/27/2023] [Indexed: 12/03/2023]
Abstract
Neural degeneration is a hallmark of healthy aging and can be associated with specific cognitive impairments. However, neural degeneration per se is not matched by unremitting declines in cognitive abilities. Instead, middle-aged and older adults typically maintain surprisingly high levels of cognitive functioning, suggesting that the human brain can adapt to structural degeneration by neural compensation. Here, we summarize prevailing theories and recent empirical studies on neural compensation with a focus on often neglected contributing factors, such as lifestyle, metabolism and neural plasticity. We suggest that these factors moderate the relationship between structural integrity and neural compensation, maintaining psychological well-being and behavioral functioning. Finally, we discuss that a breakdown in neural compensation may pose a tipping point that distinguishes the trajectories of healthy vs pathological aging, but conjoint support from psychology and cognitive neuroscience for this alluring view is still scarce. Therefore, future experiments that target the concomitant processes of neural compensation and associated behavior will foster a comprehensive understanding of both healthy and pathological aging.
Collapse
Affiliation(s)
- Nico Bunzeck
- Department of Psychology, University of Lübeck, Germany; Center of Brain, Behavior and Metabolism, University of Lübeck, Germany.
| | | | - Ulrike M Krämer
- Department of Psychology, University of Lübeck, Germany; Center of Brain, Behavior and Metabolism, University of Lübeck, Germany; Department of Neurology, University of Lübeck, Lübeck, Germany
| | - Kerstin Luedtke
- Institute of Health Sciences, Department of Physiotherapy, University of Lübeck, Germany
| | - Lisa Marshall
- Center of Brain, Behavior and Metabolism, University of Lübeck, Germany; Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany
| | - Jonas Obleser
- Department of Psychology, University of Lübeck, Germany; Center of Brain, Behavior and Metabolism, University of Lübeck, Germany
| | - Sarah Tune
- Department of Psychology, University of Lübeck, Germany; Center of Brain, Behavior and Metabolism, University of Lübeck, Germany
| |
Collapse
|
44
|
Abstract
Maintaining diversity in drug development in research into Alzheimer's disease (AD) is necessary to avoid over-reliance on targeting AD neuropathology. Treatments that reduce or prevent the generation of oxidative stress, frequently cited for its causal role in the aging process and AD, could be useful in at-risk populations or diagnosed AD patients. However, in this review, it is argued that clinical research into antioxidants in AD could provide more useful feedback as to the therapeutic value of the oxidative stress theory of AD. Improving comparability between randomized controlled trials (RCTs) is vital from a waste-reduction and priority-setting point of view for AD clinical research. For as well as attempting to improve meaningful outcomes for patients, RCTs of antioxidants in AD should strive to maximize the extraction of clinically useful information and actionable feedback from trial outcomes. Solutions to maximize information flow from RCTs of antioxidants in AD are offered here in the form of checklist questions to improve ongoing and future trials centered around the following dimensions: adhesion to reporting guidelines like CONSORT, biomarker enrichment, simple tests of treatment, and innovative trial design.
Collapse
Affiliation(s)
- Timothy Daly
- Science Norms Democracy UMR 8011, Sorbonne Université, Paris, France
- Bioethics Program, FLACSO Argentina, Buenos Aires, Argentina
| |
Collapse
|
45
|
Whiteside DJ, Holland N, Tsvetanov KA, Mak E, Malpetti M, Savulich G, Jones PS, Naessens M, Rouse MA, Fryer TD, Hong YT, Aigbirhio FI, Mulroy E, Bhatia KP, Rittman T, O'Brien JT, Rowe JB. Synaptic density affects clinical severity via network dysfunction in syndromes associated with frontotemporal lobar degeneration. Nat Commun 2023; 14:8458. [PMID: 38114493 PMCID: PMC10730886 DOI: 10.1038/s41467-023-44307-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023] Open
Abstract
There is extensive synaptic loss from frontotemporal lobar degeneration, in preclinical models and human in vivo and post mortem studies. Understanding the consequences of synaptic loss for network function is important to support translational models and guide future therapeutic strategies. To examine this relationship, we recruited 55 participants with syndromes associated with frontotemporal lobar degeneration and 24 healthy controls. We measured synaptic density with positron emission tomography using the radioligand [11C]UCB-J, which binds to the presynaptic vesicle glycoprotein SV2A, neurite dispersion with diffusion magnetic resonance imaging, and network function with task-free magnetic resonance imaging functional connectivity. Synaptic density and neurite dispersion in patients was associated with reduced connectivity beyond atrophy. Functional connectivity moderated the relationship between synaptic density and clinical severity. Our findings confirm the importance of synaptic loss in frontotemporal lobar degeneration syndromes, and the resulting effect on behaviour as a function of abnormal connectivity.
Collapse
Affiliation(s)
- David J Whiteside
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Negin Holland
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Kamen A Tsvetanov
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Elijah Mak
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - George Savulich
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - P Simon Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Michelle Naessens
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Matthew A Rouse
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Young T Hong
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Franklin I Aigbirhio
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Eoin Mulroy
- UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Kailash P Bhatia
- UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Timothy Rittman
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - John T O'Brien
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
46
|
Mitsunaga S, Fujito N, Nakaoka H, Imazeki R, Nagata E, Inoue I. Detection of APP gene recombinant in human blood plasma. Sci Rep 2023; 13:21703. [PMID: 38066066 PMCID: PMC10709617 DOI: 10.1038/s41598-023-48993-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
The pathogenesis of Alzheimer's disease (AD) is believed to involve the accumulation of amyloid-β in the brain, which is produced by the sequential cleavage of amyloid precursor protein (APP) by β-secretase and γ-secretase. Recently, analysis of genomic DNA and mRNA from postmortem brain neurons has revealed intra-exonic recombinants of APP (gencDNA), which have been implicated in the accumulation of amyloid-β. In this study, we computationally analyzed publicly available sequence data (SRA) using probe sequences we constructed to screen APP gencDNAs. APP gencDNAs were detected in SRAs constructed from both genomic DNA and RNA obtained from the postmortem brain and in the SRA constructed from plasma cell-free mRNA (cf-mRNA). The SRA constructed from plasma cf-mRNA showed a significant difference in the number of APP gencDNA reads between SAD and NCI: the p-value from the Mann-Whitney U test was 5.14 × 10-6. The transcripts were also found in circulating nucleic acids (CNA) from our plasma samples with NGS analysis. These data indicate that transcripts of APP gencDNA can be detected in blood plasma and suggest the possibility of using them as blood biomarkers for Alzheimer's disease.
Collapse
Affiliation(s)
- Shigeki Mitsunaga
- Laboratory of Human Genetics, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540, Japan.
| | - Naoko Fujito
- Laboratory of Human Genetics, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540, Japan
- Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Mishima, 411-8540, Japan
| | - Hirofumi Nakaoka
- Department of Cancer Genome Research, Sasaki Institute, Sasaki Foundation, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Ryoko Imazeki
- Department of Neurology, Tokai University School of Medicine, Isehara, Japan
| | - Eiichiro Nagata
- Department of Neurology, Tokai University School of Medicine, Isehara, Japan
| | - Ituro Inoue
- Laboratory of Human Genetics, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540, Japan.
| |
Collapse
|
47
|
Coupé P, Planche V, Mansencal B, Kamroui RA, Koubiyr I, Manjòn JV, Tourdias T. Lifespan neurodegeneration of the human brain in multiple sclerosis. Hum Brain Mapp 2023; 44:5602-5611. [PMID: 37615064 PMCID: PMC10619394 DOI: 10.1002/hbm.26464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/17/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023] Open
Abstract
Atrophy related to multiple sclerosis (MS) has been found at the early stages of the disease. However, the archetype dynamic trajectories of the neurodegenerative process, even prior to clinical diagnosis, remain unknown. We modeled the volumetric trajectories of brain structures across the entire lifespan using 40,944 subjects (38,295 healthy controls and 2649 MS patients). Then, we estimated the chronological progression of MS by assessing the divergence of lifespan trajectories between normal brain charts and MS brain charts. Chronologically, the first affected structure was the thalamus, then the putamen and the pallidum (around 4 years later), followed by the ventral diencephalon (around 7 years after thalamus) and finally the brainstem (around 9 years after thalamus). To a lesser extent, the anterior cingulate gyrus, insular cortex, occipital pole, caudate and hippocampus were impacted. Finally, the precuneus and accumbens nuclei exhibited a limited atrophy pattern. Subcortical atrophy was more pronounced than cortical atrophy. The thalamus was the most impacted structure with a very early divergence in life. Our experiments showed that lifespan models of most impacted structures could be an important tool for future preclinical/prodromal prognosis and monitoring of MS.
Collapse
Affiliation(s)
| | - Vincent Planche
- Univ. Bordeaux, CNRSBordeauxFrance
- Centre Mémoire Ressources Recherches, Pôle de Neurosciences Cliniques, CHU de BordeauxBordeauxFrance
| | | | | | - Ismail Koubiyr
- Inserm U1215 ‐ Neurocentre MagendieBordeauxFrance
- Service de Neuroimagerie diagnostique et thérapeutique, CHU de BordeauxBordeauxFrance
| | - José V. Manjòn
- Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA), Universitat Politècnica de ValènciaValenciaSpain
| | - Thomas Tourdias
- Inserm U1215 ‐ Neurocentre MagendieBordeauxFrance
- Service de Neuroimagerie diagnostique et thérapeutique, CHU de BordeauxBordeauxFrance
| |
Collapse
|
48
|
Jobin B, Boller B, Frasnelli J. Smaller grey matter volume in the central olfactory system in mild cognitive impairment. Exp Gerontol 2023; 183:112325. [PMID: 37952649 DOI: 10.1016/j.exger.2023.112325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
One of the major challenges in the diagnosis of Alzheimer's disease (AD) is to increase the specificity of the early diagnosis. While episodic memory impairment is a sensitive AD marker, other measures are needed to improve diagnostic specificity. A promising biomarker might be a cerebral atrophy of the central olfactory processing areas in the early stages of the disease since an impairment of olfactory identification is present at the clinical stage of AD. Our goal was therefore, (1) to evaluate the grey matter volume (GMV) of central olfactory processing regions in prodromal AD and (2) to assess its association with episodic memory. We included 34 cognitively normal healthy controls (HC), 92 individuals with subjective cognitive decline (SCD), and 40 with mild cognitive impairment (MCI). We performed regions of interest analysis (ROI) using two different approaches, allowing to extract GMV from (1) atlas-based anatomical ROIs and from (2) functional and non-functional subregions of these ROIs (olfactory ROIs and non-olfactory ROIs). Participants with MCI exhibited smaller olfactory ROIs GMV, including significant reductions in the piriform cortex, amygdala, entorhinal cortex, and left hippocampus compared to other groups (p ≤ 0.05, corrected). No significant effect was found regarding anatomical or non-olfactory ROIs GMV. The left hippocampus olfactory ROI GMV was correlated with episodic memory performance (p < 0.05 corrected). Limbic/medial-temporal olfactory processing areas are specifically atrophied at the MCI stage, and the degree of atrophy might predict cognitive decline in AD early stages.
Collapse
Affiliation(s)
- Benoît Jobin
- Department of Psychology, Université du Québec à Trois-Rivières, Qc, Canada; Research Centre of the Institut universitaire de Gériatrie de Montréal, Qc, Canada; Research Centre of the Hôpital du Sacré-Cœur de Montréal, Qc, Canada.
| | - Benjamin Boller
- Department of Psychology, Université du Québec à Trois-Rivières, Qc, Canada; Research Centre of the Institut universitaire de Gériatrie de Montréal, Qc, Canada
| | - Johannes Frasnelli
- Research Centre of the Hôpital du Sacré-Cœur de Montréal, Qc, Canada; Department of Anatomy, Université du Québec à Trois-Rivières, Qc, Canada
| |
Collapse
|
49
|
Mieling M, Meier H, Bunzeck N. Structural degeneration of the nucleus basalis of Meynert in mild cognitive impairment and Alzheimer's disease - Evidence from an MRI-based meta-analysis. Neurosci Biobehav Rev 2023; 154:105393. [PMID: 37717861 DOI: 10.1016/j.neubiorev.2023.105393] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/17/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023]
Abstract
Recent models of Alzheimer's disease (AD) suggest that neuropathological changes of the medial temporal lobe, especially entorhinal cortex, are preceded by degenerations of the cholinergic Nucleus basalis of Meynert (NbM). Evidence from imaging studies in humans, however, is limited. Therefore, we performed an activation-likelihood estimation meta-analysis on whole brain voxel-based morphometry (VBM) MRI data from 54 experiments and 2581 subjects in total. It revealed, compared to healthy older controls, reduced gray matter in the bilateral NbM in AD, but only limited evidence for such an effect in patients with mild cognitive impairment (MCI), which typically precedes AD. Both patient groups showed less gray matter in the amygdala and hippocampus, with hints towards more pronounced amygdala effects in AD. We discuss our findings in the context of studies that highlight the importance of the cholinergic basal forebrain in learning and memory throughout the lifespan, and conclude that they are partly compatible with pathological staging models suggesting initial and pronounced structural degenerations within the NbM in the progression of AD.
Collapse
Affiliation(s)
- Marthe Mieling
- Department of Psychology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Hannah Meier
- Department of Psychology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Nico Bunzeck
- Department of Psychology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany; Center of Brain, Behavior and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany.
| |
Collapse
|
50
|
Rosewood TJ, Nho K, Risacher SL, Gao S, Shen L, Foroud T, Saykin AJ. Genome-Wide Association Analysis across Endophenotypes in Alzheimer's Disease: Main Effects and Disease Stage-Specific Interactions. Genes (Basel) 2023; 14:2010. [PMID: 38002954 PMCID: PMC10671827 DOI: 10.3390/genes14112010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
The underlying genetic susceptibility for Alzheimer's disease (AD) is not yet fully understood. The heterogeneous nature of the disease challenges genetic association studies. Endophenotype approaches can help to address this challenge by more direct interrogation of biological traits related to the disease. AD endophenotypes based on amyloid-β, tau, and neurodegeneration (A/T/N) biomarkers and cognitive performance were selected from the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort (N = 1565). A genome-wide association study (GWAS) of quantitative phenotypes was performed using an SNP main effect and an SNP by Diagnosis interaction (SNP × DX) model to identify disease stage-specific genetic effects. Nine loci were identified as study-wide significant with one or more A/T/N endophenotypes in the main effect model, as well as additional findings significantly associated with cognitive measures. These nine loci include SNPs in or near the genes APOE, SRSF10, HLA-DQB1, XKR3, and KIAA1671. The SNP × DX model identified three study-wide significant genetic loci (BACH2, EP300, and PACRG-AS1) with a neuroprotective effect in later AD stage endophenotypes. An endophenotype approach identified novel genetic associations and provided insight into the molecular mechanisms underlying the genetic associations that may otherwise be missed using conventional case-control study designs.
Collapse
Affiliation(s)
- Thea J. Rosewood
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kwangsik Nho
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- School of Informatics and Computing, Indiana University, Indianapolis, IN 46202, USA
| | - Shannon L. Risacher
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sujuan Gao
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, The Perelman School of Medicine, Philadelphia, PA 19104, USA;
| | - Tatiana Foroud
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andrew J. Saykin
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|