51
|
Zhou TD, Zhang Z, Balachandrasekaran A, Raji CA, Becker JT, Kuller LH, Ge Y, Lopez OL, Dai W, Gach HM. Prospective Longitudinal Perfusion in Probable Alzheimer's Disease Correlated with Atrophy in Temporal Lobe. Aging Dis 2024; 15:1855-1871. [PMID: 37196135 PMCID: PMC11272196 DOI: 10.14336/ad.2023.0430] [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: 02/23/2023] [Accepted: 04/30/2023] [Indexed: 05/19/2023] Open
Abstract
Reduced cerebral blood flow (CBF) in the temporoparietal region and gray matter volumes (GMVs) in the temporal lobe were previously reported in patients with mild cognitive impairment (MCI) and Alzheimer's disease (AD). However, the temporal relationship between reductions in CBF and GMVs requires further investigation. This study sought to determine if reduced CBF is associated with reduced GMVs, or vice versa. Data came from 148 volunteers of the Cardiovascular Health Study Cognition Study (CHS-CS), including 58 normal controls (NC), 50 MCI, and 40 AD who had perfusion and structural MRIs during 2002-2003 (Time 2). Sixty-three of the 148 volunteers had follow-up perfusion and structural MRIs (Time 3). Forty out of the 63 volunteers received prior structural MRIs during 1997-1999 (Time 1). The relationships between GMVs and subsequent CBF changes, and between CBF and subsequent GMV changes were investigated. At Time 2, we observed smaller GMVs (p<0.05) in the temporal pole region in AD compared to NC and MCI. We also found associations between: (1) temporal pole GMVs at Time 2 and subsequent declines in CBF in this region (p=0.0014) and in the temporoparietal region (p=0.0032); (2) hippocampal GMVs at Time 2 and subsequent declines in CBF in the temporoparietal region (p=0.012); and (3) temporal pole CBF at Time 2 and subsequent changes in GMV in this region (p = 0.011). Therefore, hypoperfusion in the temporal pole may be an early event driving its atrophy. Perfusion declines in the temporoparietal and temporal pole follow atrophy in this temporal pole region.
Collapse
Affiliation(s)
- Tony D Zhou
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - Zongpai Zhang
- Computer Science, State University of New York at Binghamton, Binghamton, NY 13902, USA.
| | | | - Cyrus A Raji
- Departments of Radiology and Neurology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - James T Becker
- Departments of Psychiatry, Psychology, and Neurology, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | - Lewis H Kuller
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | - Yulin Ge
- Department of Radiology, New York University School of Medicine, New York, NY 10016, USA.
| | - Oscar L Lopez
- Departments of Neurology and Psychiatry, University of Pittsburgh, PA 15260, USA.
| | - Weiying Dai
- Computer Science, State University of New York at Binghamton, Binghamton, NY 13902, USA.
| | - H. Michael Gach
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
- Departments of Radiology and Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63110, USA.
| |
Collapse
|
52
|
Baiardi S, Hansson O, Levin J, Parchi P. In vivo detection of Alzheimer's and Lewy body disease concurrence: Clinical implications and future perspectives. Alzheimers Dement 2024; 20:5757-5770. [PMID: 38955137 PMCID: PMC11350051 DOI: 10.1002/alz.14039] [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: 02/21/2024] [Revised: 04/27/2024] [Accepted: 05/09/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION The recent introduction of seed amplification assays (SAAs) detecting misfolded α-synuclein, a pathology-specific marker for Lewy body disease (LBD), has allowed the in vivo identification and phenotypic characterization of patients with co-occurring Alzheimer's disease (AD) and LBD since the early clinical or even preclinical stage. METHODS We reviewed studies with an in vivo biomarker-based diagnosis of AD-LBD copathology. RESULTS Studies in large cohorts of cognitively impaired individuals have shown that cerebrospinal fluid (CSF) biomarkers detect the coexistence of AD and LB pathology in approximately 20%-25% of them, independently of the primary clinical diagnosis. Compared to those with pure AD, AD-LBD patients showed worse global cognition, especially in attentive/executive and visuospatial functions, and worse motor functions. In cognitively unimpaired individuals, concurrent AD-LBD pathologies predicted longitudinal cognitive progression with faster worsening of global cognition, memory, and attentive/executive functions. DISCUSSION Future research studies aiming for a better precision medicine approach should develop SAAs further to reach a quantitative evaluation or staging of each underlying pathology using a single biofluid sample. HIGHLIGHTS α-Synuclein seed amplification assays (SAAs) provide a specific marker for Lewy body disease (LBD). SAAs allow for the in vivo identification of co-occurring LBD in patients with Alzheimer's disease (AD). AD-LBD coexist in 20-25% of cognitively impaired elderly individuals, and ∼8% of those asymptomatic. Compared to pure AD, AD-LBD causes a faster worsening of cognitive functions. AD-LBD is associated with worse attentive/executive, memory, visuospatial and motor functions.
Collapse
Affiliation(s)
- Simone Baiardi
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| | - Oskar Hansson
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöFaculty of MedicineLund UniversityLundSweden
- Memory ClinicSkåne University HospitalLundSweden
| | - Johannes Levin
- Department of NeurologyLudwig‐Maximilians‐University MunichMunichGermany
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Munich Cluster of Systems Neurology (SyNergy)MunichGermany
| | - Piero Parchi
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| |
Collapse
|
53
|
Bermudez C, Lesnick TG, More SS, Ramanan VK, Knopman DS, Rabinstein AA, Cogswell PM, Jack CR, Vemuri P, Petersen RC, Graff-Radford J, Chen JJ. Optical Coherence Tomography Angiography Retinal Imaging Associations With Burden of Small Vessel Disease and Amyloid Positivity in the Brain. J Neuroophthalmol 2024:00041327-990000000-00691. [PMID: 39085998 DOI: 10.1097/wno.0000000000002230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
BACKGROUND Alzheimer disease (AD) and other dementias are associated with vascular changes and amyloid deposition, which may be reflected as density changes in the retinal capillaries. These changes may can be directly visualized and quantified with optical coherence tomography angiography (OCTA), making OCTA a potential noninvasive preclinical biomarker of small vessel disease and amyloid positivity. Our objective was to investigate the feasibility of retinal imaging metrics as noninvasive biomarkers of small vessel disease and amyloid positivity in the brain. METHODS We investigated associations between OCTA and neuroimaging and cognitive metrics in 41 participants without dementia from the Mayo Clinic Study of Aging and Alzheimer's Disease Research Center. OCTA metrics included superficial, deep, and full retina capillary density of the fovea, parafovea, and macula as well as the area of the foveal avascular zone (FAZ). Neuroimaging metrics included a high burden of white matter hyperintensity (WMH), presence of cerebral microbleeds (CMB), lacunar infarcts, and amyloid positivity as evidenced on positron emission tomography (PET), whereas cognitive metrics included mini-mental status examination (MMSE) score. We performed generalized estimating equations to account for measurements in each eye while controlling for age and sex to estimate associations between OCTA metrics and neuroimaging and cognitive scores. RESULTS Associations between OCTA and neuroimaging metrics were restricted to the fovea. OCTA showed decreased capillary density with high burden of WMH in both the superficial (P = 0.003), deep (P = 0.004), and full retina (P = 0.01) in the fovea but not the parafovea or whole macula. Similarly, participants with amyloid PET positivity had significantly decreased capillary density in the superficial fovea (P = 0.027) and deep fovea (P = 0.03) but higher density in the superficial parafovea (P = 0.038). Participants with amyloid PET positivity also had a significantly larger FAZ (P = 0.031), whereas in those with high WMH burden the difference did not reach statistical significance (P = 0.075). There was also a positive association between MMSE and capillary density of the full retina within the fovea (P = 0.037) and in the superficial parafovea (P = 0.046). No associations were found between OCTA metrics and presence of CMB or presence of lacunar infarcts. CONCLUSION The associations of lower foveal capillary density with cerebral WMH and amyloid positivity suggest that further research is warranted to evaluate for shared mechanisms of disease between small vessel disease and AD pathologies.
Collapse
Affiliation(s)
- Camilo Bermudez
- Department of Neurology (CB, VKR, DSK, AAR, RCP, JG-R, JJC), Mayo Clinic, Rochester, Minnesota; Center for Drug Design (SSM), College of Pharmacy, University of Minnesota, Minneapolis, Minnesota; Departments of Radiology (PMC, CRJ, PV) and Ophthalmology (JJC), Mayo Clinic, Rochester, Minnesota; and Department of Quantitative Health Sciences (TGL), Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Jack CR, Andrews JS, Beach TG, Buracchio T, Dunn B, Graf A, Hansson O, Ho C, Jagust W, McDade E, Molinuevo JL, Okonkwo OC, Pani L, Rafii MS, Scheltens P, Siemers E, Snyder HM, Sperling R, Teunissen CE, Carrillo MC. Revised criteria for diagnosis and staging of Alzheimer's disease: Alzheimer's Association Workgroup. Alzheimers Dement 2024; 20:5143-5169. [PMID: 38934362 PMCID: PMC11350039 DOI: 10.1002/alz.13859] [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: 02/07/2024] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 06/28/2024]
Abstract
The National Institute on Aging and the Alzheimer's Association convened three separate work groups in 2011 and single work groups in 2012 and 2018 to create recommendations for the diagnosis and characterization of Alzheimer's disease (AD). The present document updates the 2018 research framework in response to several recent developments. Defining diseases biologically, rather than based on syndromic presentation, has long been standard in many areas of medicine (e.g., oncology), and is becoming a unifying concept common to all neurodegenerative diseases, not just AD. The present document is consistent with this principle. Our intent is to present objective criteria for diagnosis and staging AD, incorporating recent advances in biomarkers, to serve as a bridge between research and clinical care. These criteria are not intended to provide step-by-step clinical practice guidelines for clinical workflow or specific treatment protocols, but rather serve as general principles to inform diagnosis and staging of AD that reflect current science. HIGHLIGHTS: We define Alzheimer's disease (AD) to be a biological process that begins with the appearance of AD neuropathologic change (ADNPC) while people are asymptomatic. Progression of the neuropathologic burden leads to the later appearance and progression of clinical symptoms. Early-changing Core 1 biomarkers (amyloid positron emission tomography [PET], approved cerebrospinal fluid biomarkers, and accurate plasma biomarkers [especially phosphorylated tau 217]) map onto either the amyloid beta or AD tauopathy pathway; however, these reflect the presence of ADNPC more generally (i.e., both neuritic plaques and tangles). An abnormal Core 1 biomarker result is sufficient to establish a diagnosis of AD and to inform clinical decision making throughout the disease continuum. Later-changing Core 2 biomarkers (biofluid and tau PET) can provide prognostic information, and when abnormal, will increase confidence that AD is contributing to symptoms. An integrated biological and clinical staging scheme is described that accommodates the fact that common copathologies, cognitive reserve, and resistance may modify relationships between clinical and biological AD stages.
Collapse
Affiliation(s)
| | - J. Scott Andrews
- Global Evidence & OutcomesTakeda Pharmaceuticals Company LimitedCambridgeMassachusettsUSA
| | - Thomas G. Beach
- Civin Laboratory for NeuropathologyBanner Sun Health Research InstituteSun CityArizonaUSA
| | - Teresa Buracchio
- Office of NeuroscienceU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| | - Billy Dunn
- The Michael J. Fox Foundation for Parkinson's ResearchNew YorkNew YorkUSA
| | - Ana Graf
- NovartisNeuroscience Global Drug DevelopmentBaselSwitzerland
| | - Oskar Hansson
- Department of Clinical Sciences Malmö, Faculty of MedicineLund UniversityLundSweden
- Memory ClinicSkåne University Hospital, MalmöLundSweden
| | - Carole Ho
- DevelopmentDenali TherapeuticsSouth San FranciscoCaliforniaUSA
| | - William Jagust
- School of Public Health and Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Eric McDade
- Department of NeurologyWashington University St. Louis School of MedicineSt. LouisMissouriUSA
| | - Jose Luis Molinuevo
- Department of Global Clinical Development H. Lundbeck A/SExperimental MedicineCopenhagenDenmark
| | - Ozioma C. Okonkwo
- Department of Medicine, Division of Geriatrics and GerontologyUniversity of Wisconsin School of MedicineMadisonWisconsinUSA
| | - Luca Pani
- University of MiamiMiller School of MedicineMiamiFloridaUSA
| | - Michael S. Rafii
- Alzheimer's Therapeutic Research Institute (ATRI)Keck School of Medicine at the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Philip Scheltens
- Amsterdam University Medical Center (Emeritus)NeurologyAmsterdamthe Netherlands
| | - Eric Siemers
- Clinical ResearchAcumen PharmaceuticalsZionsvilleIndianaUSA
| | - Heather M. Snyder
- Medical & Scientific Relations DivisionAlzheimer's AssociationChicagoIllinoisUSA
| | - Reisa Sperling
- Department of Neurology, Brigham and Women's HospitalMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Charlotte E. Teunissen
- Department of Laboratory MedicineAmsterdam UMC, Neurochemistry LaboratoryAmsterdamthe Netherlands
| | - Maria C. Carrillo
- Medical & Scientific Relations DivisionAlzheimer's AssociationChicagoIllinoisUSA
| |
Collapse
|
55
|
Raj A, Torok J, Ranasinghe K. Understanding the complex interplay between tau, amyloid and the network in the spatiotemporal progression of Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.05.583407. [PMID: 38559176 PMCID: PMC10979926 DOI: 10.1101/2024.03.05.583407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
INTRODUCTION The interaction of amyloid and tau in neurodegenerative diseases is a central feature of AD pathophysiology. While experimental studies point to various interaction mechanisms, their causal direction and mode (local, remote or network-mediated) remain unknown in human subjects. The aim of this study was to compare mathematical reaction-diffusion models encoding distinct cross-species couplings to identify which interactions were key to model success. METHODS We tested competing mathematical models of network spread, aggregation, and amyloid-tau interactions on publicly available data from ADNI. RESULTS Although network spread models captured the spatiotemporal evolution of tau and amyloid in human subjects, the model including a one-way amyloid-to-tau aggregation interaction performed best. DISCUSSION This mathematical exposition of the "pas de deux" of co-evolving proteins provides quantitative, whole-brain support to the concept of amyloid-facilitated-tauopathy rather than the classic amyloid-cascade or pure-tau hypotheses, and helps explain certain known but poorly understood aspects of AD.
Collapse
|
56
|
Lim AC, Weissberger GH, Axelrod J, Mosqueda L, Nguyen AL, Fenton L, Noriega D, Erdman CE, Han SD. Neuropsychological profile associated with financial exploitation vulnerability in older adults without dementia. Clin Neuropsychol 2024:1-17. [PMID: 39060956 DOI: 10.1080/13854046.2024.2378526] [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: 12/20/2023] [Accepted: 07/06/2024] [Indexed: 07/28/2024]
Abstract
Objective: Reports of financial exploitation have steadily increased among older adults. Few studies have examined neuropsychological profiles for individuals vulnerable to financial exploitation, and existing studies have focused on susceptibility to scams, one specific type of financial exploitation. The current study therefore examines whether a general measure of financial exploitation vulnerability is associated with neuropsychological performance in a community sample. Methods: A sample (n = 116) of adults aged 50 or older without dementia completed a laboratory visit that measures physical and psychological functioning and a neuropsychological assessment, the Uniform Data Set-3 (UDS-3) and California Verbal Learning Test-II. Results: After covarying for demographics, current medical problems, financial literacy, and a global cognition screen, financial exploitation vulnerability was negatively associated with scores on the Multilingual Naming Test, Craft Story Recall and Delayed Recall, California Verbal Learning Test-II Delayed Recall and Recognition Discriminability, Phonemic Fluency, and Trails B. Financial exploitation vulnerability was not associated with performance on Digit Span, Semantic Fluency, Benson Complex Figure Recall, or Trails A. Conclusions: Among older adults without dementia, individuals at higher risk for financial exploitation demonstrated worse verbal memory, confrontation naming, phonemic fluency, and set-shifting. These tests are generally sensitive to Default Mode Network functioning and Alzheimer's Disease neuropathology. Longitudinal studies in more impaired samples are warranted to further corroborate and elucidate these relationships.
Collapse
Affiliation(s)
- Aaron C Lim
- Department of Psychology, USC Dornsife College of Letters, Los Angeles, CA, USA
| | - Gali H Weissberger
- Department of Social and Health Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Jenna Axelrod
- Department of Psychology, USC Dornsife College of Letters, Los Angeles, CA, USA
| | - Laura Mosqueda
- Department of Family Medicine, USC Keck School of Medicine, Alhambra, CA, USA
| | - Annie L Nguyen
- Herbert Wertheim School of Public Health & Human Longevity Science, University of California San Diego, CA, USA
| | - Laura Fenton
- Department of Psychology, USC Dornsife College of Letters, Los Angeles, CA, USA
| | - Daisy Noriega
- Department of Psychology, USC Dornsife College of Letters, Los Angeles, CA, USA
| | - Camille E Erdman
- Department of Psychology, USC Dornsife College of Letters, Los Angeles, CA, USA
| | - S Duke Han
- Department of Psychology, USC Dornsife College of Letters, Los Angeles, CA, USA
- Department of Family Medicine, USC Keck School of Medicine, Alhambra, CA, USA
| |
Collapse
|
57
|
Ayele BA, Whitehead PL, Pascual J, Gu T, Arvizu J, Golightly CG, Adams LD, Pericak-Vance MA, Vance JM, Griswold AJ. AD plasma biomarkers are stable for an extended period at -20°C: implications for resource-constrained environments. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.17.24310504. [PMID: 39072029 PMCID: PMC11275684 DOI: 10.1101/2024.07.17.24310504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Standard procedures for measuring Alzheimer's disease (AD) plasma biomarkers include storage at -80°C. This is challenging in countries lacking research infrastructure, such -80°C freezer. To investigate stability of AD biomarkers from plasma stored at -20°C, we compared aliquots stored at -80°C and others at -20°C for two, four, six, fifteen, and thirty-five weeks. pTau181, Aβ42, Aβ40, NfL, and GFAP were measured for each timepoint. pTau181 and Aβ42/Aβ40 ratios showed minimal variation for up to 15 weeks. NfL and GFAP had higher variability. This finding of 15-week stability at -20°C enables greater participation in AD biomarker studies in resource constrained environments.
Collapse
|
58
|
Bao YW, Wang ZJ, Shea YF, Chiu PKC, Kwan JS, Chan FHW, Mak HKF. Combined quantitative amyloid-β PET and structural MRI features improve Alzheimer's Disease classification in random forest model - A multicenter study. Acad Radiol 2024:S1076-6332(24)00426-4. [PMID: 39003227 DOI: 10.1016/j.acra.2024.06.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/18/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
RATIONALE AND OBJECTIVES Prior to clinical presentations of Alzheimer's Disease (AD), neuropathological changes, such as amyloid-β and brain atrophy, have accumulated at the earlier stages of the disease. The combination of such biomarkers assessed by multiple modalities commonly improves the likelihood of AD etiology. We aimed to explore the discriminative ability of Aβ PET features and whether combining Aβ PET and structural MRI features can improve the classification performance of the machine learning model in older healthy control (OHC) and mild cognitive impairment (MCI) from AD. MATERIAL AND METHODS We collected 94 AD patients, 82 MCI patients, and 85 OHC from three different cohorts. 17 global/regional Aβ features in Centiloid, 122 regional volume, and 68 regional cortical thickness were extracted as imaging features. Single or combined modality features were used to train the random forest model on the testing set. The top 10 features were sorted based on the Gini index in each binary classification. RESULTS The results showed that AUC scores were 0.81/0.86 and 0.69/0.68 using sMRI/Aβ PET features on the testing set in differentiating OHC and MCI from AD. The performance was improved while combining two-modality features with an AUC of 0.89 and an AUC of 0.71 in two classifications. Compared to sMRI features, particular Aβ PET features contributed more to differentiating AD from others. CONCLUSION Our study demonstrated the discriminative ability of Aβ PET features in differentiating AD from OHC and MCI. A combination of Aβ PET and structural MRI features can improve the RF model performance.
Collapse
Affiliation(s)
- Yi-Wen Bao
- Department of Medical Imaging Center, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China (Y-W.B.)
| | - Zuo-Jun Wang
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China (Z-J.W., H.K-F.M.)
| | - Yat-Fung Shea
- Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China (Y-F.S., P.K-C.C., J.S.K., F.H-W.C.)
| | - Patrick Ka-Chun Chiu
- Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China (Y-F.S., P.K-C.C., J.S.K., F.H-W.C.)
| | - Joseph Sk Kwan
- Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China (Y-F.S., P.K-C.C., J.S.K., F.H-W.C.)
| | - Felix Hon-Wai Chan
- Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China (Y-F.S., P.K-C.C., J.S.K., F.H-W.C.)
| | - Henry Ka-Fung Mak
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China (Z-J.W., H.K-F.M.).
| |
Collapse
|
59
|
Mousele C, Holden D, Gnanapavan S. Neurofilaments in neurologic disease. Adv Clin Chem 2024; 123:65-128. [PMID: 39181624 DOI: 10.1016/bs.acc.2024.06.010] [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] [Indexed: 08/27/2024]
Abstract
Neurofilaments (NFs), major cytoskeletal constituents of neurons, have emerged as universal biomarkers of neuronal injury. Neuroaxonal damage underlies permanent disability in various neurological conditions. It is crucial to accurately quantify and longitudinally monitor this damage to evaluate disease progression, evaluate treatment effectiveness, contribute to novel treatment development, and offer prognostic insights. Neurofilaments show promise for this purpose, as their levels increase with neuroaxonal damage in both cerebrospinal fluid and blood, independent of specific causal pathways. New assays with high sensitivity allow reliable measurement of neurofilaments in body fluids and open avenues to investigate their role in neurological disorders. This book chapter will delve into the evolving landscape of neurofilaments, starting with their structure and cellular functions within neurons. It will then provide a comprehensive overview of their broad clinical value as biomarkers in diseases affecting the central or peripheral nervous system.
Collapse
|
60
|
Nabizadeh F, Seyedmirzaei H, Karami S. Neuroimaging biomarkers and CSF sTREM2 levels in Alzheimer's disease: a longitudinal study. Sci Rep 2024; 14:15318. [PMID: 38961148 PMCID: PMC11222555 DOI: 10.1038/s41598-024-66211-w] [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: 10/27/2023] [Accepted: 06/28/2024] [Indexed: 07/05/2024] Open
Abstract
Understanding the exact pathophysiological mechanisms underlying the involvement of triggering receptor expressed on myeloid cells 2 (TREM2) related microglia activation is crucial for the development of clinical trials targeting microglia activation at different stages of Alzheimer's disease (AD). Given the contradictory findings in the literature, it is imperative to investigate the longitudinal alterations in cerebrospinal fluid (CSF) soluble TREM2 (sTREM2) levels as a marker for microglia activation, and its potential association with AD biomarkers, in order to address the current knowledge gap. In this study, we aimed to assess the longitudinal changes in CSF sTREM2 levels within the framework of the A/T/N classification system for AD biomarkers and to explore potential associations with AD pathological features, including the presence of amyloid-beta (Aβ) plaques and tau aggregates. The baseline and longitudinal (any available follow-up visit) CSF sTREM2 levels and processed tau-PET and Aβ-PET data of 1001 subjects were recruited from the ADNI database. The participants were classified into four groups based on the A/T/N framework: A+ /TN+ , A+ /TN- , A- /TN+ , and A- /TN- . Linear regression analyses were conducted to assess the relationship between CSF sTREM2 with cognitive performance, tau and Aβ-PET adjusting for age, gender, education, and APOE ε4 status. Based on our analysis there was a significant difference in baseline and rate of change of CSF sTREM2 between ATN groups. While there was no association between baseline CSF sTREM2 and cognitive performance (ADNI-mem), we found that the rate of change of CSF sTREM2 is significantly associated with cognitive performance in the entire cohort but not the ATN groups. We found that the baseline CSF sTREM2 is significantly associated with baseline tau-PET and Aβ-PET rate of change only in the A+ /TN+ group. A significant association was found between the rate of change of CSF sTREM2 and the tau- and Aβ-PET rate of change only in the A+ /TN- group. Our study suggests that the TREM2-related microglia activation and their relations with AD markers and cognitive performance vary the in presence or absence of Aβ and tau pathology. Furthermore, our findings revealed that a faster increase in the level of CSF sTREM2 might attenuate future Aβ plaque formation and tau aggregate accumulation only in the presence of Aβ pathology.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Alzheimer's Disease Institute, Tehran, Iran.
| | - Homa Seyedmirzaei
- School of Medicine, Tehran University of Medical Science, Tehran, Iran
- Interdisciplinary Neuroscience Research Program (INRP), Tehran University of Medical Sciences, Tehran, Iran
| | - Shaghayegh Karami
- School of Medicine, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
61
|
Bollinger RM, Chen SW, Krauss MJ, Keleman AA, Kehrer-Dunlap A, Kaesler M, Ances BM, Stark SL. The Association Between Postural Sway and Preclinical Alzheimer Disease Among Community-Dwelling Older Adults. J Gerontol A Biol Sci Med Sci 2024; 79:glae091. [PMID: 38554257 PMCID: PMC11167487 DOI: 10.1093/gerona/glae091] [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: 10/26/2023] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND It is unknown whether older adults with preclinical Alzheimer disease (AD) experience changes in postural sway compared with those without preclinical AD. The purpose of this study was to understand the effect of dual tasking on standing balance, or postural sway, for people with and without preclinical AD. METHODS A cross-sectional analysis of baseline data from a longitudinal cohort study. Participants were cognitively normal older adults with and without preclinical AD. Postural sway (path length) was tested using a force plate under standard and dual task balance conditions. Dual task cost (DTC) was calculated to examine performance change in balance conditions. Logistic regression models were used to predict preclinical AD status as a function of DTC. RESULTS 203 participants (65 preclinical AD+) were included. DTC for path length was significantly greater for participants with preclinical AD (DTC path length mean difference 19.8, 95% CI 2.6-37.0, t(201) = 2.29, p = .024). Greater DTC was significantly associated with increased odds of having preclinical AD (adjusted odds ratio for a 20-unit increase in DTC 1.16, 95% CI 1.02-1.32). CONCLUSIONS Older adults with preclinical AD are more likely to demonstrate significantly greater DTC in postural sway than those without preclinical AD. Dual tasking should be integrated into balance and fall risk assessments and may inform early detection of preclinical AD.
Collapse
Affiliation(s)
- Rebecca M Bollinger
- Program in Occupational Therapy, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Szu-Wei Chen
- Program in Occupational Therapy, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Melissa J Krauss
- Program in Occupational Therapy, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Audrey A Keleman
- Program in Occupational Therapy, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Abigail Kehrer-Dunlap
- Program in Occupational Therapy, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Megan Kaesler
- Program in Occupational Therapy, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Beau M Ances
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Susan L Stark
- Program in Occupational Therapy, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
62
|
Robinson CG, Goodrich AW, Weigand SD, Pham NTT, Carlos AF, Buciuc M, Murray ME, Nguyen AT, Reichard RR, Knopman DS, Petersen RC, Dickson DW, Utianski RL, Whitwell JL, Josephs KA, Machulda MM. Determinants of confrontation naming deficits on the Boston Naming Test associated with transactive response DNA-binding protein 43 pathology. J Int Neuropsychol Soc 2024; 30:575-583. [PMID: 38525671 PMCID: PMC11422518 DOI: 10.1017/s1355617724000146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
OBJECTIVE To determine whether poorer performance on the Boston Naming Test (BNT) in individuals with transactive response DNA-binding protein 43 pathology (TDP-43+) is due to greater loss of word knowledge compared to retrieval-based deficits. METHODS Retrospective clinical-pathologic study of 282 participants with Alzheimer's disease neuropathologic changes (ADNC) and known TDP-43 status. We evaluated item-level performance on the 60-item BNT for first and last available assessment. We fit cross-sectional negative binomial count models that assessed total number of incorrect items, number correct of responses with phonemic cue (reflecting retrieval difficulties), and number of "I don't know" (IDK) responses (suggestive of loss of word knowledge) at both assessments. Models included TDP-43 status and adjusted for sex, age, education, years from test to death, and ADNC severity. Models that evaluated the last assessment adjusted for number of prior BNT exposures. RESULTS 43% were TDP-43+. The TDP-43+ group had worse performance on BNT total score at first (p = .01) and last assessments (p = .01). At first assessment, TDP-43+ individuals had an estimated 29% (CI: 7%-56%) higher mean number of incorrect items after adjusting for covariates, and a 51% (CI: 15%-98%) higher number of IDK responses compared to TDP-43-. At last assessment, compared to TDP-43-, the TDP-43+ group on average missed 31% (CI: 6%-62%; p = .01) more items and had 33% more IDK responses (CI: 1% fewer to 78% more; p = .06). CONCLUSIONS An important component of poorer performance on the BNT in participants who are TDP-43+ is having loss of word knowledge versus retrieval difficulties.
Collapse
Affiliation(s)
| | | | | | | | | | - Marina Buciuc
- Department of Neurology, Medical University of South Carolina, Charleston, SC
| | | | - Aivi T. Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | | | - Mary M. Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN
| |
Collapse
|
63
|
Garcia-Cordero I, Anastassiadis C, Khoja A, Morales-Rivero A, Thapa S, Vasilevskaya A, Davenport C, Sumra V, Couto B, Multani N, Taghdiri F, Anor C, Misquitta K, Vandevrede L, Heuer H, Tang-Wai D, Dickerson B, Pantelyat A, Litvan I, Boeve B, Rojas JC, Ljubenkov P, Huey E, Fox S, Kovacs GG, Boxer A, Lang A, Tartaglia MC. Evaluating the Effect of Alzheimer's Disease-Related Biomarker Change in Corticobasal Syndrome and Progressive Supranuclear Palsy. Ann Neurol 2024; 96:99-109. [PMID: 38578117 PMCID: PMC11249787 DOI: 10.1002/ana.26930] [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: 11/13/2023] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 04/06/2024]
Abstract
OBJECTIVES To evaluate the effect of Alzheimer's disease (AD) -related biomarker change on clinical features, brain atrophy and functional connectivity of patients with corticobasal syndrome (CBS) and progressive supranuclear palsy (PSP). METHODS Data from patients with a clinical diagnosis of CBS, PSP, and AD and healthy controls were obtained from the 4-R-Tauopathy Neuroimaging Initiative 1 and 2, the Alzheimer's Disease Neuroimaging Initiative, and a local cohort from the Toronto Western Hospital. Patients with CBS and PSP were divided into AD-positive (CBS/PSP-AD) and AD-negative (CBS/PSP-noAD) groups based on fluid biomarkers and amyloid PET scans. Cognitive, motor, and depression scores; AD fluid biomarkers (cerebrospinal p-tau, t-tau, and amyloid-beta, and plasma ptau-217); and neuroimaging data (amyloid PET, MRI and fMRI) were collected. Clinical features, whole-brain gray matter volume and functional networks connectivity were compared across groups. RESULTS Data were analyzed from 87 CBS/PSP-noAD and 23 CBS/PSP-AD, 18 AD, and 30 healthy controls. CBS/PSP-noAD showed worse performance in comparison to CBS/PSP-AD in the PSPRS [mean(SD): 34.8(15.8) vs 23.3(11.6)] and the UPDRS scores [mean(SD): 34.2(17.0) vs 21.8(13.3)]. CBS/PSP-AD demonstrated atrophy in AD signature areas and brainstem, while CBS/PSP-noAD patients displayed atrophy in frontal and temporal areas, globus pallidus, and brainstem compared to healthy controls. The default mode network showed greatest disconnection in CBS/PSP-AD compared with CBS/PSP-no AD and controls. The thalamic network connectivity was most affected in CBS/PSP-noAD. INTERPRETATION AD biomarker positivity may modulate the clinical presentation of CBS/PSP, with evidence of distinctive structural and functional brain changes associated with the AD pathology/co-pathology. ANN NEUROL 2024;96:99-109.
Collapse
Affiliation(s)
- Indira Garcia-Cordero
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Chloe Anastassiadis
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Abeer Khoja
- University Health Network Memory Clinic, Toronto, Ontario, Canada
- Neurology division, Medical Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alonso Morales-Rivero
- University Health Network Memory Clinic, Toronto, Ontario, Canada
- ABC Medical Center, Mexico City, Mexico
| | - Simrika Thapa
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Anna Vasilevskaya
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Carly Davenport
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Vishaal Sumra
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Blas Couto
- Rossy PSP Program, University Health Network and the University of Toronto, Toronto, Ontario, Canada
- The Edmond J. Safra Program in Parkinson’s Disease and Morton and Gloria Shulman Movement Disorders Clinic, Toronto, Ontario, Canada
- Institute of Cognitive and Translational Neuroscience (INCyT-INECO-CONICET), Favaloro University Hospital, Buenos Aires, Argentina
| | - Namita Multani
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Foad Taghdiri
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Cassandra Anor
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Karen Misquitta
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Lawren Vandevrede
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, California, USA
| | - Hilary Heuer
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, California, USA
| | - David Tang-Wai
- Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
| | - Bradford Dickerson
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Irene Litvan
- Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Bradley Boeve
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Julio C. Rojas
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, California, USA
| | - Peter Ljubenkov
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, California, USA
| | - Edward Huey
- Department of Psychiatry and Human Behavior, Brown University, Providence, Rhode Island, USA
| | - Susan Fox
- Rossy PSP Program, University Health Network and the University of Toronto, Toronto, Ontario, Canada
- The Edmond J. Safra Program in Parkinson’s Disease and Morton and Gloria Shulman Movement Disorders Clinic, Toronto, Ontario, Canada
| | - Gabor G. Kovacs
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Rossy PSP Program, University Health Network and the University of Toronto, Toronto, Ontario, Canada
- The Edmond J. Safra Program in Parkinson’s Disease and Morton and Gloria Shulman Movement Disorders Clinic, Toronto, Ontario, Canada
- Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
| | - Adam Boxer
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, California, USA
| | - Anthony Lang
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Rossy PSP Program, University Health Network and the University of Toronto, Toronto, Ontario, Canada
- The Edmond J. Safra Program in Parkinson’s Disease and Morton and Gloria Shulman Movement Disorders Clinic, Toronto, Ontario, Canada
- Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
| | - M. Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- University Health Network Memory Clinic, Toronto, Ontario, Canada
- Rossy PSP Program, University Health Network and the University of Toronto, Toronto, Ontario, Canada
- Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
| | | |
Collapse
|
64
|
Aschenbrenner AJ, Carr DB, Benzinger TLS, Morris JC, Babulal GM. The Influence of Personality Traits on Driving Behaviors in Preclinical Alzheimer Disease. Alzheimer Dis Assoc Disord 2024; 38:241-248. [PMID: 39177169 PMCID: PMC11524437 DOI: 10.1097/wad.0000000000000632] [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] [Accepted: 07/08/2024] [Indexed: 08/24/2024]
Abstract
INTRODUCTION Alzheimer disease (AD) has a long preclinical phase in which AD pathology is accumulating without detectable clinical symptoms. It is critical to identify participants in this preclinical phase as early as possible since treatment plans may be more effective in this stage. Monitoring for changes in driving behavior, as measured with GPS sensors, has been explored as a low-burden, easy-to-administer method for detecting AD risk. However, driving is a complex, multifaceted process that is likely influenced by other factors, including personality traits, that may change in preclinical AD. METHODS We examine the moderating influence of neuroticism and conscientiousness on longitudinal changes in driving behavior in a sample of 203 clinically normal older adults who are at varying risk of developing AD. RESULTS Neuroticism moderated rates of change in the frequency of speeding as well as the number of trips taken at night. Conscientiousness moderated rates of change in typical driving space. CONCLUSIONS Personality traits change in early AD and also influence driving behaviors. Studies that seek to utilize naturalistic driving behavior to establish AD risk need to accommodate interpersonal differences, of which personality traits are one of many possible factors. Future studies should explicitly establish how much benefit is provided by including personality traits in predictive models of AD progression.
Collapse
Affiliation(s)
| | | | | | | | - Ganesh M Babulal
- Departments of Neurology
- Institute of Public Health, Washington University School of Medicine, St. Louis, MO
- Department of Clinical Research and Leadership, The George Washington University School of Medicine and Health Sciences, Washington, DC
- Department of Psychology, Faculty of Humanities, University of Johannesburg, South Africa
| |
Collapse
|
65
|
Nicola L, Loo SJQ, Lyon G, Turknett J, Wood TR. Does resistance training in older adults lead to structural brain changes associated with a lower risk of Alzheimer's dementia? A narrative review. Ageing Res Rev 2024; 98:102356. [PMID: 38823487 DOI: 10.1016/j.arr.2024.102356] [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: 03/10/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
Dementia, particularly Alzheimer's Disease (AD), has links to several modifiable risk factors, especially physical inactivity. When considering the relationship between physcial activity and dementia risk, cognitive benefits are generally attributed to aerobic exercise, with resistance exercise (RE) receiving less attention. This review aims to address this gap by evaluating the impact of RE on brain structures and cognitive deficits associated with AD. Drawing insights from randomized controlled trials (RCTs) utilizing structural neuroimaging, the specific influence of RE on AD-affected brain structures and their correlation with cognitive function are discussed. Preliminary findings suggest that RE induces structural brain changes in older adults that could reduce the risk of AD or mitigate AD progression. Importantly, the impacts of RE appear to follow a dose-response effect, reversing pathological structural changes and improving associated cognitive functions if performed at least twice per week for at least six months, with greatest effects in those already experiencing some element of cognitive decline. While more research is eagerly awaited, this review contributes insights into the potential benefits of RE for cognitive health in the context of AD-related changes in brain structure and function.
Collapse
Affiliation(s)
| | | | | | | | - Thomas R Wood
- Department of Pediatrics, University of Washington, Seattle, WA, USA; Institute for Human and Machine Cognition, Pensacola, FL, USA.
| |
Collapse
|
66
|
Lázaro I, Grau‐Rivera O, Suárez‐Calvet M, Fauria K, Minguillón C, Shekari M, Falcón C, García‐Prat M, Huguet J, Molinuevo JL, Gispert J, Sala‐Vila A. Omega-3 blood biomarkers relate to brain glucose uptake in individuals at risk of Alzheimer's disease dementia. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12596. [PMID: 38974876 PMCID: PMC11224768 DOI: 10.1002/dad2.12596] [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: 12/01/2023] [Revised: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 07/09/2024]
Abstract
INTRODUCTION Brain glucose hypometabolism is a preclinical feature of Alzheimer's disease (AD). Dietary omega-3 fatty acids promote brain glucose metabolism, but clinical research is incipient. Circulating omega-3s objectively reflect their dietary intake. METHODS This was a cross-sectional study in 320 cognitively unimpaired participants at increased risk of AD dementia. Using lipidomics, we determined blood docosahexaenoic (DHA) and alpha-linolenic (ALA) acid levels (omega-3s from marine and plant origin, respectively). We assessed brain glucose metabolism using [18-F]-fluorodeoxyglucose (FDG) positron emission tomography (PET). RESULTS Blood ALA directly related to FDG uptake in brain areas known to be affected in AD. Stronger associations were observed in apolipoprotein E ε4 carriers and homozygotes. For DHA, significant direct associations were restricted to amyloid beta-positive tau-positive participants. DISCUSSION Blood omega-3 directly relate to preserved glucose metabolism in AD-vulnerable brain regions in individuals at increased risk of AD dementia. This adds to the benefits of omega-3 supplementation in the preclinical stage of AD dementia. Highlights Blood omega-3s were related to brain glucose uptake in participants at risk of Alzheimer's disease (AD) dementia.Complementary associations were observed for omega-3 from marine and plant sources.Foods rich in omega-3 might be useful in early features of AD.
Collapse
Affiliation(s)
- Iolanda Lázaro
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN)Instituto de Salud Carlos IIIMadridSpain
| | - Oriol Grau‐Rivera
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
| | - Marc Suárez‐Calvet
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
| | - Karine Fauria
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
| | - Carolina Minguillón
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
| | - Mahnaz Shekari
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
| | - Carles Falcón
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red BioingenieríaBiomateriales y Nanomedicina (CIBERBBN)Instituto de Salud Carlos IIIMadridSpain
| | - Marina García‐Prat
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
| | - Jordi Huguet
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Lundbeck A/SCopenhagenDenmark
| | - Juan‐Domingo Gispert
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red BioingenieríaBiomateriales y Nanomedicina (CIBERBBN)Instituto de Salud Carlos IIIMadridSpain
| | - Aleix Sala‐Vila
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN)Instituto de Salud Carlos IIIMadridSpain
- Fatty Acid Research InstituteSioux FallsSouth DakotaUSA
| | | |
Collapse
|
67
|
Qin H, Shi X, Zhu Y, Ma J, Deng X, Wang L. Alzheimer's disease early screening and staged detection with plasma proteome using machine learning and convolutional neural network. Eur J Neurosci 2024; 60:4034-4048. [PMID: 38764192 DOI: 10.1111/ejn.16392] [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: 01/18/2024] [Revised: 04/19/2024] [Accepted: 04/27/2024] [Indexed: 05/21/2024]
Abstract
Alzheimer's disease (AD) stands as the prevalent progressive neurodegenerative disease, precipitating cognitive impairment and even memory loss. Amyloid biomarkers have been extensively used in the diagnosis of AD. However, amyloid proteins offer limited information about the disease process and accurate diagnosis depends on the presence of a substantial accumulation of amyloid deposition which significantly impedes the early screening of AD. In this study, we have combined plasma proteomics with an ensemble learning model (CatBoost) to develop a cost-effective and non-invasive diagnostic method for AD. A longitudinal panel has been identified that can serve as reliable biomarkers across the entire progression of AD. Simultaneously, we have developed a neural network algorithm that utilizes plasma proteins to detect stages of Alzheimer's disease. Based on the developed longitudinal panel, the CatBoost model achieved an area under the operating curve of at least 0.90 in distinguishing mild cognitive impairment from cognitively normal. The neural network model was utilized for the detection of three stages of AD, and the results demonstrated that the neural network model exhibited an accuracy as high as 0.83, surpassing that of the traditional machine learning model.
Collapse
Affiliation(s)
- Hengyu Qin
- School of Information and Electronics, Beijing Institute of Technology, Beijing, China
| | - Xiumin Shi
- School of Information and Electronics, Beijing Institute of Technology, Beijing, China
| | - Yibo Zhu
- School of Information and Electronics, Beijing Institute of Technology, Beijing, China
| | - Jiacheng Ma
- The Department of Information Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Lu Wang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
68
|
Badhwar A, Hirschberg Y, Valle‐Tamayo N, Iulita MF, Udeh‐Momoh CT, Matton A, Tarawneh RM, Rissman RA, Ledreux A, Winston CN, Haqqani AS. Assessment of brain-derived extracellular vesicle enrichment for blood biomarker analysis in age-related neurodegenerative diseases: An international overview. Alzheimers Dement 2024; 20:4411-4422. [PMID: 38864416 PMCID: PMC11247682 DOI: 10.1002/alz.13823] [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: 08/17/2023] [Revised: 02/09/2024] [Accepted: 02/17/2024] [Indexed: 06/13/2024]
Abstract
INTRODUCTION Brain-derived extracellular vesicles (BEVs) in blood allows for minimally-invasive investigations of central nervous system (CNS) -specific markers of age-related neurodegenerative diseases (NDDs). Polymer-based EV- and immunoprecipitation (IP)-based BEV-enrichment protocols from blood have gained popularity. We systematically investigated protocol consistency across studies, and determined CNS-specificity of proteins associated with these protocols. METHODS NDD articles investigating BEVs in blood using polymer-based and/or IP-based BEV enrichment protocols were systematically identified, and protocols compared. Proteins used for BEV-enrichment and/or post-enrichment were assessed for CNS- and brain-cell-type-specificity, extracellular domains (ECD+), and presence in EV-databases. RESULTS A total of 82.1% of studies used polymer-based (ExoQuick) EV-enrichment, and 92.3% used L1CAM for IP-based BEV-enrichment. Centrifugation times differed across studies. A total of 26.8% of 82 proteins systematically identified were CNS-specific: 50% ECD+, 77.3% were listed in EV-databases. CONCLUSIONS We identified protocol steps requiring standardization, and recommend additional CNS-specific proteins that can be used for BEV-enrichment or as BEV-biomarkers. HIGHLIGHTS Across NDDs, we identified protocols commonly used for EV/BEV enrichment from blood. We identified protocol steps showing variability that require harmonization. We assessed CNS-specificity of proteins used for BEV-enrichment or found in BEV cargo. CNS-specific EV proteins with ECD+ or without were identified. We recommend evaluation of blood-BEV enrichment using these additional ECD+ proteins.
Collapse
Affiliation(s)
- AmanPreet Badhwar
- Département de pharmacologie et physiologieInstitut de Génie BiomédicalFaculté de Médecine, Université de MontréalMontréalQuebecCanada
- Multiomics Investigation of Neurodegenerative Diseases (MIND) lab, Centre de recherche de l'Institut Universitaire de GériatrieMontréalQuebecCanada
| | - Yael Hirschberg
- Centre for ProteomicsUniversity of AntwerpAntwerpBelgium
- Health Unit, Flemish Institute for Technological Research (VITO)MolBelgium
| | - Natalia Valle‐Tamayo
- Sant Pau Memory Unit, Department of NeurologyHospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant PauCalle San QuintíBarcelonaSpain
| | - M. Florencia Iulita
- Sant Pau Memory Unit, Department of NeurologyHospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant PauCalle San QuintíBarcelonaSpain
| | - Chinedu T. Udeh‐Momoh
- Ageing Epidemiology research unit, School of Public Health, Imperial College LondonLondonUK
- Division of Clinical GeriatricsDepartment of NeurobiologyCare Sciences and Society, Center for Alzheimer Research, Karolinska InstitutetSolnaSweden
- Global Brain Health InstituteUniversity of San Francisco Joan and Sanford I. Weill Neurosciences buildingSan FranciscoCaliforniaUSA
- Imarisha Centre for Brain Health and AgingBrain and Mind InstituteAga Khan UniversityNairobiKenya
| | - Anna Matton
- Ageing Epidemiology research unit, School of Public Health, Imperial College LondonLondonUK
- Division of Clinical GeriatricsDepartment of NeurobiologyCare Sciences and Society, Center for Alzheimer Research, Karolinska InstitutetSolnaSweden
- Division of NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyCenter for Alzheimer Research, Karolinska Institutet, SolnaNobels vägSweden
| | - Rawan M. Tarawneh
- Department of NeurologyCenter for Memory and AgingUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | - Robert A. Rissman
- VA San Diego Healthcare SystemSan DiegoCaliforniaUSA
- Department of Physiology and NeuroscienceAlzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Aurélie Ledreux
- Department of NeurosurgerySchool of MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Charisse N. Winston
- Department of Physiology and NeuroscienceAlzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | | | | |
Collapse
|
69
|
Whitfield T, Chouliaras L, Morrell R, Rubio D, Radford D, Marchant NL, Walker Z. The criteria used to rule out mild cognitive impairment impact dementia incidence rates in subjective cognitive decline. Alzheimers Res Ther 2024; 16:142. [PMID: 38943160 PMCID: PMC11212190 DOI: 10.1186/s13195-024-01516-6] [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: 04/16/2024] [Accepted: 06/23/2024] [Indexed: 07/01/2024]
Abstract
BACKGROUND The research criteria for subjective cognitive decline (SCD) exclude mild cognitive impairment (MCI), but do not stipulate the use of specific MCI criteria. This study compared different approaches to defining (i.e., excluding) MCI during the ascertainment of SCD, focusing on the impact on dementia incidence rates in SCD. METHODS This cohort study utilized routine healthcare data collected in the Essex Memory Clinic from 1999 to 2023. Two different operationalizations of the SCD criteria were used to categorize the cohort into two SCD patient samples. One sample was based on local clinical practice - MCI was excluded according to the Winblad criteria (this sample was termed SCDWinblad). The other sample was created via the retrospective application of the Jak/Bondi criteria for the exclusion of MCI (termed SCDJak/Bondi). Only patients aged ≥ 55 years at baseline with ≥ 12 months follow-up were considered for inclusion. The initial clinical/demographic characteristics of the samples were compared. Rates of incident dementia were calculated for each sample, and unadjusted and Mantel-Haenszel-adjusted incidence rate ratios were calculated to compare dementia incidence between the SCD samples. RESULTS The Essex Memory Clinic database included 2,233 patients in total. The SCD and study eligibility criteria were used to select SCDWinblad (n = 86) and SCDJak/Bondi (n = 185) samples from the database. Median follow-up (3 years) did not differ between the two samples. The SCDJak/Bondi sample was significantly older than the SCDWinblad at first assessment (median age: 74 versus 70 years) and had poorer scores on tests of global cognition, immediate and delayed verbal recall, and category fluency. Following adjustment for age, the dementia incidence rate ratio [95% confidence interval] was 3.7 [1.5 to 9.3], indicating a significantly greater rate of progression to dementia in SCDJak/Bondi. CONCLUSIONS This study highlights that the approach used to ascertain SCD has important implications for both SCD phenotypes and prognosis. This underscores the importance of how MCI is operationalized within SCD studies. More broadly, the findings add to a growing body of work indicating that objective cognition should not be overlooked in SCD, and offer a potential explanation for the heterogeneity across the SCD prognostic literature.
Collapse
Affiliation(s)
- Tim Whitfield
- Division of Psychiatry, University College London, London, UK.
| | - Leonidas Chouliaras
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Specialist Dementia and Frailty Service, Essex Partnership University NHS Foundation Trust, St Margaret's Hospital, Epping, UK
| | - Rachel Morrell
- Division of Psychiatry, University College London, London, UK
| | - David Rubio
- Specialist Dementia and Frailty Service, Essex Partnership University NHS Foundation Trust, St Margaret's Hospital, Epping, UK
| | - Darren Radford
- Specialist Dementia and Frailty Service, Essex Partnership University NHS Foundation Trust, St Margaret's Hospital, Epping, UK
| | | | - Zuzana Walker
- Division of Psychiatry, University College London, London, UK
- Specialist Dementia and Frailty Service, Essex Partnership University NHS Foundation Trust, St Margaret's Hospital, Epping, UK
| |
Collapse
|
70
|
Valles-Salgado M, Gil-Moreno MJ, Curiel Cid RE, Delgado-Álvarez A, Ortega-Madueño I, Delgado-Alonso C, Palacios-Sarmiento M, López-Carbonero JI, Cárdenas MC, Matías-Guiu J, Díez-Cirarda M, Loewenstein DA, Matias-Guiu JA. Detection of cerebrospinal fluid biomarkers changes of Alzheimer's disease using a cognitive stress test in persons with subjective cognitive decline and mild cognitive impairment. Front Psychol 2024; 15:1373541. [PMID: 38988382 PMCID: PMC11233766 DOI: 10.3389/fpsyg.2024.1373541] [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: 01/19/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024] Open
Abstract
Introduction Timely and accurate diagnosis of the earliest manifestations of Alzheimer's disease (AD) is critically important. Cognitive challenge tests such as the Loewenstein Acevedo Scales for Semantic Interference and Learning (LASSI-L) have shown favorable diagnostic properties in a number of previous investigations using amyloid or FDG PET. However, no studies have examined LASSI-L performance against cerebrospinal fluid biomarkers of AD, which can be affected before the distribution of fibrillar amyloid and other changes that can be observed in brain neuroimaging. Therefore, we aimed to evaluate the relationship between LASSI-L scores and CSF biomarkers and the capacity of the cognitive challenge test to detect the presence of amyloid and tau deposition in patients with subjective cognitive decline and amnestic mild cognitive impairment (MCI). Methods One hundred and seventy-nine patients consulting for memory loss without functional impairment were enrolled. Patients were examined using comprehensive neuropsychological assessment, the LASSI-L, and cerebrospinal fluid (CSF) biomarkers (Aβ1-42/Aβ1-40 and ptau181). Means comparisons, correlations, effect sizes, and ROC curves were calculated. Results LASSI-L scores were significantly associated with CSF biomarkers Aβ1-42/Aβ1-40 in patients diagnosed with MCI and subjective cognitive decline, especially those scores evaluating the capacity to recover from proactive semantic interference effects and delayed recall. A logistic regression model for the entire sample including LASSI-L and age showed an accuracy of 0.749 and an area under the curve of 0.785 to detect abnormal amyloid deposition. Conclusion Our study supports the biological validity of the LASSI-L and its semantic interference paradigm in the context of the early stages of AD. These findings emphasize the utility and the convenience of including sensitive cognitive challenge tests in the assessment of patients with suspicion of early stages of AD.
Collapse
Affiliation(s)
- Maria Valles-Salgado
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Health Research Institute (IdISSC), Universidad Complutense de Madrid, Madrid, Spain
| | - María José Gil-Moreno
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Health Research Institute (IdISSC), Universidad Complutense de Madrid, Madrid, Spain
| | - Rosie E Curiel Cid
- Center for Cognitive Neuroscience and Aging, Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Alfonso Delgado-Álvarez
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Health Research Institute (IdISSC), Universidad Complutense de Madrid, Madrid, Spain
| | - Isabel Ortega-Madueño
- Department of Clinical Analysis, Institute of Laboratory, Hospital Clínico San Carlos, San Carlos Health Research Institute (IdISSC), Universidad Complutense de Madrid, Madrid, Spain
| | - Cristina Delgado-Alonso
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Health Research Institute (IdISSC), Universidad Complutense de Madrid, Madrid, Spain
| | - Marta Palacios-Sarmiento
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Health Research Institute (IdISSC), Universidad Complutense de Madrid, Madrid, Spain
| | - Juan I López-Carbonero
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Health Research Institute (IdISSC), Universidad Complutense de Madrid, Madrid, Spain
| | - María Cruz Cárdenas
- Department of Clinical Analysis, Institute of Laboratory, Hospital Clínico San Carlos, San Carlos Health Research Institute (IdISSC), Universidad Complutense de Madrid, Madrid, Spain
| | - Jorge Matías-Guiu
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Health Research Institute (IdISSC), Universidad Complutense de Madrid, Madrid, Spain
| | - María Díez-Cirarda
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Health Research Institute (IdISSC), Universidad Complutense de Madrid, Madrid, Spain
| | - David A Loewenstein
- Center for Cognitive Neuroscience and Aging, Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Jordi A Matias-Guiu
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Health Research Institute (IdISSC), Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
71
|
Fall AB, Preti MG, Eshmawey M, Kagerer SM, Van De Ville D, Unschuld PG. Functional network centrality indicates interactions between APOE4 and age across the clinical spectrum of Alzheimer's Disease. Neuroimage Clin 2024; 43:103635. [PMID: 38941766 PMCID: PMC11260379 DOI: 10.1016/j.nicl.2024.103635] [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: 04/11/2024] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Advanced age is the most important risk factor for Alzheimer's disease (AD), and carrier-status of the Apolipoprotein E4 (APOE4) allele is the strongest known genetic risk factor. Many studies have consistently shown a link between APOE4 and synaptic dysfunction, possibly reflecting pathologically accelerated biological aging in persons at risk for AD. To test the hypothesis that distinct functional connectivity patterns characterize APOE4 carriers across the clinical spectrum of AD, we investigated 128 resting state functional Magnetic Resonance Imaging (fMRI) datasets from the Alzheimer's Disease Neuroimaging Initiative database (ADNI), representing all disease stages from cognitive normal to clinical dementia. Brain region centralities within functional networks, computed as eigenvector centrality, were tested for multivariate associations with chronological age, APOE4 carrier status and clinical stage (as well as their interactions) by partial least square analysis (PLSC). By PLSC analysis two distinct brain activity patterns could be identified, which reflected interactive effects of age, APOE4 and clinical disease stage. A first component including sensorimotor regions and parietal regions correlated with age and AD clinical stage (p < 0.001). A second component focused on medial-frontal regions and was specifically related to the interaction between age and APOE4 (p = 0.032). Our findings are consistent with earlier reports on altered network connectivity in APOE4 carriers. Results of our study highlight promise of graph-theory based network centrality to identify brain connectivity linked to genetic risk, clinical stage and age. Our data suggest the existence of brain network activity patterns that characterize APOE4 carriers across clinical stages of AD.
Collapse
Affiliation(s)
- Aïda B Fall
- Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland; Geriatric Psychiatry Service, University Hospitals of Geneva (HUG), Thônex, Switzerland; CIBM Center for Biomedical Imaging, Switzerland.
| | - Maria Giulia Preti
- Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland; CIBM Center for Biomedical Imaging, Switzerland; Neuro-X Institute, École Polytechnique Fédérale de Lausanne, Geneva, Switzerland
| | - Mohamed Eshmawey
- Geriatric Psychiatry Service, University Hospitals of Geneva (HUG), Thônex, Switzerland
| | - Sonja M Kagerer
- Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland; Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland; Psychogeriatric Medicine, Psychiatric University Hospital Zurich (PUK), Zurich, Switzerland
| | - Dimitri Van De Ville
- Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland; CIBM Center for Biomedical Imaging, Switzerland; Neuro-X Institute, École Polytechnique Fédérale de Lausanne, Geneva, Switzerland
| | - Paul G Unschuld
- Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland; Geriatric Psychiatry Service, University Hospitals of Geneva (HUG), Thônex, Switzerland
| |
Collapse
|
72
|
Koychev I, Adler AI, Edison P, Tom B, Milton JE, Butchart J, Hampshire A, Marshall C, Coulthard E, Zetterberg H, Hellyer P, Cormack F, Underwood BR, Mummery CJ, Holman RR. Protocol for a double-blind placebo-controlled randomised controlled trial assessing the impact of oral semaglutide in amyloid positivity (ISAP) in community dwelling UK adults. BMJ Open 2024; 14:e081401. [PMID: 38908839 PMCID: PMC11328662 DOI: 10.1136/bmjopen-2023-081401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 05/24/2024] [Indexed: 06/24/2024] Open
Abstract
INTRODUCTION Glucagon-like peptide-1 receptor agonists (GLP-1 RAs), currently marketed for type 2 diabetes and obesity, may offer novel mechanisms to delay or prevent neurotoxicity associated with Alzheimer's disease (AD). The impact of semaglutide in amyloid positivity (ISAP) trial is investigating whether the GLP-1 RA semaglutide reduces accumulation in the brain of cortical tau protein and neuroinflammation in individuals with preclinical/prodromal AD. METHODS AND ANALYSIS ISAP is an investigator-led, randomised, double-blind, superiority trial of oral semaglutide compared with placebo. Up to 88 individuals aged ≥55 years with brain amyloid positivity as assessed by positron emission tomography (PET) or cerebrospinal fluid, and no or mild cognitive impairment, will be randomised. People with the low-affinity binding variant of the rs6971 allele of the Translocator Protein 18 kDa (TSPO) gene, which can interfere with interpreting TSPO PET scans (a measure of neuroinflammation), will be excluded.At baseline, participants undergo tau, TSPO PET and MRI scanning, and provide data on physical activity and cognition. Eligible individuals are randomised in a 1:1 ratio to once-daily oral semaglutide or placebo, starting at 3 mg and up-titrating to 14 mg over 8 weeks. They will attend safety visits and provide blood samples to measure AD biomarkers at weeks 4, 8, 26 and 39. All cognitive assessments are repeated at week 26. The last study visit will be at week 52, when all baseline measurements will be repeated. The primary end point is the 1-year change in tau PET signal. ETHICS AND DISSEMINATION The study was approved by the West Midlands-Edgbaston Research Ethics Committee (22/WM/0013). The results of the study will be disseminated through scientific presentations and peer-reviewed publications. TRIAL REGISTRATION NUMBER ISRCTN71283871.
Collapse
Affiliation(s)
- Ivan Koychev
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Amanda I Adler
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Paul Edison
- Faculty of Medicine, Department of Brain Sciences, Imperial College London, London, UK
| | - Brian Tom
- Medical Research Council Biostatistics Unit, University of Cambridge, UK
| | - Joanne E Milton
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Joe Butchart
- Royal Devon University Healthcare Foundation Trust, Exeter, UK
- University of Exeter Medical School, Exeter, UK
| | - Adam Hampshire
- Faculty of Medicine, Department of Brain Sciences, Imperial College London, London, UK
| | - Charles Marshall
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, People's Republic of China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA18 Dementia Research Centre, Institute of Neurology, University College London, Queen Square, London, UK
| | - Peter Hellyer
- Faculty of Medicine, Department of Brain Sciences, Imperial College London, London, UK
| | | | - Benjamin R Underwood
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation trust, Cambridge, UK
| | - Catherine J Mummery
- Dementia Research Centre, Institute of Neurology, University College London, Queen Square, London, UK
| | - Rury R Holman
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
73
|
Dolci G, Cruciani F, Rahaman MA, Abrol A, Chen J, Fu Z, Galazzo IB, Menegaz G, Calhoun VD. An interpretable generative multimodal neuroimaging-genomics framework for decoding Alzheimer's disease. ARXIV 2024:arXiv:2406.13292v1. [PMID: 38947922 PMCID: PMC11213156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, affecting millions worldwide with a progressive decline in cognitive abilities. The AD continuum encompasses a prodormal stage known as Mild Cognitive Impairment (MCI), where patients may either progress to AD (MCIc) or remain stable (MCInc). Understanding the underlying mechanisms of AD requires complementary analysis derived from different data sources, leading to the development of multimodal deep learning models. In this study, we leveraged structural and functional Magnetic Resonance Imaging (sMRI/fMRI) to investigate the disease-induced grey matter and functional network connectivity changes. Moreover, considering AD's strong genetic component, we introduce Single Nucleotide Polymorphisms (SNPs) as a third channel. Given such diverse inputs, missing one or more modalities is a typical concern of multimodal methods. We hence propose a novel deep learning based classification framework where generative module employing Cycle Generative Adversarial Networks (cGAN) was adopted to impute missing data within the latent space. Additionally, we adopted an Explainable Artificial Intelligence (XAI) method, Integrated Gradients (IG), to extract input features relevance, enhancing our understanding of the learned representations. Two critical tasks were addressed: AD detection and MCI conversion prediction. Experimental results showed that our framework was able to reach the state-of-the-art in the classification of CN vs AD reaching an average test accuracy of 0.926 ± 0.02. For the MCInc vs MCIc task, we achieved an average prediction accuracy of 0.711 ± 0.01 using the pre-trained model for CN and AD. The interpretability analysis revealed that the classification performance was led by significant grey matter modulations in cortical and subcortical brain areas well known for their association with AD. Moreover, impairments in sensory-motor and visual resting state network connectivity along the disease continuum, as well as mutations in SNPs defining biological processes linked to amyloid-beta and cholesterol formation clearance and regulation, were identified as contributors to the achieved performance. Overall, our integrative deep learning approach shows promise for AD detection and MCI prediction, while shading light on important biological insights.
Collapse
Affiliation(s)
- Giorgio Dolci
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, USA
| | - Federica Cruciani
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Md Abdur Rahaman
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, USA
| | - Anees Abrol
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, USA
| | - Jiayu Chen
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, USA
| | - Zening Fu
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, USA
| | | | - Gloria Menegaz
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Vince D Calhoun
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, USA
| |
Collapse
|
74
|
Nitrini R. What is "biological Alzheimer's disease"? Dement Neuropsychol 2024; 18:e2024E001. [PMID: 38863570 PMCID: PMC11165693 DOI: 10.1590/1980-5764-dn-2024-e001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 06/13/2024] Open
Affiliation(s)
- Ricardo Nitrini
- Universidade de São Paulo, Faculdade de Medicina, São Paulo SP, Brazil
| |
Collapse
|
75
|
Burkett BJ, Johnson DR, Lowe VJ. Evaluation of Neurodegenerative Disorders with Amyloid-β, Tau, and Dopaminergic PET Imaging: Interpretation Pitfalls. J Nucl Med 2024; 65:829-837. [PMID: 38664015 DOI: 10.2967/jnumed.123.266463] [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: 11/08/2023] [Revised: 04/03/2024] [Indexed: 06/05/2024] Open
Abstract
Antiamyloid therapies for Alzheimer disease recently entered clinical practice, making imaging biomarkers for Alzheimer disease even more relevant to guiding patient management. Amyloid and tau PET are valuable tools that can provide objective evidence of Alzheimer pathophysiology in living patients and will increasingly be used to complement 18F-FDG PET in the diagnostic evaluation of cognitive impairment and dementia. Parkinsonian syndromes, also common causes of dementia, can likewise be evaluated with a PET imaging biomarker,18F-DOPA, allowing in vivo assessment of the presynaptic dopaminergic neurons. Understanding the role of these PET biomarkers will help the nuclear medicine physician contribute to the appropriate diagnosis and management of patients with cognitive impairment and dementia. To successfully evaluate brain PET examinations for neurodegenerative diseases, knowledge of the necessary protocol details for obtaining a reliable imaging study, inherent limitations for each PET radiopharmaceutical, and pitfalls in image interpretation is critical. This review will focus on underlying concepts for interpreting PET examinations, important procedural details, and guidance for avoiding potential interpretive pitfalls for amyloid, tau, and dopaminergic PET examinations.
Collapse
Affiliation(s)
| | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
76
|
Morais-Ribeiro R, Almeida FC, Coelho A, Oliveira TG. Differential atrophy along the longitudinal hippocampal axis in Alzheimer's disease. Eur J Neurosci 2024; 59:3376-3388. [PMID: 38654447 DOI: 10.1111/ejn.16361] [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: 02/04/2023] [Accepted: 04/03/2024] [Indexed: 04/26/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that primarily affects the hippocampus. Since hippocampal studies have highlighted a differential subregional regulation along its longitudinal axis, a more detailed analysis addressing subregional changes along the longitudinal hippocampal axis has the potential to provide new relevant biomarkers. This study included structural brain MRI data of 583 participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Cognitively normal (CN) subjects, mild cognitively impaired (MCI) subjects and AD patients were conveniently selected considering the age and sex match between clinical groups. Structural MRI acquisitions were pre-processed and analysed with a new longitudinal axis segmentation method, dividing the hippocampus in three subdivisions (anterior, intermediate, and posterior). When normalizing the volume of hippocampal sub-divisions to total hippocampus, the posterior hippocampus negatively correlates with age only in CN subjects (r = -.31). The longitudinal ratio of hippocampal atrophy (anterior sub-division divided by the posterior one) shows a significant increase with age only in CN (r = .25). Overall, in AD, the posterior hippocampus is predominantly atrophied early on. Consequently, the anterior/posterior hippocampal ratio is an AD differentiating metric at early disease stages with potential for diagnostic and prognostic applications.
Collapse
Affiliation(s)
- Rafaela Morais-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Francisco C Almeida
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Neuroradiology, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Ana Coelho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Division of Neuroradiology, Hospital de Braga, Braga, Portugal
| |
Collapse
|
77
|
Shanks HRC, Chen K, Reiman EM, Blennow K, Cummings JL, Massa SM, Longo FM, Börjesson-Hanson A, Windisch M, Schmitz TW. p75 neurotrophin receptor modulation in mild to moderate Alzheimer disease: a randomized, placebo-controlled phase 2a trial. Nat Med 2024; 30:1761-1770. [PMID: 38760589 PMCID: PMC11186782 DOI: 10.1038/s41591-024-02977-w] [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] [Accepted: 04/04/2024] [Indexed: 05/19/2024]
Abstract
p75 neurotrophin receptor (p75NTR) signaling pathways substantially overlap with degenerative networks active in Alzheimer disease (AD). Modulation of p75NTR with the first-in-class small molecule LM11A-31 mitigates amyloid-induced and pathological tau-induced synaptic loss in preclinical models. Here we conducted a 26-week randomized, placebo-controlled, double-blinded phase 2a safety and exploratory endpoint trial of LM11A-31 in 242 participants with mild to moderate AD with three arms: placebo, 200 mg LM11A-31 and 400 mg LM11A-31, administered twice daily by oral capsules. This trial met its primary endpoint of safety and tolerability. Within the prespecified secondary and exploratory outcome domains (structural magnetic resonance imaging, fluorodeoxyglucose positron-emission tomography and cerebrospinal fluid biomarkers), significant drug-placebo differences were found, consistent with the hypothesis that LM11A-31 slows progression of pathophysiological features of AD; no significant effect of active treatment was observed on cognitive tests. Together, these results suggest that targeting p75NTR with LM11A-31 warrants further investigation in larger-scale clinical trials of longer duration. EU Clinical Trials registration: 2015-005263-16 ; ClinicalTrials.gov registration: NCT03069014 .
Collapse
Grants
- R35 AG071476 NIA NIH HHS
- P30 AG072980 NIA NIH HHS
- SG-23-1038904 QC Alzheimer's Association
- 2022-00732 Vetenskapsrådet (Swedish Research Council)
- P20 GM109025 NIGMS NIH HHS
- R01 AG053798 NIA NIH HHS
- R35AG71476 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- ZEN-21-848495 Alzheimer's Association
- R01 AG051596 NIA NIH HHS
- P20GM109025 U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
- 453677 Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de Recherche en Santé du Canada)
- P20 AG068053 NIA NIH HHS
- 2017-00915 Vetenskapsrådet (Swedish Research Council)
- U01 AG024904 NIA NIH HHS
- R01AG053798 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- R25 AG083721-01 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- R25 AG083721 NIA NIH HHS
- Jonathan and Joshua Memorial Foundation Government of Ontario
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- State of Arizona
- Alzheimer’s Association
- the Swedish state under the agreement between the Swedish government and the County Councils, the ALF-agreement (#ALFGBG-715986 and #ALFGBG-965240), the Swedish Alzheimer Foundation (#AF-930351, #AF-939721 and #AF-968270), Hjärnfonden, Sweden (#FO2017-0243 and #ALZ2022-0006), La Fondation Recherche Alzheimer (FRA), Paris, France, the Kirsten and Freddy Johansen Foundation, Copenhagen, Denmark, and Familjen Rönströms Stiftelse, Stockholm, Sweden.
- U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
- Alzheimer’s Drug Discovery Foundation (ADDF)
- Ted and Maria Quirk Endowment; Joy Chambers-Grundy Endowment.
- San Francisco VA Health Care System
- National Institutes of Aging (NIA AD Pilot Trial 1R01AG051596) PharmatrophiX (Menlo Park, California)
- Alzheimer’s Society of Canada (176677)
Collapse
Affiliation(s)
- Hayley R C Shanks
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
- Robarts Research Institute, Western University, London, Ontario, Canada.
- Western Institute for Neuroscience, Western University, London, Ontario, Canada.
| | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA
- College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- College of Health Solutions, Arizona State University, Downtown, Phoenix, AZ, USA
| | - Eric M Reiman
- Banner Alzheimer's Institute, Phoenix, AZ, USA
- College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Translational Genomics Research Institute, Phoenix, AZ, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Stephen M Massa
- San Francisco Veterans Affairs Health Care System, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Anne Börjesson-Hanson
- Clinical Trials, Department of Aging, Karolinska University Hospital, Stockholm, Sweden
| | | | - Taylor W Schmitz
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
- Robarts Research Institute, Western University, London, Ontario, Canada.
- Western Institute for Neuroscience, Western University, London, Ontario, Canada.
| |
Collapse
|
78
|
Yu L, Wang T, Hansson O, Janelidze S, Lamar M, Arfanakis K, Bennett DA, Schneider JA, Boyle PA. MRI-Derived AD Signature of Cortical Thinning and Plasma P-Tau217 for Predicting Alzheimer Dementia Among Community-Dwelling Older Adults. Neurol Clin Pract 2024; 14:e200291. [PMID: 38720951 PMCID: PMC11073883 DOI: 10.1212/cpj.0000000000200291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/25/2024] [Indexed: 05/12/2024]
Abstract
Background and Objectives Structural brain MRI and blood-based phosphorylated tau (p-tau) measures are among the least invasive and least expensive Alzheimer's disease (AD) biomarkers to date. The extent to which these biomarkers may outperform one another in predicting future Alzheimer dementia diagnosis is poorly understood, however. This study investigated 2 specific AD biomarkers, i.e., a cortical thickness signature of AD (AD-CT) and plasma p-tau217, for predicting Alzheimer dementia. Methods Data came from community-dwelling older participants of the Religious Orders Study or the Rush Memory and Aging Project. AD-CT was obtained from 3T MRI scans using a magnetization-prepared rapid acquisition gradient echo sequence and by averaging thickness from previously identified cortical regions implicated in AD. Plasma p-tau217 was quantified using an immunoassay developed by Lilly Research Laboratories on the MSD platform. Both MRI scans and blood specimens were collected at the same visits, and subsequent diagnoses of Alzheimer dementia were determined through annual detailed clinical evaluations. Cox proportional hazards models examined the associations of the 2 biomarkers with incident Alzheimer dementia, and prediction accuracy was assessed using c-statistics. Results A total of 198 older adults, on average 84 years of age, were included. Over a mean follow-up of 4 years, 60 (30%) individuals developed Alzheimer dementia. AD-CT (hazard ratio: 1.71, 95% CI 1.26-2.31) and separately plasma p-tau217 (hazard ratio: 2.57, 95% CI 1.83-3.61) were associated with incident Alzheimer dementia. The c-statistic for prediction accuracy was consistently higher for plasma p-tau217 (between 0.74 and 0.81) than AD-CT (between 0.70 and 0.75) across a range of time horizons. Furthermore, with both biomarkers included in the same model, there was only modest improvement in the c-statistic due to AD-CT. Discussion Plasma p-tau217 outperforms an imaging-based cortical thickness signature of AD in predicting future Alzheimer dementia diagnosis. Furthermore, the AD cortical thickness signature adds little to the prediction accuracy above and beyond plasma p-tau217.
Collapse
Affiliation(s)
- Lei Yu
- Rush Alzheimer's Disease Center (LY, TW, ML, KA, DAB, JAS, PAB), Rush University Medical Center, Chicago, IL; and Clinical Memory Research Unit (OH, SJ), Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Tianhao Wang
- Rush Alzheimer's Disease Center (LY, TW, ML, KA, DAB, JAS, PAB), Rush University Medical Center, Chicago, IL; and Clinical Memory Research Unit (OH, SJ), Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Oskar Hansson
- Rush Alzheimer's Disease Center (LY, TW, ML, KA, DAB, JAS, PAB), Rush University Medical Center, Chicago, IL; and Clinical Memory Research Unit (OH, SJ), Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Shorena Janelidze
- Rush Alzheimer's Disease Center (LY, TW, ML, KA, DAB, JAS, PAB), Rush University Medical Center, Chicago, IL; and Clinical Memory Research Unit (OH, SJ), Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Melissa Lamar
- Rush Alzheimer's Disease Center (LY, TW, ML, KA, DAB, JAS, PAB), Rush University Medical Center, Chicago, IL; and Clinical Memory Research Unit (OH, SJ), Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Konstantinos Arfanakis
- Rush Alzheimer's Disease Center (LY, TW, ML, KA, DAB, JAS, PAB), Rush University Medical Center, Chicago, IL; and Clinical Memory Research Unit (OH, SJ), Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - David A Bennett
- Rush Alzheimer's Disease Center (LY, TW, ML, KA, DAB, JAS, PAB), Rush University Medical Center, Chicago, IL; and Clinical Memory Research Unit (OH, SJ), Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Julie A Schneider
- Rush Alzheimer's Disease Center (LY, TW, ML, KA, DAB, JAS, PAB), Rush University Medical Center, Chicago, IL; and Clinical Memory Research Unit (OH, SJ), Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Patricia A Boyle
- Rush Alzheimer's Disease Center (LY, TW, ML, KA, DAB, JAS, PAB), Rush University Medical Center, Chicago, IL; and Clinical Memory Research Unit (OH, SJ), Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
79
|
Vos SJB, Delvenne A, Jack CR, Thal DR, Visser PJ. The clinical importance of suspected non-Alzheimer disease pathophysiology. Nat Rev Neurol 2024; 20:337-346. [PMID: 38724589 DOI: 10.1038/s41582-024-00962-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 06/06/2024]
Abstract
The development of biomarkers for Alzheimer disease (AD) has led to the origin of suspected non-AD pathophysiology (SNAP) - a heterogeneous biomarker-based concept that describes individuals with normal amyloid and abnormal tau and/or neurodegeneration biomarker status. In this Review, we describe the origins of the SNAP construct, along with its prevalence, diagnostic and prognostic implications, and underlying neuropathology. As we discuss, SNAP can be operationalized using different biomarker modalities, which could affect prevalence estimates and reported characteristics of SNAP in ways that are not yet fully understood. Moreover, the underlying aetiologies that lead to a SNAP biomarker profile, and whether SNAP is the same in people with and without cognitive impairment, remains unclear. Improved insight into the clinical characteristics and pathophysiology of SNAP is of major importance for research and clinical practice, as well as for trial design to optimize care and treatment of individuals with SNAP.
Collapse
Affiliation(s)
- Stephanie J B Vos
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands.
| | - Aurore Delvenne
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Dietmar R Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
80
|
Plasa G, Hillier E, Luu J, Boutet D, Benovoy M, Friedrich MG. Automated Data Transformation and Feature Extraction for Oxygenation-Sensitive Cardiovascular Magnetic Resonance Images. J Cardiovasc Transl Res 2024; 17:705-715. [PMID: 38229001 DOI: 10.1007/s12265-023-10474-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/08/2023] [Indexed: 01/18/2024]
Abstract
Oxygenation-sensitive cardiovascular magnetic resonance (OS-CMR) is a novel, powerful tool for assessing coronary function in vivo. The data extraction and analysis however are labor-intensive. The objective of this study was to provide an automated approach for the extraction, visualization, and biomarker selection of OS-CMR images. We created a Python-based tool to automate extraction and export of raw patient data, featuring 3336 attributes per participant, into a template compatible with common data analytics frameworks, including the functionality to select predictive features for the given disease state. Each analysis was completed in about 2 min. The features selected by both ANOVA and MIC significantly outperformed (p < 0.001) the null set and complete set of features in two datasets, with mean AUROC scores of 0.89eatures f 0.94lete set of features in two datasets, with mean AUROC scores that our tool is suitable for automated data extraction and analysis of OS-CMR images.
Collapse
Affiliation(s)
- Glisant Plasa
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Faculty of Science, Neuroscience, McGill University, Montreal, Canada
- Area 19 Medical, Montreal, Canada
| | - Elizabeth Hillier
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Medicine and Health Sciences, Faculty of Medicine and Dentistry, McGill University, Montreal, Canada
| | - Judy Luu
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Dominic Boutet
- Faculty of Science, Neuroscience, McGill University, Montreal, Canada
| | - Mitchel Benovoy
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Area 19 Medical, Montreal, Canada
| | - Matthias G Friedrich
- Research Institute of the McGill University Health Centre, Montreal, Canada.
- Department of Medicine and Diagnostic Radiology, McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
81
|
Booth S, Ko JH. Radionuclide Imaging of the Neuroanatomical and Neurochemical Substrate of Cognitive Decline in Parkinson's Disease. Nucl Med Mol Imaging 2024; 58:213-226. [PMID: 38932760 PMCID: PMC11196570 DOI: 10.1007/s13139-024-00842-9] [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: 09/30/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 06/28/2024] Open
Abstract
Cognitive impairment is a frequent manifestation of Parkinson's disease (PD), resulting in decrease in patients' quality of life and increased societal and economic burden. However, cognitive decline in PD is highly heterogenous and the mechanisms are poorly understood. Radionuclide imaging techniques like positron emission tomography (PET) and single photon emission computed tomography (SPECT) have been used to investigate the neurochemical and neuroanatomical substrate of cognitive decline in PD. These techniques allow the assessment of different neurotransmitter systems, changes in brain glucose metabolism, proteinopathy, and neuroinflammation in vivo in PD patients. Here, we review current radionuclide imaging research on cognitive deficit in PD with a focus on predicting accelerating cognitive decline. This research could assist in the development of prognostic biomarkers for patient stratification and have utility in the development of ameliorative or disease-modifying therapies targeting cognitive deficit in PD.
Collapse
Affiliation(s)
- Samuel Booth
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, 130-745 Bannatyne Ave, Winnipeg, MB R3E 0J9 Canada
- PrairieNeuro Research Centre, Kleysen Institute of Advanced Medicine, Health Science Centre, Winnipeg, Canada
| | - Ji Hyun Ko
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, 130-745 Bannatyne Ave, Winnipeg, MB R3E 0J9 Canada
- PrairieNeuro Research Centre, Kleysen Institute of Advanced Medicine, Health Science Centre, Winnipeg, Canada
| |
Collapse
|
82
|
Sakaie K, Koenig K, Lerner A, Appleby B, Ogrocki P, Pillai JA, Rao S, Leverenz JB, Lowe MJ. Multi-shell diffusion MRI of the fornix as a biomarker for cognition in Alzheimer's disease. Magn Reson Imaging 2024; 109:221-226. [PMID: 38521367 DOI: 10.1016/j.mri.2024.03.030] [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: 07/25/2023] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND AND PURPOSE A substantial fraction of those who had Alzheimer's Disease (AD) pathology on autopsy did not have dementia in life. While biomarkers for AD pathology are well-developed, biomarkers specific to cognitive domains affected by early AD are lagging. Diffusion MRI (dMRI) of the fornix is a candidate biomarker for early AD-related cognitive changes but is susceptible to bias due to partial volume averaging (PVA) with cerebrospinal fluid. The purpose of this work is to leverage multi-shell dMRI to correct for PVA and to evaluate PVA-corrected dMRI measures in fornix as a biomarker for cognition in AD. METHODS Thirty-three participants in the Cleveland Alzheimer's Disease Research Center (CADRC) (19 with normal cognition (NC), 10 with mild cognitive impairment (MCI), 4 with dementia due to AD) were enrolled in this study. Multi-shell dMRI was acquired, and voxelwise fits were performed with two models: 1) diffusion tensor imaging (DTI) that was corrected for PVA and 2) neurite orientation dispersion and density imaging (NODDI). Values of tissue integrity in fornix were correlated with neuropsychological scores taken from the Uniform Data Set (UDS), including the UDS Global Composite 5 score (UDSGC5). RESULTS Statistically significant correlations were found between the UDSGC5 and PVA-corrected measure of mean diffusivity (MDc, r = -0.35, p < 0.05) from DTI and the intracelluar volume fraction (ficvf, r = 0.37, p < 0.04) from NODDI. A sensitivity analysis showed that the relationship to MDc was driven by episodic memory, which is often affected early in AD, and language. CONCLUSION This cross-sectional study suggests that multi-shell dMRI of the fornix that has been corrected for PVA is a potential biomarker for early cognitive domain changes in AD. A longitudinal study will be necessary to determine if the imaging measure can predict cognitive decline.
Collapse
Affiliation(s)
- Ken Sakaie
- Imaging Institute, The Cleveland Clinic, 9500 Euclid Ave, Mail code U-15, Cleveland, OH 44195, USA.
| | - Katherine Koenig
- Imaging Institute, The Cleveland Clinic, 9500 Euclid Ave, Mail code U-15, Cleveland, OH 44195, USA
| | - Alan Lerner
- Department of Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Brian Appleby
- Department of Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Paula Ogrocki
- Department of Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jagan A Pillai
- Lou Ruvo Center for Brain Health, The Cleveland Clinic, 9500 Euclid Ave, Mail code U-10, Cleveland, OH 44195, USA
| | - Stephen Rao
- Lou Ruvo Center for Brain Health, The Cleveland Clinic, 9500 Euclid Ave, Mail code U-10, Cleveland, OH 44195, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, The Cleveland Clinic, 9500 Euclid Ave, Mail code U-10, Cleveland, OH 44195, USA
| | - Mark J Lowe
- Imaging Institute, The Cleveland Clinic, 9500 Euclid Ave, Mail code U-15, Cleveland, OH 44195, USA
| |
Collapse
|
83
|
Benatar M, Wuu J, Huey ED, McMillan CT, Petersen RC, Postuma R, McHutchison C, Dratch L, Arias JJ, Crawley A, Houlden H, McDermott MP, Cai X, Thakur N, Boxer A, Rosen H, Boeve BF, Dacks P, Cosentino S, Abrahams S, Shneider N, Lingor P, Shefner J, Andersen PM, Al-Chalabi A, Turner MR. The Miami Framework for ALS and related neurodegenerative disorders: an integrated view of phenotype and biology. Nat Rev Neurol 2024; 20:364-376. [PMID: 38769202 PMCID: PMC11216694 DOI: 10.1038/s41582-024-00961-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2024] [Indexed: 05/22/2024]
Abstract
Increasing appreciation of the phenotypic and biological overlap between amyotrophic lateral sclerosis (ALS) and frontotemporal dementia, alongside evolving biomarker evidence for a pre-symptomatic stage of disease and observations that this stage of disease might not always be clinically silent, is challenging traditional views of these disorders. These advances have highlighted the need to adapt ingrained notions of these clinical syndromes to include both the full phenotypic continuum - from clinically silent, to prodromal, to clinically manifest - and the expanded phenotypic spectrum that includes ALS, frontotemporal dementia and some movement disorders. The updated clinical paradigms should also align with our understanding of the biology of these disorders, reflected in measurable biomarkers. The Miami Framework, emerging from discussions at the Second International Pre-Symptomatic ALS Workshop in Miami (February 2023; a full list of attendees and their affiliations appears in the Supplementary Information) proposes a classification system built on: first, three parallel phenotypic axes - motor neuron, frontotemporal and extrapyramidal - rather than the unitary approach of combining all phenotypic elements into a single clinical entity; and second, biomarkers that reflect different aspects of the underlying pathology and biology of neurodegeneration. This framework decouples clinical syndromes from biomarker evidence of disease and builds on experiences from other neurodegenerative diseases to offer a unified approach to specifying the pleiotropic clinical manifestations of disease and describing the trajectory of emergent biomarkers.
Collapse
Affiliation(s)
- Michael Benatar
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Joanne Wuu
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Edward D Huey
- Department of Psychiatry and Human Behaviour, Alpert Medical School of Brown University, Providence, RI, USA
| | - Corey T McMillan
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Ronald Postuma
- Department of Neurology, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Caroline McHutchison
- Human Cognitive Neuroscience, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK
| | - Laynie Dratch
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jalayne J Arias
- Department of Health Policy & Behavioral Sciences, School of Public Health, Georgia State University, Atlanta, GA, USA
| | | | - Henry Houlden
- UCL Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Michael P McDermott
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Xueya Cai
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | | | - Adam Boxer
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Howard Rosen
- Department of Neurology, University of California, San Francisco, CA, USA
| | | | - Penny Dacks
- Association for Frontotemporal Degeneration, King of Prussia, PA, USA
| | | | - Sharon Abrahams
- Human Cognitive Neuroscience, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK
| | - Neil Shneider
- Department of Neurology, Columbia University, New York, NY, USA
| | - Paul Lingor
- Department of Neurology, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Jeremy Shefner
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Peter M Andersen
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- Department of Neurology, King's College Hospital, London, UK
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
84
|
Su H, Masters CL, Bush AI, Barnham KJ, Reid GE, Vella LJ. Exploring the significance of lipids in Alzheimer's disease and the potential of extracellular vesicles. Proteomics 2024; 24:e2300063. [PMID: 37654087 DOI: 10.1002/pmic.202300063] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/07/2023] [Accepted: 08/14/2023] [Indexed: 09/02/2023]
Abstract
Lipids play a significant role in maintaining central nervous system (CNS) structure and function, and the dysregulation of lipid metabolism is known to occur in many neurological disorders, including Alzheimer's disease. Here we review what is currently known about lipid dyshomeostasis in Alzheimer's disease. We propose that small extracellular vesicle (sEV) lipids may provide insight into the pathophysiology and progression of Alzheimer's disease. This stems from the recognition that sEV likely contributes to disease pathogenesis, but also an understanding that sEV can serve as a source of potential biomarkers. While the protein and RNA content of sEV in the CNS diseases have been studied extensively, our understanding of the lipidome of sEV in the CNS is still in its infancy.
Collapse
Affiliation(s)
- Huaqi Su
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Ashley I Bush
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Kevin J Barnham
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Gavin E Reid
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - Laura J Vella
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
85
|
Bayazid AB, Lim BO. Therapeutic Effects of Plant Anthocyanin against Alzheimer's Disease and Modulate Gut Health, Short-Chain Fatty Acids. Nutrients 2024; 16:1554. [PMID: 38892488 PMCID: PMC11173718 DOI: 10.3390/nu16111554] [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: 04/23/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and neurogenerative disease (NDD), and it is also one of the leading causes of death worldwide. The number of AD patients is over 55 million according to 2020 Alzheimer's Disease International (ADI), and the number is increasing drastically without any effective cure. In this review, we discuss and analyze the potential role of anthocyanins (ACNs) against AD while understanding the molecular mechanisms. ACNs have been reported as having neuroprotective effects by mitigating cognitive impairments, apoptotic markers, neuroinflammation, aberrant amyloidogenesis, and tauopathy. Taken together, ACNs could be an important therapeutic agent for combating or delaying the onset of AD.
Collapse
Affiliation(s)
- Al Borhan Bayazid
- Medicinal Biosciences, Department of Applied Biological Sciences, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Republic of Korea
| | - Beong Ou Lim
- Medicinal Biosciences, Department of Applied Biological Sciences, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Republic of Korea
- Human Bioscience Corporate R&D Center, Human Bioscience Corp., 268 Chungwondaero, Chungju 27478, Republic of Korea
| |
Collapse
|
86
|
Jacobs T, Jacobson SR, Fortea J, Berger JS, Vedvyas A, Marsh K, He T, Gutierrez-Jimenez E, Fillmore NR, Gonzalez M, Figueredo L, Gaggi NL, Plaska CR, Pomara N, Blessing E, Betensky R, Rusinek H, Zetterberg H, Blennow K, Glodzik L, Wisniweski TM, de Leon MJ, Osorio RS, Ramos-Cejudo J. The neutrophil to lymphocyte ratio associates with markers of Alzheimer's disease pathology in cognitively unimpaired elderly people. Immun Ageing 2024; 21:32. [PMID: 38760856 PMCID: PMC11100119 DOI: 10.1186/s12979-024-00435-2] [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: 03/11/2024] [Accepted: 04/29/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND An elevated neutrophil-lymphocyte ratio (NLR) in blood has been associated with Alzheimer's disease (AD). However, an elevated NLR has also been implicated in many other conditions that are risk factors for AD, prompting investigation into whether the NLR is directly linked with AD pathology or a result of underlying comorbidities. Herein, we explored the relationship between the NLR and AD biomarkers in the cerebrospinal fluid (CSF) of cognitively unimpaired (CU) subjects. Adjusting for sociodemographics, APOE4, and common comorbidities, we investigated these associations in two cohorts: the Alzheimer's Disease Neuroimaging Initiative (ADNI) and the M.J. de Leon CSF repository at NYU. Specifically, we examined associations between the NLR and cross-sectional measures of amyloid-β42 (Aβ42), total tau (t-tau), and phosphorylated tau181 (p-tau), as well as the trajectories of these CSF measures obtained longitudinally. RESULTS A total of 111 ADNI and 190 NYU participants classified as CU with available NLR, CSF, and covariate data were included. Compared to NYU, ADNI participants were older (73.79 vs. 61.53, p < 0.001), had a higher proportion of males (49.5% vs. 36.8%, p = 0.042), higher BMIs (27.94 vs. 25.79, p < 0.001), higher prevalence of hypertensive history (47.7% vs. 16.3%, p < 0.001), and a greater percentage of Aβ-positivity (34.2% vs. 20.0%, p = 0.009). In the ADNI cohort, we found cross-sectional associations between the NLR and CSF Aβ42 (β = -12.193, p = 0.021), but not t-tau or p-tau. In the NYU cohort, we found cross-sectional associations between the NLR and CSF t-tau (β = 26.812, p = 0.019) and p-tau (β = 3.441, p = 0.015), but not Aβ42. In the NYU cohort alone, subjects classified as Aβ + (n = 38) displayed a stronger association between the NLR and t-tau (β = 100.476, p = 0.037) compared to Aβ- subjects or the non-stratified cohort. In both cohorts, the same associations observed in the cross-sectional analyses were observed after incorporating longitudinal CSF data. CONCLUSIONS We report associations between the NLR and Aβ42 in the older ADNI cohort, and between the NLR and t-tau and p-tau in the younger NYU cohort. Associations persisted after adjusting for comorbidities, suggesting a direct link between the NLR and AD. However, changes in associations between the NLR and specific AD biomarkers may occur as part of immunosenescence.
Collapse
Affiliation(s)
- Tovia Jacobs
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, Division of Brain Aging, 145 East 32Nd Street, New York, NY, 10016, USA
| | - Sean R Jacobson
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, Division of Brain Aging, 145 East 32Nd Street, New York, NY, 10016, USA
- VA Boston Cooperative Studies Program, MAVERIC, VA Boston Healthcare System, Boston, MA, USA
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de La Santa Creu y Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jeffrey S Berger
- Divisions of Cardiology and Hematology, Department of Medicine, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Alok Vedvyas
- Department of Neurology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Karyn Marsh
- Department of Neurology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Tianshe He
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, Division of Brain Aging, 145 East 32Nd Street, New York, NY, 10016, USA
| | | | - Nathanael R Fillmore
- VA Boston Cooperative Studies Program, MAVERIC, VA Boston Healthcare System, Boston, MA, USA
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Moses Gonzalez
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, Division of Brain Aging, 145 East 32Nd Street, New York, NY, 10016, USA
| | - Luisa Figueredo
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, Division of Brain Aging, 145 East 32Nd Street, New York, NY, 10016, USA
| | - Naomi L Gaggi
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, Division of Brain Aging, 145 East 32Nd Street, New York, NY, 10016, USA
| | - Chelsea Reichert Plaska
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, Division of Brain Aging, 145 East 32Nd Street, New York, NY, 10016, USA
- Nathan Kline Institute, 140 Old Orangeburg Rd, Orangeburg, NY, 10962, USA
| | - Nunzio Pomara
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, Division of Brain Aging, 145 East 32Nd Street, New York, NY, 10016, USA
- Nathan Kline Institute, 140 Old Orangeburg Rd, Orangeburg, NY, 10962, USA
- Department of Pathology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Esther Blessing
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, Division of Brain Aging, 145 East 32Nd Street, New York, NY, 10016, USA
| | - Rebecca Betensky
- Department of Neurology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Henry Rusinek
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, Division of Brain Aging, 145 East 32Nd Street, New York, NY, 10016, USA
- Department of Radiology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Inst. of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute On Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, People's Republic of China
| | - Lidia Glodzik
- Department of Neurology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Thomas M Wisniweski
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, Division of Brain Aging, 145 East 32Nd Street, New York, NY, 10016, USA
- Department of Neurology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
- Department of Pathology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Mony J de Leon
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
- Retired director of Center for Brain Health, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Ricardo S Osorio
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, Division of Brain Aging, 145 East 32Nd Street, New York, NY, 10016, USA.
- Nathan Kline Institute, 140 Old Orangeburg Rd, Orangeburg, NY, 10962, USA.
| | - Jaime Ramos-Cejudo
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, Division of Brain Aging, 145 East 32Nd Street, New York, NY, 10016, USA.
- VA Boston Cooperative Studies Program, MAVERIC, VA Boston Healthcare System, Boston, MA, USA.
| |
Collapse
|
87
|
Gaur A, Gallagher D, Herrmann N, Chen JJ, Marzolini S, Oh P, Amemiya Y, Seth A, Kiss A, Lanctôt KL. Neurofilament Light Chain as a Biomarker of Global Cognition in Individuals With Possible Vascular Mild Cognitive Impairment. J Geriatr Psychiatry Neurol 2024:8919887241254469. [PMID: 38757180 DOI: 10.1177/08919887241254469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
BACKGROUND Neurofilament Light Chain (NfL) is a biomarker of axonal injury elevated in mild cognitive impairment (MCI) and Alzheimer's disease dementia. Blood NfL also inversely correlates with cognitive performance in those conditions. However, few studies have assessed NfL as a biomarker of global cognition in individuals demonstrating mild cognitive deficits who are at risk for vascular-related cognitive decline. OBJECTIVE To assess the relationship between blood NfL and global cognition in individuals with possible vascular MCI (vMCI) throughout cardiac rehabilitation (CR). Additionally, NfL levels were compared to age/sex-matched cognitively unimpaired (CU) controls. METHOD Participants with coronary artery disease (vMCI or CU) were recruited at entry to a 24-week CR program. Global cognition was measured using the Montreal Cognitive Assessment (MoCA) and plasma NfL level (pg/ml) was quantified using a highly sensitive enzyme-linked immunosorbent assay. RESULTS Higher plasma NfL was correlated with worse MoCA scores at baseline (β = -.352, P = .029) in 43 individuals with vMCI after adjusting for age, sex, and education. An increase in NfL was associated with worse global cognition (b[SE] = -4.81[2.06], P = .023) over time, however baseline NfL did not predict a decline in global cognition. NfL levels did not differ between the vMCI (n = 39) and CU (n = 39) groups (F(1, 76) = 1.37, P = .245). CONCLUSION Plasma NfL correlates with global cognition at baseline in individuals with vMCI, and is associated with decline in global cognition during CR. Our findings increase understanding of NfL and neurobiological mechanisms associated with cognitive decline in vMCI.
Collapse
Affiliation(s)
- Amish Gaur
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
| | - Damien Gallagher
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Nathan Herrmann
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Jinghan Jenny Chen
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
| | - Susan Marzolini
- KITE Research Institute, Toronto Rehabilitation Institute, Toronto, ON, Canada
| | - Paul Oh
- KITE Research Institute, Toronto Rehabilitation Institute, Toronto, ON, Canada
| | - Yutaka Amemiya
- Genomics Core Facility, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Arun Seth
- Genomics Core Facility, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Alex Kiss
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Krista L Lanctôt
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- KITE Research Institute, Toronto Rehabilitation Institute, Toronto, ON, Canada
| |
Collapse
|
88
|
Mendes AJ, Ribaldi F, Lathuiliere A, Ashton NJ, Zetterberg H, Abramowicz M, Scheffler M, Assal F, Garibotto V, Blennow K, Frisoni GB. Comparison of plasma and neuroimaging biomarkers to predict cognitive decline in non-demented memory clinic patients. Alzheimers Res Ther 2024; 16:110. [PMID: 38755703 PMCID: PMC11097559 DOI: 10.1186/s13195-024-01478-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Plasma biomarkers of Alzheimer's disease (AD) pathology, neurodegeneration, and neuroinflammation are ideally suited for secondary prevention programs in self-sufficient persons at-risk of dementia. Plasma biomarkers have been shown to be highly correlated with traditional imaging biomarkers. However, their comparative predictive value versus traditional AD biomarkers is still unclear in cognitively unimpaired (CU) subjects and with mild cognitive impairment (MCI). METHODS Plasma (Aβ42/40, p-tau181, p-tau231, NfL, and GFAP) and neuroimaging (hippocampal volume, centiloid of amyloid-PET, and tau-SUVR of tau-PET) biomarkers were assessed at baseline in 218 non-demented subjects (CU = 140; MCI = 78) from the Geneva Memory Center. Global cognition (MMSE) was evaluated at baseline and at follow-ups up to 5.7 years. We used linear mixed-effects models and Cox proportional-hazards regression to assess the association between biomarkers and cognitive decline. Lastly, sample size calculations using the linear mixed-effects models were performed on subjects positive for amyloid-PET combined with tau-PET and plasma biomarker positivity. RESULTS Cognitive decline was significantly predicted in MCI by baseline plasma NfL (β=-0.55), GFAP (β=-0.36), hippocampal volume (β = 0.44), centiloid (β=-0.38), and tau-SUVR (β=-0.66) (all p < 0.05). Subgroup analysis with amyloid-positive MCI participants also showed that only NfL and GFAP were the only significant predictors of cognitive decline among plasma biomarkers. Overall, NfL and tau-SUVR showed the highest prognostic values (hazard ratios of 7.3 and 5.9). Lastly, we demonstrated that adding NfL to the inclusion criteria could reduce the sample sizes of future AD clinical trials by up to one-fourth in subjects with amyloid-PET positivity or by half in subjects with amyloid-PET and tau-PET positivity. CONCLUSIONS Plasma NfL and GFAP predict cognitive decline in a similar manner to traditional imaging techniques in amyloid-positive MCI patients. Hence, even though they are non-specific biomarkers of AD, both can be implemented in memory clinic workups as important prognostic biomarkers. Likewise, future clinical trials might employ plasma biomarkers as additional inclusion criteria to stratify patients at higher risk of cognitive decline to reduce sample sizes and enhance effectiveness.
Collapse
Affiliation(s)
- Augusto J Mendes
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland.
- Geneva Memory Center, Department of Rehabilitation and Geriatrics,, Geneva University Hospitals, Geneva, Switzerland.
| | - Federica Ribaldi
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Geneva Memory Center, Department of Rehabilitation and Geriatrics,, Geneva University Hospitals, Geneva, Switzerland
| | - Aurelien Lathuiliere
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Geneva Memory Center, Department of Rehabilitation and Geriatrics,, Geneva University Hospitals, Geneva, Switzerland
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer?s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Marc Abramowicz
- Genetic Medicine, Diagnostics Dept, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Max Scheffler
- Division of Radiology, Geneva University Hospitals, Geneva, Switzerland
| | - Frédéric Assal
- Division of Neurology, Department of Clinical Neurosciences, Faculty of Medicine, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Valentina Garibotto
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Faculty of Medicine, Geneva University Neurocenter, University of Geneva, Geneva, Switzerland
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, Geneva, Switzerland
- CIBM Center for Biomedical Imaging, Geneva, Switzerland
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Geneva Memory Center, Department of Rehabilitation and Geriatrics,, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
89
|
Mazzeo S, Morinelli C, Polito C, Giacomucci G, Moschini V, Ingannato A, Balestrini J, Frigerio D, Emiliani F, Galdo G, Crucitti C, Piazzesi D, Bagnoli S, Padiglioni S, Berti V, Sorbi S, Nacmias B, Bessi V. Data-driven subtypes of mixed semantic-logopenic primary progressive aphasia: Linguistic features, biomarker profiles and brain metabolic patterns. J Neurol Sci 2024; 460:122998. [PMID: 38615405 DOI: 10.1016/j.jns.2024.122998] [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: 02/12/2024] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 04/16/2024]
Abstract
Mixed primary progressive aphasia (mPPA) accounts for a substantial proportion of primary progressive aphasia (PPA) cases. However, the lack of a standardised definition of this condition has resulted in misclassification of PPA cases. In this study, we enrolled 55 patients diagnosed with PPA, comprising 12 semantic variant (svPPA), 23 logopenic variant (lvPPA), and 20 mPPA cases with linguistic characteristics consistent with both svPPA and lvPPA (s/lvPPA). All patients underwent language assessments, evaluation of Alzheimer's disease biomarkers (via cerebrospinal fluid analysis or Amyloid-PET), and 18F-FDG-PET brain scans. An agglomerative hierarchical clustering (AHC) analysis based on linguistic characteristics revealed two distinct clusters within the s/lvPPA group: cluster k1 (n = 10) displayed an AD-like biomarker profile, with lower levels of Aβ42 and Aβ42/Aβ40 ratio, along with higher levels of t-tau and p-tau compared to cluster k2 (n = 10). Interestingly, k1 exhibited linguistic features that were similar to those of svPPA. Both clusters exhibited extensive temporoparietal hypometabolism. These findings support the hypothesis that a subgroup of s/lvPPA may represent a clinical manifestation of AD-related PPA.
Collapse
Affiliation(s)
- Salvatore Mazzeo
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Italy; Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy; Vita-Salute San Raffaele University, Milan, Italy; IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Carmen Morinelli
- Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | | | - Giulia Giacomucci
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Italy
| | - Valentina Moschini
- Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Italy
| | - Juri Balestrini
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Italy
| | - Daniele Frigerio
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Italy
| | - Filippo Emiliani
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Italy
| | - Giulia Galdo
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Italy
| | - Chiara Crucitti
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Italy
| | - Diletta Piazzesi
- Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Italy
| | - Sonia Padiglioni
- Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy; Regional Referral Centre for Relational Criticalities, 50139 Tuscany Region, Italy
| | - Valentina Berti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Italy; Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.
| |
Collapse
|
90
|
Mestre TA, Stebbins GT, Stephenson D, Dexter D, Lee KK, Xiao Y, Dam T, Kopil CM, Simuni T. Patient-centered development of clinical outcome assessments in early Parkinson disease: key priorities and advances. NPJ Parkinsons Dis 2024; 10:101. [PMID: 38744872 PMCID: PMC11094181 DOI: 10.1038/s41531-024-00716-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 04/25/2024] [Indexed: 05/16/2024] Open
Affiliation(s)
- Tiago A Mestre
- Ottawa Hospital Research Institute; University of Ottawa Brain and Mind Research Institute; Division of Neurology, Department of Medicine, University of Ottawa, The Ottawa Hospital Ottawa, Ottawa, ON, Canada.
| | - Glenn T Stebbins
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | | | | | | | - Yuge Xiao
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Tien Dam
- Neumora Therapeutics, Cambridge, MA, USA
| | - Catherine M Kopil
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Tanya Simuni
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
91
|
Burkhart MC, Lee LY, Vaghari D, Toh AQ, Chong E, Chen C, Tiňo P, Kourtzi Z. Unsupervised multimodal modeling of cognitive and brain health trajectories for early dementia prediction. Sci Rep 2024; 14:10755. [PMID: 38729989 PMCID: PMC11087538 DOI: 10.1038/s41598-024-60914-w] [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: 12/13/2023] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Predicting the course of neurodegenerative disorders early has potential to greatly improve clinical management and patient outcomes. A key challenge for early prediction in real-world clinical settings is the lack of labeled data (i.e., clinical diagnosis). In contrast to supervised classification approaches that require labeled data, we propose an unsupervised multimodal trajectory modeling (MTM) approach based on a mixture of state space models that captures changes in longitudinal data (i.e., trajectories) and stratifies individuals without using clinical diagnosis for model training. MTM learns the relationship between states comprising expensive, invasive biomarkers (β-amyloid, grey matter density) and readily obtainable cognitive observations. MTM training on trajectories stratifies individuals into clinically meaningful clusters more reliably than MTM training on baseline data alone and is robust to missing data (i.e., cognitive data alone or single assessments). Extracting an individualized cognitive health index (i.e., MTM-derived cluster membership index) allows us to predict progression to AD more precisely than standard clinical assessments (i.e., cognitive tests or MRI scans alone). Importantly, MTM generalizes successfully from research cohort to real-world clinical data from memory clinic patients with missing data, enhancing the clinical utility of our approach. Thus, our multimodal trajectory modeling approach provides a cost-effective and non-invasive tool for early dementia prediction without labeled data (i.e., clinical diagnosis) with strong potential for translation to clinical practice.
Collapse
Affiliation(s)
- Michael C Burkhart
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK
| | - Liz Y Lee
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK
| | - Delshad Vaghari
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK
| | - An Qi Toh
- Department of Pharmacology, Memory, Aging, and Cognition Center, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Eddie Chong
- Department of Pharmacology, Memory, Aging, and Cognition Center, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christopher Chen
- Department of Pharmacology, Memory, Aging, and Cognition Center, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Peter Tiňo
- School of Computer Science, University of Birmingham, Birmingham, B15 2TT, UK
| | - Zoe Kourtzi
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK.
| |
Collapse
|
92
|
Genius P, Calle ML, Rodríguez-Fernández B, Minguillon C, Cacciaglia R, Garrido-Martin D, Esteller M, Navarro A, Gispert JD, Vilor-Tejedor N. Compositional structural brain signatures capture Alzheimer's genetic risk on brain structure along the disease continuum. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.08.24307046. [PMID: 38766190 PMCID: PMC11100942 DOI: 10.1101/2024.05.08.24307046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
INTRODUCTION Traditional brain imaging genetics studies have primarily focused on how genetic factors influence the volume of specific brain regions, often neglecting the overall complexity of brain architecture and its genetic underpinnings. METHODS This study analyzed data from participants across the Alzheimer's disease (AD) continuum from the ALFA and ADNI studies. We exploited compositional data analysis to examine relative brain volumetric variations that (i) differentiate cognitively unimpaired (CU) individuals, defined as amyloid-negative (A-) based on CSF profiling, from those at different AD stages, and (ii) associated with increased genetic susceptibility to AD, assessed using polygenic risk scores. RESULTS Distinct brain signatures differentiated CU A-individuals from amyloid-positive MCI and AD. Moreover, disease stage-specific signatures were associated with higher genetic risk of AD. DISCUSSION The findings underscore the complex interplay between genetics and disease stages in shaping brain structure, which could inform targeted preventive strategies and interventions in preclinical AD.
Collapse
|
93
|
Subedi L, Gaire BP, Koronyo Y, Koronyo-Hamaoui M, Crother TR. Chlamydia pneumoniae in Alzheimer's disease pathology. Front Neurosci 2024; 18:1393293. [PMID: 38770241 PMCID: PMC11102982 DOI: 10.3389/fnins.2024.1393293] [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: 02/28/2024] [Accepted: 04/22/2024] [Indexed: 05/22/2024] Open
Abstract
While recent advances in diagnostics and therapeutics offer promising new approaches for Alzheimer's disease (AD) diagnosis and treatment, there is still an unmet need for an effective remedy, suggesting new avenues of research are required. Besides many plausible etiologies for AD pathogenesis, mounting evidence supports a possible role for microbial infections. Various microbes have been identified in the postmortem brain tissues of human AD patients. Among bacterial pathogens in AD, Chlamydia pneumoniae (Cp) has been well characterized in human AD brains and is a leading candidate for an infectious involvement. However, no definitive studies have been performed proving or disproving Cp's role as a causative or accelerating agent in AD pathology and cognitive decline. In this review, we discuss recent updates for the role of Cp in human AD brains as well as experimental models of AD. Furthermore, based on the current literature, we have compiled a list of potential mechanistic pathways which may connect Cp with AD pathology.
Collapse
Affiliation(s)
- Lalita Subedi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children's at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Bhakta Prasad Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Timothy R. Crother
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children's at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
94
|
He K, Li B, Wang J, Wang Y, You Z, Chen X, Chen H, Li J, Huang Q, Guo Q, Huang YH, Guan Y, Chen K, Zhao J, Deng Y, Xie F. APOE ε4 is associated with decreased synaptic density in cognitively impaired participants. Alzheimers Dement 2024; 20:3157-3166. [PMID: 38477490 PMCID: PMC11095422 DOI: 10.1002/alz.13775] [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: 09/30/2023] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 03/14/2024]
Abstract
INTRODUCTION We aimed to investigate the effect of apolipoprotein E4 (APOE) ε4 on synaptic density in cognitively impaired (CI) participants. METHODS One hundred ten CI participants underwent amyloid positron emission tomography (PET) with 18F-florbetapir and synaptic density PET with 18F-SynVesT-1. We evaluated the influence of APOE ε4 allele on synaptic density and investigated the effects of ε4 genotype on the associations of synaptic density with Alzheimer's disease (AD) biomarkers. The mediation effects of AD biomarkers on ε4-associated synaptic density loss were analyzed. RESULTS Compared with non-carriers, APOE ε4 allele carriers exhibited significant synaptic loss in the medial temporal lobe. Amyloid beta (Aβ) and tau pathology mediated the effects of APOE ε4 on synaptic density to different extents. The associations between synaptic density and tau pathology were regulated by the APOE ε4 genotype. DISCUSSION The APOE ε4 allele was associated with decreased synaptic density in CI individuals and may be driven by AD biomarkers.
Collapse
Affiliation(s)
- Kun He
- Department of Nuclear Medicine & PET CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Binyin Li
- Department of Neurology & Institute of Neurology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Clinical Neuroscience CenterRuijin Hospital LuWan BranchShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jie Wang
- Department of Nuclear Medicine & PET CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Ying Wang
- Department of GerontologyShanghai Jiaotong University Affiliated Sixth People's HospitalShanghaiChina
| | - Zhiwen You
- Department of Nuclear MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Xing Chen
- Department of Nuclear MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Haijuan Chen
- Department of Neurology & Institute of Neurology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Junpeng Li
- Department of Nuclear Medicine & PET CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Qi Huang
- Department of Nuclear Medicine & PET CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Qihao Guo
- Department of GerontologyShanghai Jiaotong University Affiliated Sixth People's HospitalShanghaiChina
| | - Yiyun Henry Huang
- PET CenterDepartment of Radiology and Biomedical ImagingYale University School of MedicineNew HavenUSA
| | - Yihui Guan
- Department of Nuclear Medicine & PET CenterHuashan Hospital, Fudan UniversityShanghaiChina
| | - Kewei Chen
- Banner Alzheimer InstituteArizona State University, University of Arizona and Arizona Alzheimer's ConsortiumPhoenixUSA
| | - Jun Zhao
- Department of Nuclear MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Yulei Deng
- Department of Neurology & Institute of Neurology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Clinical Neuroscience CenterRuijin Hospital LuWan BranchShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fang Xie
- Department of Nuclear Medicine & PET CenterHuashan Hospital, Fudan UniversityShanghaiChina
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|
95
|
Salvadó G, Horie K, Barthélemy NR, Vogel JW, Pichet Binette A, Chen CD, Aschenbrenner AJ, Gordon BA, Benzinger TLS, Holtzman DM, Morris JC, Palmqvist S, Stomrud E, Janelidze S, Ossenkoppele R, Schindler SE, Bateman RJ, Hansson O. Disease staging of Alzheimer's disease using a CSF-based biomarker model. NATURE AGING 2024; 4:694-708. [PMID: 38514824 PMCID: PMC11108782 DOI: 10.1038/s43587-024-00599-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 02/20/2024] [Indexed: 03/23/2024]
Abstract
Biological staging of individuals with Alzheimer's disease (AD) may improve diagnostic and prognostic workup of dementia in clinical practice and the design of clinical trials. In this study, we used the Subtype and Stage Inference (SuStaIn) algorithm to establish a robust biological staging model for AD using cerebrospinal fluid (CSF) biomarkers. Our analysis involved 426 participants from BioFINDER-2 and was validated in 222 participants from the Knight Alzheimer Disease Research Center cohort. SuStaIn identified a singular biomarker sequence and revealed that five CSF biomarkers effectively constituted a reliable staging model (ordered: Aβ42/40, pT217/T217, pT205/T205, MTBR-tau243 and non-phosphorylated mid-region tau). The CSF stages (0-5) demonstrated a correlation with increased abnormalities in other AD-related biomarkers, such as Aβ-PET and tau-PET, and aligned with longitudinal biomarker changes reflective of AD progression. Higher CSF stages at baseline were associated with an elevated hazard ratio of clinical decline. This study highlights a common molecular pathway underlying AD pathophysiology across all patients, suggesting that a single CSF collection can accurately indicate the presence of AD pathologies and characterize the stage of disease progression. The proposed staging model has implications for enhancing diagnostic and prognostic assessments in both clinical practice and the design of clinical trials.
Collapse
Affiliation(s)
- Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
| | - Kanta Horie
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Eisai, Inc., Nutley, NJ, USA
| | - Nicolas R Barthélemy
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jacob W Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Clinical Science, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Charles D Chen
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew J Aschenbrenner
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
96
|
Ansari MA, Tripathi T, Venkidasamy B, Monziani A, Rajakumar G, Alomary MN, Alyahya SA, Onimus O, D'souza N, Barkat MA, Al-Suhaimi EA, Samynathan R, Thiruvengadam M. Multifunctional Nanocarriers for Alzheimer's Disease: Befriending the Barriers. Mol Neurobiol 2024; 61:3042-3089. [PMID: 37966683 DOI: 10.1007/s12035-023-03730-z] [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: 04/19/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023]
Abstract
Neurodegenerative diseases (NDDs) have been increasing in incidence in recent years and are now widespread worldwide. Neuronal death is defined as the progressive loss of neuronal structure or function which is closely associated with NDDs and represents the intrinsic features of such disorders. Amyotrophic lateral sclerosis, frontotemporal dementia, Alzheimer's, Parkinson's, and Huntington's diseases (AD, PD, and HD, respectively) are considered neurodegenerative diseases that affect a large number of people worldwide. Despite the testing of various drugs, there is currently no available therapy that can remedy or effectively slow the progression of these diseases. Nanomedicine has the potential to revolutionize drug delivery for the management of NDDs. The use of nanoparticles (NPs) has recently been developed to improve drug delivery efficiency and is currently subjected to extensive studies. Nanoengineered particles, known as nanodrugs, can cross the blood-brain barrier while also being less invasive compared to the most treatment strategies in use. Polymeric, magnetic, carbonic, and inorganic NPs are examples of NPs that have been developed to improve drug delivery efficiency. Primary research studies using NPs to cure AD are promising, but thorough research is needed to introduce these approaches to clinical use. In the present review, we discussed the role of metal-based NPs, polymeric nanogels, nanocarrier systems such as liposomes, solid lipid NPs, polymeric NPs, exosomes, quantum dots, dendrimers, polymersomes, carbon nanotubes, and nanofibers and surfactant-based systems for the therapy of neurodegenerative diseases. In addition, we highlighted nanoformulations such as N-butyl cyanoacrylate, poly(butyl cyanoacrylate), D-penicillamine, citrate-coated peptide, magnetic iron oxide, chitosan (CS), lipoprotein, ceria, silica, metallic nanoparticles, cholinesterase inhibitors, an acetylcholinesterase inhibitors, metal chelators, anti-amyloid, protein, and peptide-loaded NPs for the treatment of AD.
Collapse
Affiliation(s)
- Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research & Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441, Dammam, Saudi Arabia
| | - Takshashila Tripathi
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Baskar Venkidasamy
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Alan Monziani
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Govindasamy Rajakumar
- Department of Orthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Mohammad N Alomary
- Advanced Diagnostic and Therapeutic Institute, King Abdulaziz City for Science and Technology, 11442, Riyadh, Saudi Arabia
| | - Sami A Alyahya
- Wellness and Preventive Medicine Institute, King Abdulaziz City for Science and Technology, 11442, Riyadh, Saudi Arabia
| | - Oriane Onimus
- Faculty of Basic and Biomedical Sciences, University of Paris, Paris, France
| | - Naomi D'souza
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Md Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin, Saudi Arabia
| | - Ebtesam A Al-Suhaimi
- Research Consultation Department, Vice Presidency for Scientific Research and Innovation, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441, Dammam, Saudi Arabia
| | - Ramkumar Samynathan
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
97
|
Gérard T, Colmant L, Malotaux V, Salman Y, Huyghe L, Quenon L, Dricot L, Ivanoiu A, Lhommel R, Hanseeuw B. The spatial extent of tauopathy on [ 18F]MK-6240 tau PET shows stronger association with cognitive performances than the standard uptake value ratio in Alzheimer's disease. Eur J Nucl Med Mol Imaging 2024; 51:1662-1674. [PMID: 38228971 PMCID: PMC11043108 DOI: 10.1007/s00259-024-06603-2] [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] [Accepted: 01/04/2024] [Indexed: 01/18/2024]
Abstract
PURPOSE [18F]MK-6240, a second-generation tau PET tracer, is increasingly used for the detection and the quantification of in vivo cerebral tauopathy in Alzheimer's disease (AD). Given that neurological symptoms are better explained by the topography rather than by the nature of brain lesions, our study aimed to evaluate whether cognitive impairment would be more closely associated with the spatial extent than with the intensity of tau-PET signal, as measured by the standard uptake value ratio (SUVr). METHODS [18F]MK6240 tau-PET data from 82 participants in the AD spectrum were quantified in three different brain regions (Braak ≤ 2, Braak ≤ 4, and Braak ≤ 6) using SUVr and the extent of tauopathy (EOT, percentage of voxels with SUVr ≥ 1.3). PET data were first compared between diagnostic categories, and ROC curves were computed to evaluate sensitivity and specificity. PET data were then correlated to cognitive performances and cerebrospinal fluid (CSF) tau values. RESULTS The EOT in the Braak ≤ 2 region provided the highest diagnostic accuracies, distinguishing between amyloid-negative and positive clinically unimpaired individuals (threshold = 9%, sensitivity = 79%, specificity = 82%) as well as between prodromal AD and preclinical AD (threshold = 38%, sensitivity = 81%, specificity = 93%). The EOT better correlated with cognition than SUVr (∆R2 + 0.08-0.09) with the best correlation observed for EOT in the Braak ≤ 4 region (R2 = 0.64). Cognitive performances were more closely associated with PET metrics than with CSF values. CONCLUSIONS Quantifying [18F]MK-6240 tau PET in terms of EOT rather than SUVr significantly increases the correlation with cognitive performances. Quantification in the mesiotemporal lobe is the most useful to diagnose preclinical AD or prodromal AD.
Collapse
Affiliation(s)
- Thomas Gérard
- Nuclear Medicine Department, Cliniques Universitaires Saint Luc, Brussels, Belgium.
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium.
| | - Lise Colmant
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
- Neurology Department, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Vincent Malotaux
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
| | - Yasmine Salman
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
| | - Lara Huyghe
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
| | - Lisa Quenon
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
- Neurology Department, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Laurence Dricot
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
| | - Adrian Ivanoiu
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
- Neurology Department, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Renaud Lhommel
- Nuclear Medicine Department, Cliniques Universitaires Saint Luc, Brussels, Belgium
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
| | - Bernard Hanseeuw
- Institute of Neurosciences, Université Catholique de Louvain, Brussels, Belgium
- Neurology Department, Cliniques Universitaires Saint Luc, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Avenue Pasteur, 6, 1300, Wavre, Belgium
- Department of Radiology, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
98
|
Takemaru L, Yang S, Wu R, He B, Davtzikos C, Yan J, Shen L. MAPPING ALZHEIMER'S DISEASE PSEUDO-PROGRESSION WITH MULTIMODAL BIOMARKER TRAJECTORY EMBEDDINGS. PROCEEDINGS. IEEE INTERNATIONAL SYMPOSIUM ON BIOMEDICAL IMAGING 2024; 2024:10.1109/isbi56570.2024.10635249. [PMID: 39371469 PMCID: PMC11452153 DOI: 10.1109/isbi56570.2024.10635249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder characterized by progressive cognitive degeneration and motor impairment, affecting millions worldwide. Mapping the progression of AD is crucial for early detection of loss of brain function, timely intervention, and development of effective treatments. However, accurate measurements of disease progression are still challenging at present. This study presents a novel approach to understanding the heterogeneous pathways of AD through longitudinal biomarker data from medical imaging and other modalities. We propose an analytical pipeline adopting two popular machine learning methods from the single-cell transcriptomics domain, PHATE and Slingshot, to project multimodal biomarker trajectories to a low-dimensional space. These embeddings serve as our pseudotime estimates. We applied this pipeline to the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset to align longitudinal data across individuals at various disease stages. Our approach mirrors the technique used to cluster single-cell data into cell types based on developmental timelines. Our pseudotime estimates revealed distinct patterns of disease evolution and biomarker changes over time, providing a deeper understanding of the temporal dynamics of AD. The results show the potential of the approach in the clinical domain of neurodegenerative diseases, enabling more precise disease modeling and early diagnosis.
Collapse
Affiliation(s)
- Lina Takemaru
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shu Yang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruiming Wu
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bing He
- School of Informatics and Computing, Indiana University, Indianapolis, IN, USA
| | - Christos Davtzikos
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingwen Yan
- School of Informatics and Computing, Indiana University, Indianapolis, IN, USA
| | - Li Shen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
99
|
Doering S, McCullough A, Gordon BA, Chen CD, McKay N, Hobbs D, Keefe S, Flores S, Scott J, Smith H, Jarman S, Jackson K, Hornbeck RC, Ances BM, Xiong C, Aschenbrenner AJ, Hassenstab J, Cruchaga C, Daniels A, Bateman RJ, Morris JC, Benzinger TLS. Deconstructing pathological tau by biological process in early stages of Alzheimer disease: a method for quantifying tau spatial spread in neuroimaging. EBioMedicine 2024; 103:105080. [PMID: 38552342 PMCID: PMC10995809 DOI: 10.1016/j.ebiom.2024.105080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Neuroimaging studies often quantify tau burden in standardized brain regions to assess Alzheimer disease (AD) progression. However, this method ignores another key biological process in which tau spreads to additional brain regions. We have developed a metric for calculating the extent tau pathology has spread throughout the brain and evaluate the relationship between this metric and tau burden across early stages of AD. METHODS 445 cross-sectional participants (aged ≥ 50) who had MRI, amyloid PET, tau PET, and clinical testing were separated into disease-stage groups based on amyloid positivity and cognitive status (older cognitively normal control, preclinical AD, and symptomatic AD). Tau burden and tau spatial spread were calculated for all participants. FINDINGS We found both tau metrics significantly elevated across increasing disease stages (p < 0.0001) and as a function of increasing amyloid burden for participants with preclinical (p < 0.0001, p = 0.0056) and symptomatic (p = 0.010, p = 0.0021) AD. An interaction was found between tau burden and tau spatial spread when predicting amyloid burden (p = 0.00013). Analyses of slope between tau metrics demonstrated more spread than burden in preclinical AD (β = 0.59), but then tau burden elevated relative to spread (β = 0.42) once participants had symptomatic AD, when the tau metrics became highly correlated (R = 0.83). INTERPRETATION Tau burden and tau spatial spread are both strong biomarkers for early AD but provide unique information, particularly at the preclinical stage. Tau spatial spread may demonstrate earlier changes than tau burden which could have broad impact in clinical trial design. FUNDING This research was supported by the Knight Alzheimer Disease Research Center (Knight ADRC, NIH grants P30AG066444, P01AG026276, P01AG003991), Dominantly Inherited Alzheimer Network (DIAN, NIH grants U01AG042791, U19AG03243808, R01AG052550-01A1, R01AG05255003), and the Barnes-Jewish Hospital Foundation Willman Scholar Fund.
Collapse
Affiliation(s)
- Stephanie Doering
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Austin McCullough
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Brian A Gordon
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Charles D Chen
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Nicole McKay
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Diana Hobbs
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Sarah Keefe
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Shaney Flores
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Jalen Scott
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Hunter Smith
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Stephen Jarman
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Kelley Jackson
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Russ C Hornbeck
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Beau M Ances
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Chengjie Xiong
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | | | - Jason Hassenstab
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Carlos Cruchaga
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Alisha Daniels
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Randall J Bateman
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - John C Morris
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | | |
Collapse
|
100
|
Wisch JK, McKay NS, Boerwinkle AH, Kennedy J, Flores S, Handen BL, Christian BT, Head E, Mapstone M, Rafii MS, O'Bryant SE, Price JC, Laymon CM, Krinsky-McHale SJ, Lai F, Rosas HD, Hartley SL, Zaman S, Lott IT, Tudorascu D, Zammit M, Brickman AM, Lee JH, Bird TD, Cohen A, Chrem P, Daniels A, Chhatwal JP, Cruchaga C, Ibanez L, Jucker M, Karch CM, Day GS, Lee JH, Levin J, Llibre-Guerra J, Li Y, Lopera F, Roh JH, Ringman JM, Supnet-Bell C, van Dyck CH, Xiong C, Wang G, Morris JC, McDade E, Bateman RJ, Benzinger TLS, Gordon BA, Ances BM. Comparison of tau spread in people with Down syndrome versus autosomal-dominant Alzheimer's disease: a cross-sectional study. Lancet Neurol 2024; 23:500-510. [PMID: 38631766 PMCID: PMC11209765 DOI: 10.1016/s1474-4422(24)00084-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/01/2024] [Accepted: 02/21/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND In people with genetic forms of Alzheimer's disease, such as in Down syndrome and autosomal-dominant Alzheimer's disease, pathological changes specific to Alzheimer's disease (ie, accumulation of amyloid and tau) occur in the brain at a young age, when comorbidities related to ageing are not present. Studies including these cohorts could, therefore, improve our understanding of the early pathogenesis of Alzheimer's disease and be useful when designing preventive interventions targeted at disease pathology or when planning clinical trials. We compared the magnitude, spatial extent, and temporal ordering of tau spread in people with Down syndrome and autosomal-dominant Alzheimer's disease. METHODS In this cross-sectional observational study, we included participants (aged ≥25 years) from two cohort studies. First, we collected data from the Dominantly Inherited Alzheimer's Network studies (DIAN-OBS and DIAN-TU), which include carriers of autosomal-dominant Alzheimer's disease genetic mutations and non-carrier familial controls recruited in Australia, Europe, and the USA between 2008 and 2022. Second, we collected data from the Alzheimer Biomarkers Consortium-Down Syndrome study, which includes people with Down syndrome and sibling controls recruited from the UK and USA between 2015 and 2021. Controls from the two studies were combined into a single group of familial controls. All participants had completed structural MRI and tau PET (18F-flortaucipir) imaging. We applied Gaussian mixture modelling to identify regions of high tau PET burden and regions with the earliest changes in tau binding for each cohort separately. We estimated regional tau PET burden as a function of cortical amyloid burden for both cohorts. Finally, we compared the temporal pattern of tau PET burden relative to that of amyloid. FINDINGS We included 137 people with Down syndrome (mean age 38·5 years [SD 8·2], 74 [54%] male, and 63 [46%] female), 49 individuals with autosomal-dominant Alzheimer's disease (mean age 43·9 years [11·2], 22 [45%] male, and 27 [55%] female), and 85 familial controls, pooled from across both studies (mean age 41·5 years [12·1], 28 [33%] male, and 57 [67%] female), who satisfied the PET quality-control procedure for tau-PET imaging processing. 134 (98%) people with Down syndrome, 44 (90%) with autosomal-dominant Alzheimer's disease, and 77 (91%) controls also completed an amyloid PET scan within 3 years of tau PET imaging. Spatially, tau PET burden was observed most frequently in subcortical and medial temporal regions in people with Down syndrome, and within the medial temporal lobe in people with autosomal-dominant Alzheimer's disease. Across the brain, people with Down syndrome had greater concentrations of tau for a given level of amyloid compared with people with autosomal-dominant Alzheimer's disease. Temporally, increases in tau were more strongly associated with increases in amyloid for people with Down syndrome compared with autosomal-dominant Alzheimer's disease. INTERPRETATION Although the general progression of amyloid followed by tau is similar for people Down syndrome and people with autosomal-dominant Alzheimer's disease, we found subtle differences in the spatial distribution, timing, and magnitude of the tau burden between these two cohorts. These differences might have important implications; differences in the temporal pattern of tau accumulation might influence the timing of drug administration in clinical trials, whereas differences in the spatial pattern and magnitude of tau burden might affect disease progression. FUNDING None.
Collapse
Affiliation(s)
- Julie K Wisch
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA.
| | - Nicole S McKay
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Anna H Boerwinkle
- McGovern Medical School, University of Texas in Houston, Houston, TX, USA
| | - James Kennedy
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Shaney Flores
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Benjamin L Handen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bradley T Christian
- Department of Medical Physics and Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth Head
- Department of Pathology, Gillespie Neuroscience Research Facility, University of California, Irvine, CA, USA
| | - Mark Mapstone
- Department of Neurology, University of California Irvine School of Medicine, Irvine, CA, USA
| | - Michael S Rafii
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Sid E O'Bryant
- Institute for Translational Research Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Julie C Price
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Charles M Laymon
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sharon J Krinsky-McHale
- Department of Psychology, New York State Institute for Basic Research in Developmental Disabilities, New York, NY, USA
| | - Florence Lai
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - H Diana Rosas
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA; Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Sigan L Hartley
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Shahid Zaman
- Cambridge Intellectual and Developmental Disabilities Research Group, University of Cambridge, Cambridge, UK
| | - Ira T Lott
- Department of Pediatrics, University of California Irvine School of Medicine, Irvine, CA, USA
| | - Dana Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew Zammit
- Department of Medical Physics and Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Adam M Brickman
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Joseph H Lee
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA; Department of Epidemiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Thomas D Bird
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Annie Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patricio Chrem
- Centro de Memoria y Envejecimiento, Buenos Aires, Argentina
| | - Alisha Daniels
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA; Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, USA
| | - Laura Ibanez
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Mathias Jucker
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Celeste M Karch
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA; Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA; German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Gregory S Day
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Jae-Hong Lee
- Department of Neurology, University of Ulsan College of Medicine, Asian Medical Center, Seoul, South Korea
| | - Johannes Levin
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases, site Munich, Munich, Germany; Munich Cluster for Systems Neurology, Munich, Germany
| | - Jorge Llibre-Guerra
- Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA; Department of Biostatistics, Washington University in St Louis, St Louis, MO, USA
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Jee Hoon Roh
- Departments of Physiology and Neurology, Korea University College of Medicine, Seoul, South Korea
| | - John M Ringman
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of USC, Los Angeles, CA, USA
| | | | | | - Chengjie Xiong
- Department of Biostatistics, Washington University in St Louis, St Louis, MO, USA
| | - Guoqiao Wang
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA; Department of Biostatistics, Washington University in St Louis, St Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | | | - Brian A Gordon
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Beau M Ances
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| |
Collapse
|