151
|
Padovani A, Benussi A, Cotelli MS, Ferrari C, Cantoni V, Dell'Era V, Turrone R, Paghera B, Borroni B. Transcranial magnetic stimulation and amyloid markers in mild cognitive impairment: impact on diagnostic confidence and diagnostic accuracy. ALZHEIMERS RESEARCH & THERAPY 2019; 11:95. [PMID: 31787103 PMCID: PMC6886207 DOI: 10.1186/s13195-019-0555-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 11/08/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND The development of diagnostic tools capable of accurately identifying the pathophysiology of mild cognitive impairment (MCI) has become a crucial target considering the claim that disease-modifying treatments should be administered as early as possible in the disease course. Transcranial magnetic stimulation (TMS) protocols have demonstrated analytical validity in discriminating different forms of dementia; however, its value in daily clinical practice in MCI subjects is still unknown. OBJECTIVE To evaluate the clinical value of TMS compared to amyloid markers on diagnostic confidence and accuracy in MCI subjects, considering clinicians' expertise. METHODS One hundred seven MCI subjects were included and classified as MCI-Alzheimer disease (MCI-AD), MCI-frontotemporal dementia (MCI-FTD), MCI-dementia with Lewy bodies (MCI-DLB), or MCI-other in a three-step process based on (i) demographic, clinical, and neuropsychological evaluation (clinical work-up); (ii) clinical work-up PLUS amyloidosis markers or clinical work-up PLUS TMS measures; and (iii) clinical work-up PLUS both markers. Two blinded neurologists with different clinical expertise were asked to express a diagnostic confidence for each MCI subgroup, and ROC curve analyses were performed at each step. RESULTS The addition of TMS markers to clinical work-up significantly increased the diagnostic confidence for MCI-AD (p = 0.003), MCI-FTD (p = 0.044), and MCI-DLB (p = 0.033) compared to clinical work-up alone, but not for MCI-other (p > 0.05). No significant differences between the add-on effect of TMS and the add-on effect of amyloid markers to clinical work-up were observed (p > 0.732), while the diagnostic confidence further increased when both markers were available. The greater the clinical expertise, the greater the flexibility in considering alternative diagnosis, and the greater the ability to modify diagnostic confidence with TMS and amyloid markers. CONCLUSIONS TMS in addition to routine clinical assessment in MCI subjects has a significant effect on diagnostic accuracy and confidence, comparable to well-established biomarkers of amyloidosis.
Collapse
Affiliation(s)
- Alessandro Padovani
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, Clinica Neurologica, University of Brescia, P.le Spedali Civili, 1, 25100, Brescia, Italy
| | - Alberto Benussi
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, Clinica Neurologica, University of Brescia, P.le Spedali Civili, 1, 25100, Brescia, Italy
| | | | - Clarissa Ferrari
- Service of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Valentina Cantoni
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, Clinica Neurologica, University of Brescia, P.le Spedali Civili, 1, 25100, Brescia, Italy.,Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Valentina Dell'Era
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, Clinica Neurologica, University of Brescia, P.le Spedali Civili, 1, 25100, Brescia, Italy
| | - Rosanna Turrone
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, Clinica Neurologica, University of Brescia, P.le Spedali Civili, 1, 25100, Brescia, Italy
| | - Barbara Paghera
- Nuclear Medicine Unit, Spedali Civili Brescia, Brescia, Italy
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, Clinica Neurologica, University of Brescia, P.le Spedali Civili, 1, 25100, Brescia, Italy.
| |
Collapse
|
152
|
Ashton NJ, Ide M, Zetterberg H, Blennow K. Salivary Biomarkers for Alzheimer's Disease and Related Disorders. Neurol Ther 2019; 8:83-94. [PMID: 31833026 PMCID: PMC6908535 DOI: 10.1007/s40120-019-00168-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Indexed: 12/13/2022] Open
Abstract
The search for accessible and cost-effective biomarkers to complement current cerebrospinal fluid (CSF) and imaging biomarkers in the accurate detection of Alzheimer disease (AD) and other common neurodegenerative disorders remains a challenging task. The advances in ultra-sensitive detection methods has highlighted blood biomarkers (e.g. amyloid-β and neurofilament light) as a valuable and realistic tool in a diagnostic or screening process. Saliva, however, is also a rich source of potential biomarkers for disease detection and offers several practical advantages over biofluids that are currently examined for neurodegenerative disorders. However, while this may be true for the general population, challenges in collecting saliva from an elderly population should be seriously considered. In this review, we begin by discussing how saliva is produced and how age-related conditions can modify saliva production and composition. We then focus on the data available which support the concept of salivary amyloid-β, tau species and novel biomarkers in detecting AD and alpha-synuclein (α-syn) in Parkinson's disease (PD).
Collapse
Affiliation(s)
- Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute 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.
| | - Mark Ide
- Periodontology, Centre for Host Microbiome Interactions, Faculty of Dental, Oral and Craniofacial Sciences, King's College London, London, UK
- Mucosal and Salivary Biology, Centre for Host Microbiome Interactions, Faculty of Dental, Oral and Craniofacial Sciences, King's College London, London, UK
| | - 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
| | - Kaj Blennow
- 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
| |
Collapse
|
153
|
Okada Y, Kato T, Iwata K, Kimura Y, Nakamura A, Hattori H, Toyama H, Ishii K, Ishii K, Senda M, Ito K, Iwatsubo T. Evaluation of PiB visual interpretation with CSF Aβ and longitudinal SUVR in J-ADNI study. Ann Nucl Med 2019; 34:108-118. [PMID: 31749127 PMCID: PMC7026272 DOI: 10.1007/s12149-019-01420-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/11/2019] [Indexed: 12/14/2022]
Abstract
Objective The objectives of the present study were to investigate (1) whether trinary visual interpretation of amyloid positron emission tomography (PET) imaging (negative/equivocal/positive) reflects quantitative amyloid measurements and the time course of 11C-Pittsburgh compound B (PiB) amyloid accumulation, and (2) whether visually equivocal scans represent an early stage of the Alzheimer’s disease (AD) continuum in terms of an intermediate state of quantitative amyloid measurements and the changes in amyloid accumulation over time. Methods From the National Bioscience Database Center Human Database of the Japanese Alzheimer’s Disease Neuroimaging Initiative, we selected 133 individuals for this study including 33 with Alzheimer’s disease dementia (ADD), 52 with late mild cognitive impairment (LMCI), and 48 cognitively normal (CN) subjects who underwent clinical assessment, PiB PET, and structural magnetic resonance imaging (MRI) with 2 or 3-years of follow-up. Sixty-eight of the 133 individuals underwent cerebrospinal fluid amyloid-β1-42 (CSF-Ab42) analysis at baseline. The standard uptake value ratio (SUVR) of PiB PET was calculated with a method using MRI at each visit. The cross-sectional values, longitudinal changes in SUVR, and baseline CSF-Ab42 were compared among groups, which were categorized based on trinary visual reads of amyloid PET (negative/equivocal/positive). Results From the trinary visual interpretation of the PiB PET images, 55 subjects were negative, 8 were equivocal, and 70 were positive. Negative interpretation was most frequent in the CN group (70.8/10.4/18.8%: negative/equivocal/positive), and positive was most frequent in the LMCI group (34.6/1.9/63.5%) and in the ADD group (9.1/6.1/84.8%). The baseline SUVRs were 1.08 ± 0.06 in the negative group, 1.23 ± 0.15 in the equivocal group, and 1.86 ± 0.31 in the positive group (F = 174.9, p < 0.001). The baseline CSF-Ab42 level was 463 ± 112 pg/mL in the negative group, 383 ± 125 pg/mL in the equivocal group, and 264 ± 69 pg/mL in the positive group (F = 37, p < 0.001). Over the 3-year follow-up, annual changes in SUVR were − 0.00 ± 0.02 in the negative group, 0.02 ± 0.02 in the equivocal group, and 0.04 ± 0.07 in the positive group (F = 8.4, p < 0.001). Conclusions Trinary visual interpretation (negative/equivocal/positive) of amyloid PET imaging reflects quantitative amyloid measurements evaluated with PET and the CSF amyloid test as well as the amyloid accumulation over time evaluated with PET over 3 years. Subjects in the early stage of the AD continuum could be identified with an equivocal scan, because they showed intermediate quantitative amyloid PET, CSF measurements, and the amyloid accumulation over time. Electronic supplementary material The online version of this article (10.1007/s12149-019-01420-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yusuke Okada
- Department of Radiology, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, 470-1192, Aichi, Japan. .,Department of Psychiatry, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan. .,Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan.
| | - Takashi Kato
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan.,Department of Radiology, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan
| | - Kaori Iwata
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan
| | - Yasuyuki Kimura
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan
| | - Akinori Nakamura
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan
| | - Hideyuki Hattori
- Department of Psychiatry, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan
| | - Hiroshi Toyama
- Department of Radiology, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, 470-1192, Aichi, Japan
| | - Kazunari Ishii
- Department of Radiology, Kindai University Faculty of Medicine, 377-2 Onohigashi, Osakasayama, 589-8511, Osaka, Japan
| | - Kenji Ishii
- Diagnostic Neuroimaging Research, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Michio Senda
- Division of Molecular Imaging, Kobe City Medical Center General Hospital, 2-1-1, Minatojimaminamimachi, Chuo-ku, Kobe, 650-0047, Hyogo, Japan
| | - Kengo Ito
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan.,Department of Radiology, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan
| | - Takeshi Iwatsubo
- Department of Neuropathology, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | | |
Collapse
|
154
|
Umlauf A, Soontornniyomkij B, Sundermann EE, Gouaux B, Ellis RJ, Levine AJ, Moore DJ, Soontornniyomkij V. Risk of developing cerebral β-amyloid plaques with posttranslational modification among HIV-infected adults. AIDS 2019; 33:2157-2166. [PMID: 31688040 PMCID: PMC6852888 DOI: 10.1097/qad.0000000000002336] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Evidence of accelerated brain aging among HIV-infected adults argues for the increased risk of developing cerebral β-amyloid (Aβ) plaques. We compared the frequency of Aβ plaque-bearing cases in our HIV cohort with that in a general cohort reported by Braak et al. We explored posttranslationally modified Aβ forms (N3pE, E22P, phospho-Ser8) in plaques and E22P-Aβ in the postmortem cerebrospinal fluid (CSF) in the HIV cohort. DESIGN Clinicopathological study of HIV-infected adults. METHODS To assess frontal Aβ plaque deposition, we conducted immunohistochemistry for generic Aβ (4G8) and three modified Aβ forms. We determined CSF E22P-Aβ levels by ELISA. RESULTS We found 4G8-Aβ plaques in 29% of 279 HIV-infected cases. Within the age range of 31-70 years, the frequency of 4G8-Aβ plaque-bearing cases was higher in our HIV cohort (n = 273) compared with the general cohort (n = 1110) overall (29.3 vs. 25.8%) and across four age groups by decade (odds ratio 2.35, P < 0.0001). In HIV-infected cases with (n = 37) and without (n = 12) 4G8-Aβ plaques, modified Aβ forms occurred in order: N3pE, E22P, and phospho-Ser8. In CSF assays of HIV-infected cases with (n = 27; 17 focal, 10 widespread) and without (n = 11) 4G8-Aβ plaques, the median E22P-Aβ/Aβ40 ratio was higher among cases with widespread plaques than in cases with focal or absent plaques (P = 0.047). CONCLUSION Our findings suggest HIV-infected adults are at increased risk of developing cerebral Aβ plaques. The occurrence of modified Aβ forms in order suggests the progression stages of Aβ plaque deposition. The potential for E22P-Aβ as a CSF biomarker of cerebral Aβ plaques should be investigated.
Collapse
Affiliation(s)
- Anya Umlauf
- aDepartment of Psychiatry bDepartment of Neurosciences, School of Medicine, University of California San Diego, La Jolla cDepartment of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
155
|
When Does Alzheimer's Disease Really Start? The Role of Biomarkers. Int J Mol Sci 2019; 20:ijms20225536. [PMID: 31698826 PMCID: PMC6888399 DOI: 10.3390/ijms20225536] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/01/2019] [Accepted: 11/04/2019] [Indexed: 12/16/2022] Open
Abstract
While Alzheimer’s disease (AD) classical diagnostic criteria rely on clinical data from a stablished symptomatic disease, newer criteria aim to identify the disease in its earlier stages. For that, they incorporated the use of AD’s specific biomarkers to reach a diagnosis, including the identification of Aβ and tau depositions, glucose hypometabolism, and cerebral atrophy. These biomarkers created a new concept of the disease, in which AD’s main pathological processes have already taken place decades before we can clinically diagnose the first symptoms. Therefore, AD is now considered a dynamic disease with a gradual progression, and dementia is its final stage. With that in mind, new models were proposed, considering the orderly increment of biomarkers and the disease as a continuum, or the variable time needed for the disease’s progression. In 2011, the National Institute on Aging and the Alzheimer’s Association (NIA-AA) created separate diagnostic recommendations for each stage of the disease continuum—preclinical, mild cognitive impairment, and dementia. However, new scientific advances have led them to create a unifying research framework in 2018 that, although not intended for clinical use as of yet, is a step toward shifting the focus from the clinical symptoms to the biological alterations and toward changing the future diagnostic and treatment possibilities. This review aims to discuss the role of biomarkers in the onset of AD.
Collapse
|
156
|
Alavi A, Barrio JR, Werner TJ, Khosravi M, Newberg A, Høilund-Carlsen PF. Suboptimal validity of amyloid imaging-based diagnosis and management of Alzheimer’s disease: why it is time to abandon the approach. Eur J Nucl Med Mol Imaging 2019; 47:225-230. [DOI: 10.1007/s00259-019-04564-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
157
|
Lesman-Segev OH, La Joie R, Stephens ML, Sonni I, Tsai R, Bourakova V, Visani AV, Edwards L, O'Neil JP, Baker SL, Gardner RC, Janabi M, Chaudhary K, Perry DC, Kramer JH, Miller BL, Jagust WJ, Rabinovici GD. Tau PET and multimodal brain imaging in patients at risk for chronic traumatic encephalopathy. Neuroimage Clin 2019; 24:102025. [PMID: 31670152 PMCID: PMC6831941 DOI: 10.1016/j.nicl.2019.102025] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/03/2019] [Accepted: 09/27/2019] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To characterize individual and group-level neuroimaging findings in patients at risk for Chronic Traumatic Encephalopathy (CTE). METHODS Eleven male patients meeting criteria for Traumatic Encephalopathy Syndrome (TES, median age: 64) underwent neurologic evaluation, 3-Tesla MRI, and PET with [18F]-Flortaucipir (FTP, tau-PET) and [11C]-Pittsburgh compound B (PIB, amyloid-PET). Six patients underwent [18F]-Fluorodeoxyglucose-PET (FDG, glucose metabolism). We assessed imaging findings at the individual patient level, and in group-level comparisons with modality-specific groups of cognitively normal older adults (CN). Tau-PET findings in patients with TES were also compared to a matched group of patients with mild cognitive impairment or dementia due to Alzheimer's disease (AD). RESULTS All patients with TES sustained repetitive head injury participating in impact sports, ten in American football. Three patients met criteria for dementia and eight had mild cognitive impairment. Two patients were amyloid-PET positive and harbored the most severe MRI atrophy, FDG hypometabolism, and FTP-tau PET binding. Among the nine amyloid-negative patients, tau-PET showed either mildly elevated frontotemporal binding, a "dot-like" pattern, or no elevated binding. Medial temporal FTP was mildly elevated in a subset of amyloid-negative patients, but values were considerably lower than in AD. Voxelwise analyses revealed a convergence of imaging abnormalities (higher FTP binding, lower FDG, lower gray matter volumes) in frontotemporal areas in TES compared to controls. CONCLUSIONS Mildly elevated tau-PET binding was observed in a subset of amyloid-negative patients at risk for CTE, in a distribution consistent with CTE pathology stages III-IV. FTP-PET may be useful as a biomarker of tau pathology in CTE but is unlikely to be sensitive to early disease stages.
Collapse
Affiliation(s)
- Orit H Lesman-Segev
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, United States.
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, United States
| | - Melanie L Stephens
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, United States
| | - Ida Sonni
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Richard Tsai
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, United States
| | - Viktoriya Bourakova
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, United States
| | - Adrienne V Visani
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, United States
| | - Lauren Edwards
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, United States
| | - James P O'Neil
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Suzanne L Baker
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Raquel C Gardner
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, United States; San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121, United States
| | - Mustafa Janabi
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Kiran Chaudhary
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, United States
| | - David C Perry
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, United States
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, United States
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, United States
| | - William J Jagust
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, United States
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, United States; Departments of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, United States; Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, United States
| |
Collapse
|
158
|
Surkova EV, Tanashyan MM, Bespalov AI, Naminov AV. [Diabetes mellitus and cognitive impairment]. TERAPEVT ARKH 2019; 91:112-118. [PMID: 32598641 DOI: 10.26442/00403660.2019.10.000362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Indexed: 11/22/2022]
Abstract
The review discusses literature data and the results of our own studies on the effect of diabetes on cognitive functions and cerebrovascular pathology, as well as possible ptogenetic mechanisms for the implementation of this effect. The results of studies on the effects of antidiabetic drugs on cognitive function are presented.
Collapse
|
159
|
Weldon Furr J, Morales-Scheihing D, Manwani B, Lee J, McCullough LD. Cerebral Amyloid Angiopathy, Alzheimer's Disease and MicroRNA: miRNA as Diagnostic Biomarkers and Potential Therapeutic Targets. Neuromolecular Med 2019; 21:369-390. [PMID: 31586276 DOI: 10.1007/s12017-019-08568-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022]
Abstract
The protein molecules must fold into unique conformations to acquire functional activity. Misfolding, aggregation, and deposition of proteins in diverse organs, the so-called "protein misfolding disorders (PMDs)", represent the conformational diseases with highly ordered assemblies, including oligomers and fibrils that are linked to neurodegeneration in brain illnesses such as cerebral amyloid angiopathy (CAA) and Alzheimer's disease (AD). Recent studies have revealed several aspects of brain pathology in CAA and AD, but both the classification and underlying mechanisms need to be further refined. MicroRNAs (miRNAs) are critical regulators of gene expression at the post-transcriptional level. Increasing evidence with the advent of RNA sequencing technology suggests possible links between miRNAs and these neurodegenerative disorders. To provide insights on the small RNA-mediated regulatory circuitry and the translational significance of miRNAs in PMDs, this review will discuss the characteristics and mechanisms of the diseases and summarize circulating or tissue-resident miRNAs associated with AD and CAA.
Collapse
Affiliation(s)
- J Weldon Furr
- BRAINS Research Laboratory, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Diego Morales-Scheihing
- BRAINS Research Laboratory, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Bharti Manwani
- BRAINS Research Laboratory, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Juneyoung Lee
- BRAINS Research Laboratory, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Louise D McCullough
- BRAINS Research Laboratory, University of Texas McGovern Medical School, Houston, TX, 77030, USA.
| |
Collapse
|
160
|
Gomez-Isla T, Frosch MP. The Challenge of Defining Alzheimer Disease Based on Biomarkers in the Absence of Symptoms. JAMA Neurol 2019; 76:1143-1144. [PMID: 31305888 DOI: 10.1001/jamaneurol.2019.1667] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Teresa Gomez-Isla
- Department of Neurology, Massachusetts General Hospital, Boston
- Massachusetts Alzheimer's Disease Research Center, Boston
| | - Matthew P Frosch
- Massachusetts Alzheimer's Disease Research Center, Boston
- C. S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston
| |
Collapse
|
161
|
Zhen X, Chakraborty R, Vogt N, Bendlin BB, Singh V. Dilated Convolutional Neural Networks for Sequential Manifold-valued Data. PROCEEDINGS. IEEE INTERNATIONAL CONFERENCE ON COMPUTER VISION 2019; 2019:10620-10630. [PMID: 32405275 PMCID: PMC7220031 DOI: 10.1109/iccv.2019.01072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Efforts are underway to study ways via which the power of deep neural networks can be extended to non-standard data types such as structured data (e.g., graphs) or manifold-valued data (e.g., unit vectors or special matrices). Often, sizable empirical improvements are possible when the geometry of such data spaces are incorporated into the design of the model, architecture, and the algorithms. Motivated by neuroimaging applications, we study formulations where the data are sequential manifold-valued measurements. This case is common in brain imaging, where the samples correspond to symmetric positive definite matrices or orientation distribution functions. Instead of a recurrent model which poses computational/technical issues, and inspired by recent results showing the viability of dilated convolutional models for sequence prediction, we develop a dilated convolutional neural network architecture for this task. On the technical side, we show how the modules needed in our network can be derived while explicitly taking the Riemannian manifold structure into account. We show how the operations needed can leverage known results for calculating the weighted Fréchet Mean (wFM). Finally, we present scientific results for group difference analysis in Alzheimer's disease (AD) where the groups are derived using AD pathology load: here the model finds several brain fiber bundles that are related to AD even when the subjects are all still cognitively healthy.
Collapse
|
162
|
Musiek ES, Bhimasani M, Zangrilli MA, Morris JC, Holtzman DM, Ju YES. Circadian Rest-Activity Pattern Changes in Aging and Preclinical Alzheimer Disease. JAMA Neurol 2019; 75:582-590. [PMID: 29379963 DOI: 10.1001/jamaneurol.2017.4719] [Citation(s) in RCA: 306] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Importance Circadian rhythm disturbances occur in symptomatic Alzheimer disease (AD) and have been hypothesized to contribute to disease pathogenesis. However, it is unknown whether circadian changes occur during the presymptomatic phase of the disease. Objective To examine the associations between circadian function, aging, and preclinical AD pathology in cognitively normal adults. Design, Setting, and Participants This cross-sectional study was conducted using community volunteers from the Knight Alzheimer's Disease Research Center at Washington University in St Louis. Cognitively normal participants (n = 205) underwent 7 to 14 days of actigraphy in their home environment between 2010 and 2012, in addition to clinical assessment, amyloid imaging with Pittsburgh Compound B (PiB), and cerebrospinal fluid biomarker collection. Data collected from 3 years before to 6 months after actigraphy were included. Sixteen participants were excluded owing to incomplete data collection. Main Outcomes and Measures Circadian rhythm analysis was performed on actigraphy data using 3 methods: cosinor, nonparametric, and empirical mode decomposition. Preclinical AD was assessed by longitudinal clinical assessment, amyloid imaging with PiB, and cerebrospinal fluid biomarker collection. Results Data from 189 participants were included in the analyses. The mean (SD) age was 66.6 (8.3) years, and 121 participants (64%) were women. Older age (β = .247; P = .003) and male sex (β = .170; P = .04), in the absence of amyloid pathology, were associated with a significant increase in intradaily variability, a nonparametric measure of rest-activity rhythm fragmentation, as well as decreased amplitude by several measures. After correction for age and sex, the presence of preclinical amyloid plaque pathology, assessed by positive PiB imaging (mean [SD], 0.804 [0.187] for PiB negative vs 0.875 [0.178] for PiB positive; P = .05) or increasing cerebrospinal fluid phosphorylated-tau to amyloid β 42 ratio (β = .231; P = .008), was associated with increased intradaily variability, indicating rest-activity rhythm fragmentation. Conclusions and Relevance Preclinical AD is associated with rest-activity rhythm fragmentation, independent of age or sex. Aging was also associated with circadian dysfunction independently of preclinical AD pathology, particularly in men. The presence of circadian rhythm abnormalities in the preclinical phase of AD suggests that circadian dysfunction could contribute to early disease pathogenesis or serve as a biomarker of preclinical disease.
Collapse
Affiliation(s)
- Erik S Musiek
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, Missouri
| | - Meghana Bhimasani
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Margaret A Zangrilli
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, Missouri
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, Missouri
| | - Yo-El S Ju
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
163
|
Banning LCP, Ramakers IHGB, Deckers K, Verhey FRJ, Aalten P. Affective symptoms and AT(N) biomarkers in mild cognitive impairment and Alzheimer's disease: A systematic literature review. Neurosci Biobehav Rev 2019; 107:346-359. [PMID: 31525387 DOI: 10.1016/j.neubiorev.2019.09.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) biomarkers such as amyloid, p-tau and neuronal injury markers have been associated with affective symptoms in cognitively impaired individuals, but results are conflicting. METHODS CINAHL, Embase, PsycINFO and PubMed were searched for studies evaluating AD biomarkers with affective symptoms in mild cognitive impairment and AD dementia. Studies were classified according to AT(N) research criteria. RESULT Forty-five abstracts fulfilled eligibility criteria, including in total 8,293 patients (41 cross-sectional studies and 7 longitudinal studies). Depression and night-time behaviour disturbances were not related to AT(N) markers. Apathy was associated with A markers (PET, not CSF). Mixed findings were reported for the association between apathy and T(N) markers; anxiety and AT(N) markers; and between agitation and irritability and A markers. Agitation and irritability were not associated with T(N) markers. DISCUSSION Whereas some AD biomarkers showed to be associated with affective symptoms in AD, most evidence was inconsistent. This is likely due to differences in study design or heterogeneity in affective symptoms. Directions for future research are given.
Collapse
Affiliation(s)
- Leonie C P Banning
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, the Netherlands.
| | - Inez H G B Ramakers
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, the Netherlands.
| | - Kay Deckers
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, the Netherlands.
| | - Frans R J Verhey
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, the Netherlands.
| | - Pauline Aalten
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, the Netherlands.
| |
Collapse
|
164
|
Schreiber S, Wilisch-Neumann A, Schreiber F, Assmann A, Scheumann V, Perosa V, Jandke S, Mawrin C, Carare RO, Werring DJ. Invited Review: The spectrum of age-related small vessel diseases: potential overlap and interactions of amyloid and nonamyloid vasculopathies. Neuropathol Appl Neurobiol 2019; 46:219-239. [PMID: 31386773 DOI: 10.1111/nan.12576] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 07/10/2019] [Accepted: 07/11/2019] [Indexed: 12/12/2022]
Abstract
Deep perforator arteriopathy (DPA) and cerebral amyloid angiopathy (CAA) are the commonest known cerebral small vessel diseases (CSVD), which cause ischaemic stroke, intracebral haemorrhage (ICH) and vascular cognitive impairment (VCI). While thus far mainly considered as separate entities, we here propose that DPA and CAA share similarities, overlap and interact, so that 'pure' DPA or CAA are extremes along a continuum of age-related small vessel pathologies. We suggest blood-brain barrier (BBB) breakdown, endothelial damage and impaired perivascular β-amyloid (Aβ) drainage are hallmark common mechanisms connecting DPA and CAA. We also suggest a need for new biomarkers (e.g. high-resolution imaging) to deepen understanding of the complex relationships between DPA and CAA.
Collapse
Affiliation(s)
- S Schreiber
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany.,Center for behavioral brain sciences (CBBS), Magdeburg, Germany
| | - A Wilisch-Neumann
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - F Schreiber
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - A Assmann
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - V Scheumann
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - V Perosa
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - S Jandke
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - C Mawrin
- Department of Neuropathology, Otto-von-Guericke University, Magdeburg, Germany
| | - R O Carare
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - D J Werring
- Stroke Research Centre, Department of Brain Repair & Rehabilitation, UCL Institute of Neurology, The National Hospital for Neurology and Neurosurgery, London, UK
| |
Collapse
|
165
|
Matsuda H, Shigemoto Y, Sato N. Neuroimaging of Alzheimer's disease: focus on amyloid and tau PET. Jpn J Radiol 2019; 37:735-749. [PMID: 31493197 DOI: 10.1007/s11604-019-00867-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 08/28/2019] [Indexed: 12/14/2022]
Abstract
Although the diagnosis of dementia is still largely a clinical one, based on history and disease course, neuroimaging has dramatically increased our ability to accurately diagnose it. Neuroimaging modalities now play a wider role in dementia beyond their traditional role of excluding neurosurgical lesions and are recommended in most clinical guidelines for dementia. In addition, new neuroimaging methods facilitate the diagnosis of most neurodegenerative conditions after symptom onset and show diagnostic promise even in the very early or presymptomatic phases of some diseases. In the case of Alzheimer's disease (AD), extracellular amyloid-β (Aβ) aggregates and intracellular tau neurofibrillary tangles are the two neuropathological hallmarks of the disease. Recent molecular imaging techniques using amyloid and tau PET ligands have led to preclinical diagnosis and improved differential diagnosis as well as narrowed subject selection and treatment monitoring in clinical trials aimed at delaying or preventing the symptomatic phase of AD. This review discusses the recent progress in amyloid and tau PET imaging and the key findings achieved by the use of this molecular imaging modality related to the respective roles of Aβ and tau in AD, as well as its specific limitations.
Collapse
Affiliation(s)
- Hiroshi Matsuda
- Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo, 187-8551, Japan.
| | - Yoko Shigemoto
- Department of Radiology, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo, 187-8551, Japan
| | - Noriko Sato
- Department of Radiology, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo, 187-8551, Japan
| |
Collapse
|
166
|
Blautzik J, Kotz S, Brendel M, Sauerbeck J, Vettermann F, Winter Y, Bartenstein P, Ishii K, Rominger A. Relationship Between Body Mass Index, ApoE4 Status, and PET-Based Amyloid and Neurodegeneration Markers in Amyloid-Positive Subjects with Normal Cognition or Mild Cognitive Impairment. J Alzheimers Dis 2019; 65:781-791. [PMID: 28697560 DOI: 10.3233/jad-170064] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Body weight loss in late-life is known to occur at a very early stage of Alzheimer's disease (AD). Apolipoprotein E4 (ApoE4) represents a major genetic risk factor for AD and is linked to an increased cortical amyloid-β (Aβ) accumulation. Since the relationship between body weight, ApoE4, and AD pathology is poorly investigated, we aimed to evaluate whether ApoE4 allelic status modifies the association of body mass index (BMI) with markers of AD pathology. A total of 368 Aβ-positive cognitively healthy or mild cognitive impaired subjects had undergone [18F]-AV45-PET, [18F]-FDG-PET, and T1w-MRI examinations. Composite cortical [18F]-AV45 uptake and [18F]-FDG uptake in posterior cingulate cortex were calculated as surrogates of cortical Aβ load and glucose metabolism, respectively. Multiple linear regressions were performed to assess the relationships between these PET biomarkers with BMI, present cognitive performance, and cognitive changes over time. Multivariate analysis of covariance was conducted to test for statistical differences between ApoE4/BMI categories on the PET markers and cognitive scores. In carriers of the ApoE4 allele only, BMI was inversely associated with cortical amlyoid load (β= -0.193, p < 0.005) and recent cognitive decline (β= -0.209, p < 0.05), and positively associated with cortical glucose metabolism in an AD-vulnerable region (β= 0.145, p < 0.05). ApoE4/BMI category analyses demonstrated lower Aβ load, higher posterior cingulate glucose metabolism, improved cognitive performance, and lower progression of cognitive decline in obese ApoE4 carriers. The effect of ApoE4 in promoting the accumulation of cortical amyoid, which may itself be a driver for weight loss, may be moderated by altering leptin signaling in the hypothalamus.
Collapse
Affiliation(s)
- Janusch Blautzik
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sebastian Kotz
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Julia Sauerbeck
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Franziska Vettermann
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Yaroslav Winter
- Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Kazunari Ishii
- Department of Radiology, Kindai University Hospital, Osaka, Japan
| | - Axel Rominger
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | | |
Collapse
|
167
|
Abrahamson EE, Head E, Lott IT, Handen BL, Mufson EJ, Christian BT, Klunk WE, Ikonomovic MD. Neuropathological correlates of amyloid PET imaging in Down syndrome. Dev Neurobiol 2019; 79:750-766. [PMID: 31379087 DOI: 10.1002/dneu.22713] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 11/07/2022]
Abstract
Down syndrome (DS) results in an overproduction of amyloid-β (Aβ) peptide associated with early onset of Alzheimer's disease (AD). DS cases have Aβ deposits detectable histologically as young as 12-30 years of age, primarily in the form of diffuse plaques, the type of early amyloid pathology also seen at pre-clinical (i.e., pathological aging) and prodromal stages of sporadic late onset AD. In DS subjects aged >40 years, levels of cortical Aβ deposition are similar to those observed in late onset AD and in addition to diffuse plaques involve cored plaques associated with dystrophic neurites (neuritic plaques), which are of neuropathological diagnostic significance in AD. The purpose of this review is to summarize and discuss findings from amyloid PET imaging studies of DS in reference to postmortem amyloid-based neuropathology. PET neuroimaging applied to subjects with DS has the potential to (a) track the natural progression of brain pathology, including the earliest stages of amyloid accumulation, and (b) determine whether amyloid PET biomarkers predict the onset of dementia. In addition, the question that is still incompletely understood and relevant to both applications is the ability of amyloid PET to detect Aβ deposits in their earliest form.
Collapse
Affiliation(s)
- Eric E Abrahamson
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania.,Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, UC Irvine School of Medicine, Orange, California
| | - Ira T Lott
- Department of Neurology, UC Irvine School of Medicine, Orange, California
| | - Benjamin L Handen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Elliott J Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona
| | - Bradley T Christian
- Departments of Medical Physics and Psychiatry, Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - William E Klunk
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Milos D Ikonomovic
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania.,Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
168
|
Piri R, Naghavi-Behzad M, Gerke O, Høilund-Carlsen PF, Vafaee MS. Investigations of possible links between Alzheimer’s disease and type 2 diabetes mellitus by positron emission tomography: a systematic review. Clin Transl Imaging 2019. [DOI: 10.1007/s40336-019-00339-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
169
|
Li J, Jin D, Li A, Liu B, Song C, Wang P, Wang D, Xu K, Yang H, Yao H, Zhou B, Bejanin A, Chetelat G, Han T, Lu J, Wang Q, Yu C, Zhang X, Zhou Y, Zhang X, Jiang T, Liu Y, Han Y. ASAF: altered spontaneous activity fingerprinting in Alzheimer's disease based on multisite fMRI. Sci Bull (Beijing) 2019; 64:998-1010. [PMID: 36659811 DOI: 10.1016/j.scib.2019.04.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/22/2019] [Accepted: 03/25/2019] [Indexed: 01/21/2023]
Abstract
Several monocentric studies have noted alterations in spontaneous brain activity in Alzheimer's disease (AD), although there is no consensus on the altered amplitude of low-frequency fluctuations in AD patients. The main aim of the present study was to identify a reliable and reproducible abnormal brain activity pattern in AD. The amplitude of local brain activity (AM), which can provide fast mapping of spontaneous brain activity across the whole brain, was evaluated based on multisite rs-fMRI data for 688 subjects (215 normal controls (NCs), 221 amnestic mild cognitive impairment (aMCI) 252 AD). Two-sample t-tests were used to detect group differences between AD patients and NCs from the same site. Differences in the AM maps were statistically analyzed via the Stouffer's meta-analysis. Consistent regions of lower spontaneous brain activity in the default mode network and increased activity in the bilateral hippocampus/parahippocampus, thalamus, caudate nucleus, orbital part of the middle frontal gyrus and left fusiform were observed in the AD patients compared with those in NCs. Significant correlations (P < 0.05, Bonferroni corrected) between the normalized amplitude index and Mini-Mental State Examination scores were found in the identified brain regions, which indicates that the altered brain activity was associated with cognitive decline in the patients. Multivariate analysis and leave-one-site-out cross-validation led to a 78.49% prediction accuracy for single-patient classification. The altered activity patterns of the identified brain regions were largely correlated with the FDG-PET results from another independent study. These results emphasized the impaired brain activity to provide a robust and reproducible imaging signature of AD.
Collapse
Affiliation(s)
- Jiachen Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Dan Jin
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ang Li
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bing Liu
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China; Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Chengyuan Song
- Department of Neurology, Qilu Hospital of Shandong University, Ji'nan 250012, China
| | - Pan Wang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin 300350, China; Institute of Geriatrics and Gerontology, Chinese PLA General Hospital, Beijing 100853, China
| | - Dawei Wang
- Department of Radiology, Qilu Hospital, Ji'nan 250012, China
| | - Kaibin Xu
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Hongwei Yang
- Department of Radiology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Hongxiang Yao
- Department of Radiology, Chinese PLA General Hospital, Beijing 100853, China
| | - Bo Zhou
- Institute of Geriatrics and Gerontology, Chinese PLA General Hospital, Beijing 100853, China
| | - Alexandre Bejanin
- Université Normandie, Inserm, Université de Caen-Normandie, Inserm UMR-S U1237, GIP Cyceron, Caen 14000, France
| | - Gael Chetelat
- Université Normandie, Inserm, Université de Caen-Normandie, Inserm UMR-S U1237, GIP Cyceron, Caen 14000, France
| | - Tong Han
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Jie Lu
- Department of Radiology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Qing Wang
- Department of Radiology, Qilu Hospital, Ji'nan 250012, China
| | - Chunshui Yu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xinqing Zhang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yuying Zhou
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Xi Zhang
- Institute of Geriatrics and Gerontology, Chinese PLA General Hospital, Beijing 100853, China
| | - Tianzi Jiang
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China; Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yong Liu
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China; Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing 100053, China; Beijing Institute of Geriatrics, Beijing 100053, China; National Clinical Research Center for Geriatric Disorders, Beijing 100053, China.
| |
Collapse
|
170
|
Shirvan J, Clement N, Ye R, Katz S, Schultz A, Johnson KA, Gomez-Isla T, Frosch M, Growdon JH, Gomperts SN. Neuropathologic correlates of amyloid and dopamine transporter imaging in Lewy body disease. Neurology 2019; 93:e476-e484. [PMID: 31243072 DOI: 10.1212/wnl.0000000000007855] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 03/14/2019] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To develop imaging biomarkers of diseases in the Lewy body spectrum and to validate these markers against postmortem neuropathologic findings. METHODS Four cognitively normal participants with Parkinson disease (PD), 4 with PD with cognitive impairments, and 10 with dementia with Lewy bodies underwent amyloid imaging with [11C]Pittsburgh compound B (PiB) and dopamine transporter (DAT) imaging with [11C]Altropane. All 18 had annual neurologic examinations. All cognitively normal participants with PD developed cognitive impairment before death. Neuropathologic examinations assessed and scored Braak Lewy bodies, Thal distribution of amyloid, Consortium to Establish a Registry for Alzheimer's Disease neuritic amyloid plaques, Braak neurofibrillary tangles, and cerebral amyloid angiopathy, as well as total amyloid plaque burden in the superior frontal, superior parietal, occipital, and inferior temporal cortical regions. PET data were expressed as the standardized uptake value ratio with cerebellar reference. Analyses accounted for the interval between imaging and autopsy. RESULTS All 18 patients met neuropathologic criteria for Lewy body disease; the DAT concentration was low in each case. All patients with elevated [11C]PiB retention measured in a neocortical aggregate had β-amyloid deposits at autopsy. [11C]PiB retention significantly correlated with neuritic plaque burden and with total plaque burden. [11C]PiB retention also significantly correlated with the severity of both Braak stages of neurofibrillary tangle and Lewy body scores. Neuritic plaque burden was significantly associated with neurofibrillary tangle pathology. CONCLUSION Antemortem [11C]Altropane PET is a sensitive measure of substantia nigra degeneration. [11C]PiB scans accurately reflect cortical amyloid deposits seen at autopsy. These findings support the use of molecular imaging in the evaluation of patients with Lewy body diseases.
Collapse
Affiliation(s)
- Julia Shirvan
- From the Departments of Neurology (J.S., R.Y., K.A.J., T.G.-I., J.H.G., S.N.G.), Pathology (N.C., M.F.), and Radiology (S.K., A.S., K.A.J.), Massachusetts General Hospital, Boston
| | - Nathan Clement
- From the Departments of Neurology (J.S., R.Y., K.A.J., T.G.-I., J.H.G., S.N.G.), Pathology (N.C., M.F.), and Radiology (S.K., A.S., K.A.J.), Massachusetts General Hospital, Boston
| | - Rong Ye
- From the Departments of Neurology (J.S., R.Y., K.A.J., T.G.-I., J.H.G., S.N.G.), Pathology (N.C., M.F.), and Radiology (S.K., A.S., K.A.J.), Massachusetts General Hospital, Boston
| | - Samantha Katz
- From the Departments of Neurology (J.S., R.Y., K.A.J., T.G.-I., J.H.G., S.N.G.), Pathology (N.C., M.F.), and Radiology (S.K., A.S., K.A.J.), Massachusetts General Hospital, Boston
| | - Aaron Schultz
- From the Departments of Neurology (J.S., R.Y., K.A.J., T.G.-I., J.H.G., S.N.G.), Pathology (N.C., M.F.), and Radiology (S.K., A.S., K.A.J.), Massachusetts General Hospital, Boston
| | - Keith A Johnson
- From the Departments of Neurology (J.S., R.Y., K.A.J., T.G.-I., J.H.G., S.N.G.), Pathology (N.C., M.F.), and Radiology (S.K., A.S., K.A.J.), Massachusetts General Hospital, Boston
| | - Teresa Gomez-Isla
- From the Departments of Neurology (J.S., R.Y., K.A.J., T.G.-I., J.H.G., S.N.G.), Pathology (N.C., M.F.), and Radiology (S.K., A.S., K.A.J.), Massachusetts General Hospital, Boston
| | - Matthew Frosch
- From the Departments of Neurology (J.S., R.Y., K.A.J., T.G.-I., J.H.G., S.N.G.), Pathology (N.C., M.F.), and Radiology (S.K., A.S., K.A.J.), Massachusetts General Hospital, Boston
| | - John H Growdon
- From the Departments of Neurology (J.S., R.Y., K.A.J., T.G.-I., J.H.G., S.N.G.), Pathology (N.C., M.F.), and Radiology (S.K., A.S., K.A.J.), Massachusetts General Hospital, Boston
| | - Stephen N Gomperts
- From the Departments of Neurology (J.S., R.Y., K.A.J., T.G.-I., J.H.G., S.N.G.), Pathology (N.C., M.F.), and Radiology (S.K., A.S., K.A.J.), Massachusetts General Hospital, Boston.
| |
Collapse
|
171
|
Shea YF, Barker W, Greig-Gusto MT, Loewenstein DA, Duara R, DeKosky ST. Impact of Amyloid PET Imaging in the Memory Clinic: A Systematic Review and Meta-Analysis. J Alzheimers Dis 2019; 64:323-335. [PMID: 29889075 DOI: 10.3233/jad-180239] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Patients with cognitive impairment or dementias of uncertain etiology are frequently referred to a memory disorders specialty clinic. The impact of and role for amyloid PET imaging (Aβ-PET) may be most appropriate in this clinical setting. OBJECTIVE The primary objective of this study was to perform a systematic review and meta-analysis of the impact of Aβ-PET on etiological diagnosis and clinical management in the memory clinic setting. METHODS A search of the literature on the impact of Aβ-PET in the memory clinic setting between 1 January 2004 and 12 February 2018 was conducted. Meta-analysis using a random effects model was performed to determine the pooled estimate of the impact of Aβ-PET in the changes of diagnoses and changes in management plan. RESULTS After rigorous review, results from 13 studies were extracted, involving 1,489 patients. Meta-analysis revealed a pooled effect of change in diagnoses of 35.2% (95% CI 24.6-47.5). Sub-analyses showed that the pooled effect in change in diagnoses if Aβ-PET was used under the appropriate use criteria (AUC) or non-AUC criteria were 47.8% (95% CI 25.9-70.5) and 29.6% (95% CI: 21.5-39.3), respectively. The pooled effect of a change of diagnosis from Alzheimer's disease (AD) to non-AD and from non-AD to AD were 22.7% (95% CI: 17.1-29.5) and 25.6% (95% CI: 17.6-35.8), respectively. The pooled effect leading to a change of management was 59.6% (95% CI 39.4-77.0). CONCLUSIONS Aβ-PET has a highly significant impact on both changes in diagnosis and management among patients being seen at a specialty memory clinic.
Collapse
Affiliation(s)
- Yat-Fung Shea
- Wien Center for Alzheimer's Disease and Memory Disorder, Mount Sinai Medical Center, Miami Beach, FL, USA.,Department of Medicine, LKS Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Warren Barker
- Wien Center for Alzheimer's Disease and Memory Disorder, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Maria T Greig-Gusto
- Wien Center for Alzheimer's Disease and Memory Disorder, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - David A Loewenstein
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, FL, USA
| | - Ranjan Duara
- Wien Center for Alzheimer's Disease and Memory Disorder, Mount Sinai Medical Center, Miami Beach, FL, USA.,Departments of Neurology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL and University of Florida College of Medicine, Gainesville, FL, USA
| | - Steven T DeKosky
- Department of Neurology, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
172
|
Mormino EC, Papp KV. Amyloid Accumulation and Cognitive Decline in Clinically Normal Older Individuals: Implications for Aging and Early Alzheimer's Disease. J Alzheimers Dis 2019; 64:S633-S646. [PMID: 29782318 DOI: 10.3233/jad-179928] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The aberrant accumulation of the amyloid protein is a critical and early event in the Alzheimer's disease (AD) cascade. Given the early involvement of this pathological process, it is not surprising that many clinically normal (CN) older individuals demonstrate evidence of abnormal Aβ at postmortem examination and in vivo using either CSF or PET imaging. Converging evidence across multiple research groups suggests that the presence of abnormal Aβ among CN individuals is associated with elevated risk of future clinical impairment and cognitive decline. Amyloid positivity in conjunction with biomarkers of neuronal injury offers further insight into which CN are most at risk for short-term decline. Although in its infancy, tau PET has demonstrated early increases among Aβ+ that will likely be an important indicator of risk among CN. Overall, the detection of early Aβ among CN individuals has provided an important opportunity to understand the contributions of this pathology to age-related cognitive decline and to explore early intervention with disease modifying strategies.
Collapse
Affiliation(s)
- Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Kathryn V Papp
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
173
|
Meyer PF, McSweeney M, Gonneaud J, Villeneuve S. AD molecular: PET amyloid imaging across the Alzheimer's disease spectrum: From disease mechanisms to prevention. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 165:63-106. [PMID: 31481172 DOI: 10.1016/bs.pmbts.2019.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The advent of amyloid-beta (Aβ) positron emission tomography (PET) imaging has transformed the field of Alzheimer's disease (AD) by enabling the quantification of cortical Aβ accumulation and propagation in vivo. This revolutionary tool has made it possible to measure direct associations between Aβ and other AD biomarkers, to identify factors that influence Aβ accumulation and to redefine entry criteria into clinical trials as well as measure drug target engagement. This chapter summarizes the main findings on the associations of Aβ with other biomarkers of disease progression across the AD spectrum. It discusses investigations of the timing at which Aβ pathology starts to accumulate, demonstrates the clinical utility of Aβ PET imaging and discusses some ethical implications. Finally, it presents genetic and potentially modifiable lifestyle factors that might influence Aβ accumulation and therefore be targets for AD prevention.
Collapse
Affiliation(s)
- Pierre-François Meyer
- Centre for Studies on the Prevention of Alzheimer's Disease, Douglas Mental Health University Institute, Montréal, Canada; McGill University, Montréal, Canada
| | - Melissa McSweeney
- Centre for Studies on the Prevention of Alzheimer's Disease, Douglas Mental Health University Institute, Montréal, Canada; McGill University, Montréal, Canada
| | - Julie Gonneaud
- Centre for Studies on the Prevention of Alzheimer's Disease, Douglas Mental Health University Institute, Montréal, Canada; McGill University, Montréal, Canada
| | - Sylvia Villeneuve
- Centre for Studies on the Prevention of Alzheimer's Disease, Douglas Mental Health University Institute, Montréal, Canada; McGill University, Montréal, Canada.
| |
Collapse
|
174
|
Frederiksen KS, Madsen K, Andersen BB, Beyer N, Garde E, Høgh P, Waldemar G, Hasselbalch SG, Law I. Moderate- to high-intensity exercise does not modify cortical β-amyloid in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2019; 5:208-215. [PMID: 31198839 PMCID: PMC6556817 DOI: 10.1016/j.trci.2019.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Animal models of Alzheimer's disease show that exercise may modify β-amyloid (Aβ) deposition. We examined the effect of a 16-week exercise intervention on cortical Aβ in patients with mild-to-moderate Alzheimer's disease. METHODS Thirty-six patients with Alzheimer's disease were randomized to either one hour of aerobic exercise three times weekly for 16 weeks or usual care. Pre and post intervention, 11Carbon-Pittsburgh compound B positron emission tomography was carried out to assess cortical Aβ, and quantified using standardized uptake value rations (SUVRs). RESULTS The intervention showed no effect on follow-up SUVRs in a covariance analysis with group allocation, baseline intervention SUVR, age, sex, and baseline Mini-Mental State Examination as predictors. Change in SUVRs did not correlate with changes in measures of physical or aerobic fitness. DISCUSSION The present findings do not support an effect of exercise on Aβ. However, the relatively short intervention period may account for a lack of efficacy. Further studies should test earlier and longer interventions.
Collapse
Affiliation(s)
- Kristian S. Frederiksen
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Karine Madsen
- Neurobiology Research Unit, Copenhagen, Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte B. Andersen
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Nina Beyer
- Musculoskeletal Rehabilitation Research Unit and Institute of Sports Medicine, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ellen Garde
- Danish Research Centre for Magnetic Resonance, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Department of Public Health and Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Peter Høgh
- Zealand University Hospital, Department of Neurology, Roskilde, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Steen G. Hasselbalch
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
175
|
NIA-AA Research Framework: Toward a biological definition of Alzheimer's disease. Alzheimers Dement 2019; 14:535-562. [PMID: 29653606 PMCID: PMC5958625 DOI: 10.1016/j.jalz.2018.02.018] [Citation(s) in RCA: 6455] [Impact Index Per Article: 1075.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/21/2018] [Accepted: 02/27/2018] [Indexed: 02/06/2023]
Abstract
In 2011, the National Institute on Aging and Alzheimer’s Association created separate diagnostic recommendations for the preclinical, mild cognitive impairment, and dementia stages of Alzheimer’s disease. Scientific progress in the interim led to an initiative by the National Institute on Aging and Alzheimer’s Association to update and unify the 2011 guidelines. This unifying update is labeled a “research framework” because its intended use is for observational and interventional research, not routine clinical care. In the National Institute on Aging and Alzheimer’s Association Research Framework, Alzheimer’s disease (AD) is defined by its underlying pathologic processes that can be documented by postmortem examination or in vivo by biomarkers. The diagnosis is not based on the clinical consequences of the disease (i.e., symptoms/signs) in this research framework, which shifts the definition of AD in living people from a syndromal to a biological construct. The research framework focuses on the diagnosis of AD with biomarkers in living persons. Biomarkers are grouped into those of β amyloid deposition, pathologic tau, and neurodegeneration [AT(N)]. This ATN classification system groups different biomarkers (imaging and biofluids) by the pathologic process each measures. The AT(N) system is flexible in that new biomarkers can be added to the three existing AT(N) groups, and new biomarker groups beyond AT(N) can be added when they become available. We focus on AD as a continuum, and cognitive staging may be accomplished using continuous measures. However, we also outline two different categorical cognitive schemes for staging the severity of cognitive impairment: a scheme using three traditional syndromal categories and a six-stage numeric scheme. It is important to stress that this framework seeks to create a common language with which investigators can generate and test hypotheses about the interactions among different pathologic processes (denoted by biomarkers) and cognitive symptoms. We appreciate the concern that this biomarker-based research framework has the potential to be misused. Therefore, we emphasize, first, it is premature and inappropriate to use this research framework in general medical practice. Second, this research framework should not be used to restrict alternative approaches to hypothesis testing that do not use biomarkers. There will be situations where biomarkers are not available or requiring them would be counterproductive to the specific research goals (discussed in more detail later in the document). Thus, biomarker-based research should not be considered a template for all research into age-related cognitive impairment and dementia; rather, it should be applied when it is fit for the purpose of the specific research goals of a study. Importantly, this framework should be examined in diverse populations. Although it is possible that β-amyloid plaques and neurofibrillary tau deposits are not causal in AD pathogenesis, it is these abnormal protein deposits that define AD as a unique neurodegenerative disease among different disorders that can lead to dementia. We envision that defining AD as a biological construct will enable a more accurate characterization and understanding of the sequence of events that lead to cognitive impairment that is associated with AD, as well as the multifactorial etiology of dementia. This approach also will enable a more precise approach to interventional trials where specific pathways can be targeted in the disease process and in the appropriate people.
Collapse
|
176
|
Bischof GN, Jacobs HIL. Subthreshold amyloid and its biological and clinical meaning: Long way ahead. Neurology 2019; 93:72-79. [PMID: 31167933 DOI: 10.1212/wnl.0000000000007747] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 03/26/2019] [Indexed: 01/22/2023] Open
Abstract
The development of in vivo imaging of the pathologic hallmark of Alzheimer disease (AD), β-amyloid (Aβ), altered the framing of its pathophysiology and formulation of inclusion criteria for clinical trials. Recent evidence suggests that in vivo measures of Aβ deposition below a threshold indicative of Aβ positivity carry critical information on future cognitive decline and accumulation of AD pathology, potentially already at a younger age. Here, we integrate the existing literature on histopathology of Aβ and its convergence and divergence with in vivo Aβ imaging. The evidence presented amounts to a reconceptualization, in which we advocate for a closer look into Aβ accumulation rates in earlier life, the factors that promote accumulation, comparative studies with different markers of Aβ, and longitudinal designs to elucidate when AD pathology rises and how it shifts from benign to malignant stages that ultimately define AD. These efforts open a new window of opportunity for disease-modifying interventions.
Collapse
Affiliation(s)
- Gérard N Bischof
- From the Multimodal Imaging Group (G.N.B.), Department of Nuclear Medicine, University Hospital Cologne, Germany; Cognitive Neuroscience (H.I.L.J.), Faculty of Psychology and Neuroscience, and School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Alzheimer Centre Limburg, Maastricht University, the Netherlands; and Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Harvard Medical School, Massachusetts General Hospital, Boston.
| | - Heidi I L Jacobs
- From the Multimodal Imaging Group (G.N.B.), Department of Nuclear Medicine, University Hospital Cologne, Germany; Cognitive Neuroscience (H.I.L.J.), Faculty of Psychology and Neuroscience, and School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Alzheimer Centre Limburg, Maastricht University, the Netherlands; and Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Harvard Medical School, Massachusetts General Hospital, Boston
| |
Collapse
|
177
|
Lowe VJ, Lundt ES, Albertson SM, Przybelski SA, Senjem ML, Parisi JE, Kantarci K, Boeve B, Jones DT, Knopman D, Jack CR, Dickson DW, Petersen RC, Murray ME. Neuroimaging correlates with neuropathologic schemes in neurodegenerative disease. Alzheimers Dement 2019; 15:927-939. [PMID: 31175025 DOI: 10.1016/j.jalz.2019.03.016] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 02/05/2019] [Accepted: 03/07/2019] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Neuroimaging biomarkers are important for early diagnosis of Alzheimer's disease, and comparing multimodality neuroimaging to autopsy data is essential. METHODS We compared the pathologic findings from a prospective autopsy cohort (n = 100) to Pittsburgh compound B PET (PiB-PET), 18F-fluorodeoxyglucose PET (FDG-PET), and MRI. Correlations between neuroimaging biomarkers and neuropathologic schemes were assessed. RESULTS PiB-PET showed strong correlations with Thal amyloid phase and Consortium to Establish a Registry for Alzheimer's Disease score and categorized 44% of Thal phase 1 participants as positive. FDG-PET and MRI correlated modestly with Braak tangle stage in Alzheimer's type pathology. A subset of participants with "none" or "sparse" neuritic plaque scores had elevated PiB-PET signal due to diffuse amyloid plaque. Participants with findings characterized as "suspected non-Alzheimer's pathophysiology" represented 15% of the group. DISCUSSION PiB-PET is associated with Alzheimer's disease, neuritic plaques, and diffuse plaques. FDG-PET and MRI have modest correlation with neuropathologic schemes. Participants with findings characterized as suspected non-Alzheimer's pathophysiology most commonly had primary age-related tauopathy.
Collapse
Affiliation(s)
- Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA.
| | - Emily S Lundt
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | | | - Matthew L Senjem
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - Joseph E Parisi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Bradley Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - David Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | |
Collapse
|
178
|
Doré V, Bullich S, Rowe CC, Bourgeat P, Konate S, Sabri O, Stephens AW, Barthel H, Fripp J, Masters CL, Dinkelborg L, Salvado O, Villemagne VL, De Santi S. Comparison of 18F-florbetaben quantification results using the standard Centiloid, MR-based, and MR-less CapAIBL ® approaches: Validation against histopathology. Alzheimers Dement 2019; 15:807-816. [PMID: 31101517 DOI: 10.1016/j.jalz.2019.02.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 02/15/2019] [Accepted: 02/25/2019] [Indexed: 01/22/2023]
Abstract
INTRODUCTION 18F-florbetaben is currently approved for the visual rule out of β-amyloid (Aβ) pathology. It is also used for recruitment and as an outcome measure in therapeutic trials, requiring accurate and reproducible quantification of Aβ burden in the brain. METHODS Data from eighty-eight subjects (52 male subjects, aged 79.8 ± 10.6 years) who underwent antemortem 18F-florbetaben positron emission tomography scan and magnetic resonance imaging less than a year before neuropathological assessment at autopsy were evaluated. Image analysis was performed using the standard Centiloid (CL) statistical parametric mapping approach and CapAIBL®. Imaging results were compared against autopsy data. RESULTS Against combined Bielschowsky silver staining and immunohistochemistry histopathological scores, statistical parametric mapping had 96% sensitivity, 96% specificity, and 95% accuracy, whereas magnetic resonance-less CapAIBL standardized uptake value ratioWhole Cerebellum had 94% sensitivity, 96% specificity, and 95% accuracy. Based on the combined histopathological scores, a CL threshold band of 19 ± 7 CL was determined. DISCUSSION Quantification of 18F-florbetaben positron emission tomography scans using magnetic resonance-based and magnetic resonance-less CapAIBL® approaches showed high agreement, establishing a pathology-based threshold in CL.
Collapse
Affiliation(s)
- Vincent Doré
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia; CSIRO Health and Biosecurity Flagship: The Australian e-Health Research Centre, Brisbane, QLD, Australia.
| | | | - Christopher C Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia; Department of Medicine, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Pierrick Bourgeat
- CSIRO Health and Biosecurity Flagship: The Australian e-Health Research Centre, Brisbane, QLD, Australia
| | - Salamata Konate
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia; CSIRO Health and Biosecurity Flagship: The Australian e-Health Research Centre, Brisbane, QLD, Australia
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | | | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Jurgen Fripp
- CSIRO Health and Biosecurity Flagship: The Australian e-Health Research Centre, Brisbane, QLD, Australia
| | - Colin L Masters
- The Florey Institute of Neurosciences and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | | | - Olivier Salvado
- CSIRO Health and Biosecurity Flagship: The Australian e-Health Research Centre, Brisbane, QLD, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia; Department of Medicine, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| | | |
Collapse
|
179
|
Sheikh-Bahaei N, Manavaki R, Sajjadi SA, Priest AN, O’Brien JT, Gillard JH. Correlation of Lobar Cerebral Microbleeds with Amyloid, Perfusion, and Metabolism in Alzheimer’s Disease. J Alzheimers Dis 2019; 68:1489-1497. [DOI: 10.3233/jad-180443] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Nasim Sheikh-Bahaei
- Department of Radiology, Keck School of Medicine of USC, University of Southern California, USA
| | - Roido Manavaki
- Department of Radiology, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - S. Ahmad Sajjadi
- Department of Neurology, University of California Irvine, CA, USA
| | - Andrew N. Priest
- Department of Radiology, Cambridge University Hospitals, Cambridge, UK
| | - John T. O’Brien
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Jonathan H. Gillard
- Department of Radiology, University of Cambridge School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
180
|
Clinical significance of visually equivocal amyloid PET findings from the Alzheimer's Disease Neuroimaging Initiative cohort. Neuroreport 2019; 29:553-558. [PMID: 29438267 DOI: 10.1097/wnr.0000000000000986] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
To evaluate the clinical and imaging characteristics of patients with visually equivocal amyloid PET images, patients from the Alzheimer's Disease Neuroimaging Initiative cohort who had fluorine-18-florbetapir PET scans both at baseline and 24 months were selected. Five nuclear medicine physicians visually assessed the PET images and classified them as either positive or negative. Images not reaching a majority agreement were classified as equivocal. Among a total of 379 patients, the number of patients in each fluorine-18-florbetapir PET negative/equivocal/positive categories was 218 (57.5%), 32 (8.4%), and 129 (34.0%). Eight to 9% of patients with normal cognition (N=12/141), mild cognitive impairment (N=20/214), and no Alzheimer's disease (N=0/24) showed equivocal PET finding for each. In negative/equivocal/positive groups, positive cerebrospinal fluid Aβ1-42 was observed in 25.7, 81.5, and 98.3%, respectively. Baseline standardized uptake value ratios of fluorine-18-florbetapir PET were 0.75±0.05, 0.86±0.09, and 1.01±0.09, respectively [F(2, 376)=603.547; P<0.001]. After 24 months of follow-up, the standardized uptake value ratios increased by 0.81±2.62, 2.81±2.90, and 2.17±3.66%, respectively [F(2, 376)=7.905, P<0.05 vs. the negative group]. Among mild cognitive impairment patients, the equivocal group showed a more rapid decline in glucose metabolism than the negative group [5.52±5.36 vs. 0.67±4.45; F(2, 122)=9.028, P<0.01]. 8.4% of the patients in this study showed a visually equivocal result of amyloid PET. These patients showed a moderate amount of amyloid accumulation and a rapid rate of accumulation.
Collapse
|
181
|
Yang J, Cheng R, Fu H, Yang J, Kumar M, Lu J, Xu Y, Liang SH, Cui M, Ran C. Half-curcumin analogues as PET imaging probes for amyloid beta species. Chem Commun (Camb) 2019; 55:3630-3633. [PMID: 30849141 DOI: 10.1039/c8cc10166c] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this report, we demonstrate that half-curcuminoid could be a better scaffold for PET tracer development. F-CRANAD-101 was designed and found to show significant response to both soluble and insoluble Aβs in the fluorescent spectral tests. PET imaging results indicated that 14 month and 5 month old APP/PS1 AD mice had higher signals in the brain than age-matched wild type mice.
Collapse
Affiliation(s)
- Jian Yang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Duara R, Loewenstein DA, Lizarraga G, Adjouadi M, Barker WW, Greig-Custo MT, Rosselli M, Penate A, Shea YF, Behar R, Ollarves A, Robayo C, Hanson K, Marsiske M, Burke S, Ertekin-Taner N, Vaillancourt D, De Santi S, Golde T, St D. Effect of age, ethnicity, sex, cognitive status and APOE genotype on amyloid load and the threshold for amyloid positivity. NEUROIMAGE-CLINICAL 2019; 22:101800. [PMID: 30991618 PMCID: PMC6447735 DOI: 10.1016/j.nicl.2019.101800] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 02/08/2019] [Accepted: 03/26/2019] [Indexed: 11/30/2022]
Abstract
The threshold for amyloid positivity by visual assessment on PET has been validated by comparison to amyloid load measured histopathologically and biochemically at post mortem. As such, it is now feasible to use qualitative visual assessment of amyloid positivity as an in-vivo gold standard to determine those factors which can modify the quantitative threshold for amyloid positivity. We calculated quantitative amyloid load, measured as Standardized Uptake Value Ratios (SUVRs) using [18-F]florbetaben PET scans, for 159 Hispanic and non-Hispanic participants, who had been classified clinically as Cognitively Normal (CN), Mild Cognitive Impairment (MCI) or Dementia (DEM). PET scans were visually rated as amyloid positive (A+) or negative (A-), and these judgments were used as the gold standard with which to determine (using ROC analyses) the SUVR threshold for amyloid positivity considering factors such as age, ethnicity (Hispanic versus non-Hispanic), gender, cognitive status, and apolipoprotein E ε4 carrier status. Visually rated scans were A+ for 11% of CN, 39.0% of MCI and 70% of DEM participants. The optimal SUVR threshold for A+ among all participants was 1.42 (sensitivity = 94%; specificity = 92.5%), but this quantitative threshold was higher among E4 carriers (SUVR = 1.52) than non-carriers (SUVR = 1.31). While mean SUVRs did not differ between Hispanic and non-Hispanic participants;, a statistically significant interaction term indicated that the effect of E4 carrier status on amyloid load was greater among non-Hispanics than Hispanics. Visual assessment, as the gold standard for A+, facilitates determination of the effects of various factors on quantitative thresholds for amyloid positivity. A continuous relationship was found between amyloid load and global cognitive scores, suggesting that any calculated threshold for the whole group, or a subgroup, is artefactual and that the lowest calculated threshold may be optimal for the purposes of early diagnosis and intervention. Demographic factors did not affect the threshold for amyloid positivity. Cognitive status did not affect this threshold for amyloid positivity. APOE4 carriers had a higher threshold for amyloid positivity than non-carriers. Among APOE4 carriers, non-Hispanics had higher amyloid load than non- Hispanics. There was a continuous relationship between amyloid load and cognitive status.
Collapse
Affiliation(s)
- R Duara
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA; College of Engineering and Computing, Florida International University, Miami, FL, USA; University of Florida College of Medicine, Gainesville, FL, USA.
| | - D A Loewenstein
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA; Miller School of Medicine, University of Miami, Miami, FL, USA
| | - G Lizarraga
- Florida ADRC, USA; College of Engineering and Computing, Florida International University, Miami, FL, USA
| | - M Adjouadi
- Florida ADRC, USA; College of Engineering and Computing, Florida International University, Miami, FL, USA
| | - W W Barker
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA
| | - M T Greig-Custo
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA
| | - M Rosselli
- Florida ADRC, USA; Florida Atlantic University, USA
| | - A Penate
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA
| | - Y F Shea
- Mount Sinai Medical Center, Miami Beach, USA; Department of Medicine, University of Hong Kong, Hong Kong
| | - R Behar
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA
| | - A Ollarves
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA
| | - C Robayo
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA
| | - K Hanson
- Florida ADRC, USA; University of Florida College of Medicine, Gainesville, FL, USA
| | - M Marsiske
- Florida ADRC, USA; University of Florida College of Medicine, Gainesville, FL, USA; University of Florida, College of Public Health and Health Professions, USA
| | - S Burke
- Florida ADRC, USA; Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - N Ertekin-Taner
- Mayo Clinic Florida, Department of Neurology, Jacksonville, FL, USA; Mayo Clinic Florida, Department of Neuroscience, Jacksonville, FL, USA
| | - D Vaillancourt
- Florida ADRC, USA; University of Florida College of Medicine, Gainesville, FL, USA
| | | | - T Golde
- Florida ADRC, USA; University of Florida College of Medicine, Gainesville, FL, USA
| | - DeKosky St
- Florida ADRC, USA; University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
183
|
Conrad J, Kremmyda O, Högen T, Brendel M, Rominger A, Levin J, Danek A. [Posterior cortical atrophy-a heterogeneous syndrome : A case series]. DER NERVENARZT 2019; 90:1045-1050. [PMID: 30903200 DOI: 10.1007/s00115-019-0697-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Julian Conrad
- Klinik und Poliklinik für Neurologie, Ludwig-Maximilians-Universität München, Marchioninistraße 15, 81377, München, Deutschland.
- Deutsches Schwindel- und Gleichgewichtszentrum (DSGZ), Ludwig-Maximilians-Universität München, München, Deutschland.
| | - Olympia Kremmyda
- Klinik und Poliklinik für Neurologie, Ludwig-Maximilians-Universität München, Marchioninistraße 15, 81377, München, Deutschland
- Deutsches Schwindel- und Gleichgewichtszentrum (DSGZ), Ludwig-Maximilians-Universität München, München, Deutschland
| | - Tobias Högen
- Klinik und Poliklinik für Neurologie, Ludwig-Maximilians-Universität München, Marchioninistraße 15, 81377, München, Deutschland
| | - Matthias Brendel
- Klinik und Poliklinik für Nuklearmedizin, Ludwig-Maximilians-Universität München, München, Deutschland
| | - Axel Rominger
- Universitätsklinik für Nuklearmedizin, Inselspital, Universitätsspital Bern, Bern, Schweiz
| | - Johannes Levin
- Klinik und Poliklinik für Neurologie, Ludwig-Maximilians-Universität München, Marchioninistraße 15, 81377, München, Deutschland
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Feodor-Lynen-Straße 17, 81377, München, Deutschland
- Munich Cluster for Systems Neurology (SyNergy), München, Deutschland
| | - Adrian Danek
- Klinik und Poliklinik für Neurologie, Ludwig-Maximilians-Universität München, Marchioninistraße 15, 81377, München, Deutschland
| |
Collapse
|
184
|
Zou R, Kuang G, Ågren H, Nordberg A, Långström B, Tu Y. Free Energy Profile for Penetration of Pittsburgh Compound-B into the Amyloid β Fibril. ACS Chem Neurosci 2019; 10:1783-1790. [PMID: 30698013 DOI: 10.1021/acschemneuro.8b00662] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The amyloid β (Aβ) fibril is a hallmark of Alzheimer's disease (AD) and has therefore served as an important target for early diagnosis of AD. The Pittsburgh Compound-B (PiB) is one of the most famous positron emission tomography (PET) tracers commonly used for in vivo detection of Aβ fibrils. Many theoretical studies have predicted the existence of various core binding sites with different microenvironments for probes binding to the Aβ fibril. However, little attention has been devoted to how the probes actually penetrate into the different core binding sites. In this study, an integrated molecular modeling scheme is used to study the penetration of PiB into the core binding sites of the Aβ1-42 fibril structure recently obtained by cryogenic electron microscopy. We find that there are two core binding sites for PiB with dramatic differences in cavity size and microenvironment properties, and furthermore that the penetration of PiB into site-1 is energetically prohibitive, whereas the penetration into site-2 is much more favorable. Therefore, the binding capacity at site-2 may be larger than that at site-1 despite its lower binding affinity. Our results thus suggest that site-2 may be a major binding site for PiB binding to Aβ fibril and emphasize the importance to adopt a full dynamical picture when studying tracer-fibril binding problems in general, something that in turn can be used to guide the development of tracers with higher affinity and selectivity for the Aβ fibril.
Collapse
Affiliation(s)
- Rongfeng Zou
- Department of Theoretical Chemistry and Biology, Royal Institute of Technology (KTH), AlbaNova University Center, S-106 91 Stockholm, Sweden
| | - Guanglin Kuang
- Department of Theoretical Chemistry and Biology, Royal Institute of Technology (KTH), AlbaNova University Center, S-106 91 Stockholm, Sweden
| | - Hans Ågren
- Department of Theoretical Chemistry and Biology, Royal Institute of Technology (KTH), AlbaNova University Center, S-106 91 Stockholm, Sweden
- College of Chemistry and Chemical Engineering, Henan University, Kaifeng, Henan 475004, P. R. China
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Center of Alzheimer Research, Clinical Geriatrics, Neo and Theme Aging, Karolinska University Hospital, Karolinska Institute, 141 83 Huddinge, Sweden
| | - Bengt Långström
- Department of Chemistry—BMC, Physical Organic Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Yaoquan Tu
- Department of Theoretical Chemistry and Biology, Royal Institute of Technology (KTH), AlbaNova University Center, S-106 91 Stockholm, Sweden
| |
Collapse
|
185
|
Li HC, Chen PY, Cheng HF, Kuo YM, Huang CC. In Vivo Visualization of Brain Vasculature in Alzheimer's Disease Mice by High-Frequency Micro-Doppler Imaging. IEEE Trans Biomed Eng 2019; 66:3393-3401. [PMID: 30872220 DOI: 10.1109/tbme.2019.2904702] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Cerebrovascular disorders are associated with Alzheimer's disease (AD). Functional analysis of the cerebral vasculature requires an in vivo approach to visualize the blood flow in small animal brains. This paper proposes a high-frequency micro-Doppler imaging (HFμDI) technology for mapping mouse cerebral vasculature. METHODS HFμDI used a 40-MHz transducer with an ultrafast ultrasound imaging technology that enabled in vivo visualization of the mouse brain up to 3 mm in depth; furthermore, a minimal vessel diameter of 48 μm could be determined. RESULTS Animal experiments determined that the cortical and hippocampal vessel density in young wild-type (WT) mice was similar to that in middle-aged WT mice. However, compared with the vessel density in middle-aged WT mice, that in middle-aged mice with AD was significantly lower, particularly in the hippocampus. DISCUSSION In vivo observation of cerebral vasculature demonstrated the effectiveness of HFμDI for the preclinical study of AD, and a potential way for human diagnosis was provided.
Collapse
|
186
|
Perez SE, Miguel JC, He B, Malek-Ahmadi M, Abrahamson EE, Ikonomovic MD, Lott I, Doran E, Alldred MJ, Ginsberg SD, Mufson EJ. Frontal cortex and striatal cellular and molecular pathobiology in individuals with Down syndrome with and without dementia. Acta Neuropathol 2019; 137:413-436. [PMID: 30734106 PMCID: PMC6541490 DOI: 10.1007/s00401-019-01965-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/18/2019] [Accepted: 01/22/2019] [Indexed: 02/06/2023]
Abstract
Although, by age 40, individuals with Down syndrome (DS) develop amyloid-β (Aβ) plaques and tau-containing neurofibrillary tangles (NFTs) linked to cognitive impairment in Alzheimer's disease (AD), not all people with DS develop dementia. Whether Aβ plaques and NFTs are associated with individuals with DS with (DSD +) and without dementia (DSD -) is under-investigated. Here, we applied quantitative immunocytochemistry and fluorescent procedures to characterize NFT pathology using antibodies specific for tau phosphorylation (pS422, AT8), truncation (TauC3, MN423), and conformational (Alz50, MC1) epitopes, as well as Aβ and its precursor protein (APP) to frontal cortex (FC) and striatal tissue from DSD + to DSD - cases. Expression profiling of single pS422 labeled FC layer V and VI neurons was also determined using laser capture microdissection and custom-designed microarray analysis. Analysis revealed that cortical and striatal Aβ plaque burdens were similar in DSD + and DSD - cases. In both groups, most FC plaques were neuritic, while striatal plaques were diffuse. By contrast, FC AT8-positive NFTs and neuropil thread densities were significantly greater in DSD + compared to DSD -, while striatal NFT densities were similar between groups. FC pS422-positive and TauC3 NFT densities were significantly greater than Alz50-labeled NFTs in DSD + , but not DSD - cases. Putaminal, but not caudate pS422-positive NFT density, was significantly greater than TauC3-positive NFTs. In the FC, AT8 + pS422 + Alz50, TauC3 + pS422 + Alz50, pS422 + Alz50, and TauC3 + pS422 positive NFTs were more frequent in DSD + compared to DSD- cases. Single gene-array profiling of FC pS422 positive neurons revealed downregulation of 63 of a total of 864 transcripts related to Aβ/tau biology, glutamatergic, cholinergic, and monoaminergic metabolism, intracellular signaling, cell homeostasis, and cell death in DSD + compared DSD - cases. These observations suggest that abnormal tau aggregation plays a critical role in the development of dementia in DS.
Collapse
Affiliation(s)
- Sylvia E Perez
- Department of Neurobiology and Neurology, Barrow Neurological Institute, 350 W. Thomas St, Phoenix, AZ, 85013, USA
- School of Life Sciences, College of Liberal Arts and Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Jennifer C Miguel
- Department of Neurobiology and Neurology, Barrow Neurological Institute, 350 W. Thomas St, Phoenix, AZ, 85013, USA
| | - Bin He
- Department of Neurobiology and Neurology, Barrow Neurological Institute, 350 W. Thomas St, Phoenix, AZ, 85013, USA
| | | | - Eric E Abrahamson
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, 15213, USA
- Departments of Neurology and Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Milos D Ikonomovic
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, 15213, USA
- Departments of Neurology and Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Ira Lott
- Departments of Pediatrics and Neurology, University of California, Irvine, CA, 92697, USA
| | - Eric Doran
- Departments of Pediatrics and Neurology, University of California, Irvine, CA, 92697, USA
| | - Melissa J Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
- Departments of Psychiatry, NYU Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10021, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
- Departments of Psychiatry, NYU Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10021, USA
- Departments of Neuroscience and Physiology, The NYU Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10021, USA
| | - Elliott J Mufson
- Department of Neurobiology and Neurology, Barrow Neurological Institute, 350 W. Thomas St, Phoenix, AZ, 85013, USA.
| |
Collapse
|
187
|
Brown BM, Peiffer J, Rainey-Smith SR. Exploring the relationship between physical activity, beta-amyloid and tau: A narrative review. Ageing Res Rev 2019; 50:9-18. [PMID: 30615936 DOI: 10.1016/j.arr.2019.01.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/06/2018] [Accepted: 01/03/2019] [Indexed: 12/19/2022]
Abstract
Several prospective cohort studies have reported an association between higher levels of physical activity and decreased risk of cognitive decline and dementia, years later. To support physical activity as a preventative measure against dementia, including Alzheimer's disease (AD; the most common form of dementia), evidence regarding the underlying mechanisms is vital. Here, we review previous work examining the role of physical activity in modulating levels of AD pathological hallmarks, beta-amyloid (Aβ) and tau (in the brain, cerebrospinal fluid and blood). Robust evidence from transgenic animal studies suggests that physical activity (voluntary wheel running) and exercise (forced wheel running) are implicated in lowering levels of brain Aβ and tau. Nevertheless, evidence from human studies, utilising measurements from positron emission tomography and cerebrospinal fluid biomarkers, is less consistent. Rigorous randomised controlled trials utilising long exercise interventions are vital to further understand the relationship between physical activity and Alzheimer's disease.
Collapse
|
188
|
Lin C, Huang C, Huang K, Lin K, Yen T, Kuo H. A metabolomic approach to identifying biomarkers in blood of Alzheimer's disease. Ann Clin Transl Neurol 2019; 6:537-545. [PMID: 30911577 PMCID: PMC6414491 DOI: 10.1002/acn3.726] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/20/2018] [Accepted: 12/31/2018] [Indexed: 01/28/2023] Open
Abstract
Objective This study aims to identify metabolites with altered levels of expression in patients with early and progressive stages of Alzheimer's disease (AD). Methods All participants of the study underwent genetic screening and were diagnosed using both neuropsychological assessment and amyloid imaging before metabolome analysis. According to these assessments, the patients were classified as normal (n = 15), with mild cognitive impairment (n = 10), and with AD (n = 15). Results Using a targeted metabolomic approach, we found that plasma levels of C3, C5, and C5-DC acylcarnitines, arginine, phenylalanine, creatinine, symmetric dimethylarginine (SDMA) and phosphatidylcholine ae C38:2 were significantly altered in patients with early and progressive stages of AD. We created a predictive model based on the decision tree that included three main parameters: age, arginine and C5 plasma concentrations. The model distinguished AD patients from other participants with 60% sensitivity and 86.7% specificity. For healthy controls, the sensitivity was 85.7% and specificity was 61.5%. Multivariate ROC analysis to develop a decision tree showed that our model reached moderate diagnostic power in differentiating between older adults who are cognitively normal (AUC = 0.77) and those with AD (AUC = 0.72). Interpretation The plasma levels of arginine and valeryl carnitine, together with subject age, are promising as biomarkers for the diagnosis of AD in older adults.
Collapse
Affiliation(s)
- Chia‐Ni Lin
- Department of Laboratory MedicineChang Gung Memorial HospitalTaoyuanTaiwan
- Department of Medical Biotechnology and Laboratory ScienceCollege of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Chin‐Chang Huang
- Department of NeurologyChang Gung Memorial Hospital at Linkou Medical CenterChang Gung University College of MedicineTaoyuanTaiwan
| | - Kuo‐Lun Huang
- Department of NeurologyChang Gung Memorial Hospital at Linkou Medical CenterChang Gung University College of MedicineTaoyuanTaiwan
| | - Kun‐Ju Lin
- Molecular Imaging Center and Department of Nuclear MedicineChang Gung Memorial HospitalTaoyuanTaiwan
- Department of Medical Imaging and Radiological SciencesHealthy Aging Research CenterChang Gung UniversityTaoyuanTaiwan
| | - Tzu‐Chen Yen
- Molecular Imaging Center and Department of Nuclear MedicineChang Gung Memorial HospitalTaoyuanTaiwan
- Department of Medical Imaging and Radiological SciencesHealthy Aging Research CenterChang Gung UniversityTaoyuanTaiwan
| | - Hung‐Chou Kuo
- Department of NeurologyChang Gung Memorial Hospital at Linkou Medical CenterChang Gung University College of MedicineTaoyuanTaiwan
| |
Collapse
|
189
|
Wilson H, Pagano G, Politis M. Dementia spectrum disorders: lessons learnt from decades with PET research. J Neural Transm (Vienna) 2019; 126:233-251. [PMID: 30762136 PMCID: PMC6449308 DOI: 10.1007/s00702-019-01975-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/21/2019] [Indexed: 02/07/2023]
Abstract
The dementia spectrum encompasses a range of disorders with complex diagnosis, pathophysiology and limited treatment options. Positron emission tomography (PET) imaging provides insights into specific neurodegenerative processes underlying dementia disorders in vivo. Here we focus on some of the most common dementias: Alzheimer's disease, Parkinsonism dementias including Parkinson's disease with dementia, dementia with Lewy bodies, progressive supranuclear palsy and corticobasal syndrome, and frontotemporal lobe degeneration. PET tracers have been developed to target specific proteinopathies (amyloid, tau and α-synuclein), glucose metabolism, cholinergic system and neuroinflammation. Studies have shown distinct imaging abnormalities can be detected early, in some cases prior to symptom onset, allowing disease progression to be monitored and providing the potential to predict symptom onset. Furthermore, advances in PET imaging have identified potential therapeutic targets and novel methods to accurately discriminate between different types of dementias in vivo. There are promising imaging markers with a clinical application on the horizon, however, further studies are required before they can be implantation into clinical practice.
Collapse
Affiliation(s)
- Heather Wilson
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, 125 Coldharbour Lane, Camberwell, London, SE5 9NU, UK
| | - Gennaro Pagano
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, 125 Coldharbour Lane, Camberwell, London, SE5 9NU, UK
| | - Marios Politis
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, 125 Coldharbour Lane, Camberwell, London, SE5 9NU, UK.
| |
Collapse
|
190
|
Clark LR, Koscik RL, Allison SL, Berman SE, Norton D, Carlsson CM, Betthauser TJ, Bendlin BB, Christian BT, Chin NA, Asthana S, Johnson SC. Hypertension and obesity moderate the relationship between β-amyloid and cognitive decline in midlife. Alzheimers Dement 2019; 15:418-428. [PMID: 30367828 PMCID: PMC6408972 DOI: 10.1016/j.jalz.2018.09.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/14/2018] [Accepted: 09/09/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND This study tested if central obesity, hypertension, or depressive symptoms moderated the relationship between β-amyloid (Aβ) and longitudinal cognitive performance in late middle-aged adults enriched for Alzheimer's disease (AD) risk. METHODS Participants (n = 207; ages = 40-70 years; 73% parental AD) in the Wisconsin Registry for Alzheimer's Prevention study completed 3+ neuropsychological evaluations and a [11C]PiB positron emission tomography scan or lumbar puncture. Linear mixed-effects regression models tested interactions of risk factor × Aβ × visit age on longitudinal Verbal Learning & Memory and Speed & Flexibility factor scores. RESULTS The relationship between Aβ and Verbal Learning & Memory decline was moderated by hypertension (χ2(1) = 3.85, P = .04) and obesity (χ2(1) = 6.12, P = .01); those with both elevated Aβ and the risk factor declined at faster rates than those with only elevated Aβ or elevated risk factors. CONCLUSION In this cohort, hypertension and obesity moderated the relationship between Aβ and cognitive decline.
Collapse
Affiliation(s)
- Lindsay R Clark
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| | - Rebecca L Koscik
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Samantha L Allison
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sara E Berman
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Medical Scientist and Neuroscience Training Programs, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Derek Norton
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA
| | - Cynthia M Carlsson
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Tobey J Betthauser
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Waisman Center and Department of Medical Physics, University of Wisconsin, Madison, WI, USA
| | - Barbara B Bendlin
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Bradley T Christian
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Waisman Center and Department of Medical Physics, University of Wisconsin, Madison, WI, USA
| | - Nathaniel A Chin
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sanjay Asthana
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Sterling C Johnson
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| |
Collapse
|
191
|
Jonkman LE, Kenkhuis B, Geurts JJG, van de Berg WDJ. Post-Mortem MRI and Histopathology in Neurologic Disease: A Translational Approach. Neurosci Bull 2019; 35:229-243. [PMID: 30790214 DOI: 10.1007/s12264-019-00342-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/29/2018] [Indexed: 01/28/2023] Open
Abstract
In this review, combined post-mortem brain magnetic resonance imaging (MRI) and histology studies are highlighted, illustrating the relevance of translational approaches to define novel MRI signatures of neuropathological lesions in neuroinflammatory and neurodegenerative disorders. Initial studies combining post-mortem MRI and histology have validated various MRI sequences, assessing their sensitivity and specificity as diagnostic biomarkers in neurologic disease. More recent studies have focused on defining new radiological (bio)markers and implementing them in the clinical (research) setting. By combining neurological and neuroanatomical expertise with radiological development and pathological validation, a cycle emerges that allows for the discovery of novel MRI biomarkers to be implemented in vivo. Examples of this cycle are presented for multiple sclerosis, Alzheimer's disease, Parkinson's disease, and traumatic brain injury. Some applications have been shown to be successful, while others require further validation. In conclusion, there is much to explore with post-mortem MRI and histology studies, which can eventually be of high relevance for clinical practice.
Collapse
Affiliation(s)
- Laura E Jonkman
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
| | - Boyd Kenkhuis
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen J G Geurts
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Wilma D J van de Berg
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| |
Collapse
|
192
|
Rinne JO, Suotunen T, Rummukainen J, Herukka SK, Nerg O, Koivisto AM, Rauramaa T, Någren K, Hiltunen M, Alafuzoff I, Rinne J, Jääskeläinen JE, Soininen H, Leinonen V. [11C]PIB PET Is Associated with the Brain Biopsy Amyloid-β Load in Subjects Examined for Normal Pressure Hydrocephalus. J Alzheimers Dis 2019; 67:1343-1351. [DOI: 10.3233/jad-180645] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Juha O. Rinne
- Turku PET Centre, University of Turku, Turku, Finland
- Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland
| | - Timo Suotunen
- Turku PET Centre, University of Turku, Turku, Finland
| | - Jaana Rummukainen
- Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland
| | - Sanna-Kaisa Herukka
- Neurology of NeuroCenter, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Kuopio, Finland
| | - Ossi Nerg
- Neurology of NeuroCenter, Kuopio University Hospital, Kuopio, Finland
| | - Anne M. Koivisto
- Neurology of NeuroCenter, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Kuopio, Finland
| | - Tuomas Rauramaa
- Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine, Pathology, University of Eastern Finland, Kuopio, Finland
| | - Kjell Någren
- Department of Nuclear Medicine, PET and Cyclotron Unit, Odense University Hospital, Odense, Denmark
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Irina Alafuzoff
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Jaakko Rinne
- Clinical Neurosciences, Department of Neurosurgery, Turku University Hospital, Turku, Finland
| | - Juha E. Jääskeläinen
- Institute of Clinical Medicine, Neurosurgery, University of Eastern Finland, Kuopio, Finland
- Neurosurgery of NeuroCenter, Kuopio University Hospital, Kuopio, Finland
| | - Hilkka Soininen
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Kuopio, Finland
| | - Ville Leinonen
- Institute of Clinical Medicine, Neurosurgery, University of Eastern Finland, Kuopio, Finland
- Neurosurgery of NeuroCenter, Kuopio University Hospital, Kuopio, Finland
- Unit of Clinical Neuroscience, Neurosurgery, University of Oulu and Medical Research Center, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
193
|
Bouter C, Vogelgsang J, Wiltfang J. Comparison between amyloid-PET and CSF amyloid-β biomarkers in a clinical cohort with memory deficits. Clin Chim Acta 2019; 492:62-68. [PMID: 30735665 DOI: 10.1016/j.cca.2019.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 11/25/2022]
Abstract
With increasing prevalence of Alzheimer's disease (AD) and advances in research of therapeutic approaches, an early and accurate in-vivo diagnosis is crucial. Different biomarkers that are able to identify AD are currently in focus. However, whether and to which extend results of cerebrospinal fluid (CSF) and imaging biomarkers are comparable, is unclear. This study aims to correlate CSF and amyloid imaging biomarkers comparing them to cognitive measurements in order to determine whether these methods provide identical or complementary information. The study comprises 33 consecutive patients with suspected cognitive decline that underwent lumbar puncture for CSF biomarker analysis and Amyloid-PET/CT within the diagnostic evaluation of memory impairment. Amyloid PET/CTs were evaluated visually and quantitatively. CSF and imaging data were retrospectively evaluated and results were compared to cognition tests, age, gender, and ApoE status. Global cortex SUVr levels correlated highly with CSF Aβ42/40 and moderately with Aβ42 but not with Aβ40. Global cortex SUVr and Aβ42/40 correlated with mini mental status examination. This study indicates that Amyloid-PET and CSF biomarkers might not reflect identical clinical information and a combination of both seems to be the most accurate way to characterize clinically unclear cognitive decline.
Collapse
Affiliation(s)
- Caroline Bouter
- University Medical Center Goettingen (UMG), Georg-August-University, Dept. of Nuclear Medicine, Robert-Koch-Str. 40, D-37075 Goettingen, Germany.
| | - Jonathan Vogelgsang
- University Medical Center Goettingen (UMG), Georg-August-University, Dept. of Psychiatry and Psychotherapy, Von-Siebold-Str. 5, D-37075 Goettingen, Germany
| | - Jens Wiltfang
- University Medical Center Goettingen (UMG), Georg-August-University, Dept. of Psychiatry and Psychotherapy, Von-Siebold-Str. 5, D-37075 Goettingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, D-37075 Goettingen, Germany; iBiMED, Medical Science Department, University of Aveiro, Portugal
| |
Collapse
|
194
|
La Joie R, Ayakta N, Seeley WW, Borys E, Boxer AL, DeCarli C, Doré V, Grinberg LT, Huang E, Hwang JH, Ikonomovic MD, Jack C, Jagust WJ, Jin LW, Klunk WE, Kofler J, Lesman-Segev OH, Lockhart SN, Lowe VJ, Masters CL, Mathis CA, McLean CL, Miller BL, Mungas D, O'Neil JP, Olichney JM, Parisi JE, Petersen RC, Rosen HJ, Rowe CC, Spina S, Vemuri P, Villemagne VL, Murray ME, Rabinovici GD. Multisite study of the relationships between antemortem [ 11C]PIB-PET Centiloid values and postmortem measures of Alzheimer's disease neuropathology. Alzheimers Dement 2019; 15:205-216. [PMID: 30347188 PMCID: PMC6368897 DOI: 10.1016/j.jalz.2018.09.001] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/08/2018] [Accepted: 09/03/2018] [Indexed: 10/28/2022]
Abstract
INTRODUCTION We sought to establish the relationships between standard postmortem measures of AD neuropathology and antemortem [11C]PIB-positron emission tomography ([11C]PIB-PET) analyzed with the Centiloid (CL) method, a standardized scale for Aβ-PET quantification. METHODS Four centers contributed 179 participants encompassing a broad range of clinical diagnoses, PET data, and autopsy findings. RESULTS CL values increased with each CERAD neuritic plaque score increment (median -3 CL for no plaques and 92 CL for frequent plaques) and nonlinearly with Thal Aβ phases (increases were detected starting at phase 2) with overlap between scores/phases. PET-pathology associations were comparable across sites and unchanged when restricting the analyses to the 56 patients who died within 2 years of PET. A threshold of 12.2 CL detected CERAD moderate-to-frequent neuritic plaques (area under the curve = 0.910, sensitivity = 89.2%, specificity = 86.4%), whereas 24.4 CL identified intermediate-to-high AD neuropathological changes (area under the curve = 0.894, sensitivity = 84.1%, specificity = 87.9%). DISCUSSION Our study demonstrated the robustness of a multisite Centiloid [11C]PIB-PET study and established a range of pathology-based CL thresholds.
Collapse
Affiliation(s)
- Renaud La Joie
- Memory & Aging Center, Department of Neurology, University of California, San Francisco, CA, USA.
| | - Nagehan Ayakta
- Memory & Aging Center, Department of Neurology, University of California, San Francisco, CA, USA; Helen Wills Neuroscience Institute, University of California Berkeley, CA, USA
| | - William W Seeley
- Memory & Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Ewa Borys
- Department of Pathology, Stritch School of Medicine, Loyola University, Maywood, IL, USA
| | - Adam L Boxer
- Memory & Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Charles DeCarli
- Department of Neurology, University of California, Davis, CA, USA
| | - Vincent Doré
- Department of Molecular Imaging & Therapy, Centre for PET, Austin Health, Heidelberg, Victoria, Australia
| | - Lea T Grinberg
- Memory & Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Eric Huang
- Memory & Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Ji-Hye Hwang
- Memory & Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, PA, USA
| | - Clifford Jack
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California Berkeley, CA, USA
| | - Lee-Way Jin
- Alzheimer's Disease Center, Department of Pathology, University of California Davis, CA, USA
| | - William E Klunk
- Department of Neurology, University of Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, PA, USA; Alzheimer's Disease Research Center, University of Pittsburgh, PA, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh, Pennsylvania, USA
| | - Orit H Lesman-Segev
- Memory & Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Samuel N Lockhart
- Helen Wills Neuroscience Institute, University of California Berkeley, CA, USA; Department of Internal Medicine, Division of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Val J Lowe
- Department of Nuclear Medicine, Mayo Clinic, Rochester, MN, USA
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | | | - Catriona L McLean
- Department of Anatomical Pathology, Alfred Hospital, Melbourne, Australia
| | - Bruce L Miller
- Memory & Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Daniel Mungas
- Department of Neurology, University of California, Davis, CA, USA
| | - James P O'Neil
- Helen Wills Neuroscience Institute, University of California Berkeley, CA, USA; Biomedical Isotope Facility, MBIB Division, Lawrence Berkeley National Laboratory, CA, USA
| | - John M Olichney
- Department of Neurology, University of California, Davis, CA, USA
| | - Joseph E Parisi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Howard J Rosen
- Memory & Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Christopher C Rowe
- Department of Molecular Imaging & Therapy, Centre for PET, Austin Health, Heidelberg, Victoria, Australia
| | - Salvatore Spina
- Memory & Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | | | - Victor L Villemagne
- Department of Molecular Imaging & Therapy, Centre for PET, Austin Health, Heidelberg, Victoria, Australia; The Florey Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | | | - Gil D Rabinovici
- Memory & Aging Center, Department of Neurology, University of California, San Francisco, CA, USA; Helen Wills Neuroscience Institute, University of California Berkeley, CA, USA
| |
Collapse
|
195
|
Striley CW, Milani SA, Kwiatkowski E, DeKosky ST, Cottler LB. Community perceptions related to brain donation: Evidence for intervention. Alzheimers Dement 2019; 15:267-272. [PMID: 30365929 PMCID: PMC7224447 DOI: 10.1016/j.jalz.2018.09.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 09/01/2018] [Accepted: 09/15/2018] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Research progress on neurocognitive disorders requires donation of both healthy and diseased brains. Here, we describe attitudes toward brain donation among a large community sample in Florida. METHODS HealthStreet, a community engagement program at the University of Florida, used community health workers to assess community attitudes toward research participation, including brain donation. RESULTS Over 60% of people, primarily Caucasian and employed, indicated that they would be likely or somewhat likely to donate their brain for research. Those who would be willing to donate were also more likely to be willing to participate in other research studies and to have participated in research. DISCUSSION Brain donation will add to the science of disorders of aging, including accurate diagnoses and validation of in vivo biomarkers. Increasing willingness to donate is a first step toward donation. Community populations are willing; community health workers can educate others about the need for this initiative in communities.
Collapse
|
196
|
Racine AM, Merluzzi AP, Adluru N, Norton D, Koscik RL, Clark LR, Berman SE, Nicholas CR, Asthana S, Alexander AL, Blennow K, Zetterberg H, Kim WH, Singh V, Carlsson CM, Bendlin BB, Johnson SC. Association of longitudinal white matter degeneration and cerebrospinal fluid biomarkers of neurodegeneration, inflammation and Alzheimer's disease in late-middle-aged adults. Brain Imaging Behav 2019; 13:41-52. [PMID: 28600739 PMCID: PMC5723250 DOI: 10.1007/s11682-017-9732-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is characterized by substantial neurodegeneration, including both cortical atrophy and loss of underlying white matter fiber tracts. Understanding longitudinal alterations to white matter may provide new insights into trajectories of brain change in both healthy aging and AD, and fluid biomarkers may be particularly useful in this effort. To examine this, 151 late-middle-aged participants enriched with risk for AD with at least one lumbar puncture and two diffusion tensor imaging (DTI) scans were selected for analysis from two large observational and longitudinally followed cohorts. Cerebrospinal fluid (CSF) was assayed for biomarkers of AD-specific pathology (phosphorylated-tau/Aβ42 ratio), axonal degeneration (neurofilament light chain protein, NFL), dendritic degeneration (neurogranin), and inflammation (chitinase-3-like protein 1, YKL-40). Linear mixed effects models were performed to test the hypothesis that biomarkers for AD, neurodegeneration, and inflammation, or two-year change in those biomarkers, would be associated with worse white matter health overall and/or progressively worsening white matter health over time. At baseline in the cingulum, phosphorylated-tau/Aβ42 was associated with higher mean diffusivity (MD) overall (intercept) and YKL-40 was associated with increases in MD over time. Two-year change in neurogranin was associated with higher mean diffusivity and lower fractional anisotropy overall (intercepts) across white matter in the entire brain and in the cingulum. These findings suggest that biomarkers for AD, neurodegeneration, and inflammation are potentially important indicators of declining white matter health in a cognitively healthy, late-middle-aged cohort.
Collapse
Affiliation(s)
- Annie M Racine
- Neuroscience and Public Policy Program, University of Wisconsin, Madison, WI, USA
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Andrew P Merluzzi
- Neuroscience and Public Policy Program, University of Wisconsin, Madison, WI, USA
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Nagesh Adluru
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Derek Norton
- Department of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI, 53792, USA
| | - Rebecca L Koscik
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Lindsay R Clark
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, 2500 Overlook Terrace, Madison, WI, 53705, USA
| | - Sara E Berman
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Christopher R Nicholas
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, 2500 Overlook Terrace, Madison, WI, 53705, USA
| | - Sanjay Asthana
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, 2500 Overlook Terrace, Madison, WI, 53705, USA
| | - Andrew L Alexander
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Kaj Blennow
- 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
| | - 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
- Institute of Neurology, University College London, London, UK
| | - Won Hwa Kim
- Department of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI, 53792, USA
- Department of Computer Sciences, University of Wisconsin - Madison, Madison, WI, 53706, USA
| | - Vikas Singh
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI, 53792, USA
- Department of Computer Sciences, University of Wisconsin - Madison, Madison, WI, 53706, USA
| | - Cynthia M Carlsson
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, 2500 Overlook Terrace, Madison, WI, 53705, USA
| | - Barbara B Bendlin
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Sterling C Johnson
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, 2500 Overlook Terrace, Madison, WI, 53705, USA.
| |
Collapse
|
197
|
Ba M, Ng KP, Gao X, Kong M, Guan L, Yu L. The combination of apolipoprotein E4, age and Alzheimer's Disease Assessment Scale - Cognitive Subscale improves the prediction of amyloid positron emission tomography status in clinically diagnosed mild cognitive impairment. Eur J Neurol 2019; 26:733-e53. [PMID: 30561868 DOI: 10.1111/ene.13881] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 12/06/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND PURPOSE Randomized clinical trials involving anti-amyloid interventions focus on the early stages of Alzheimer's disease (AD) with proven amyloid pathology, using amyloid positron emission tomography (amyloid-PET) imaging or cerebrospinal fluid analysis. However, these investigations are either expensive or invasive and are not readily available in resource-limited centres. Hence, the identification of cost-effective clinical alternatives to amyloid-PET is highly desirable. This study aimed to investigate the accuracy of combined clinical markers in predicting amyloid-PET status in mild cognitive impairment (MCI) individuals. METHODS In all, 406 MCI participants from the Alzheimer's Disease Neuroimaging Initiative database were dichotomized into amyloid-PET(+) and amyloid-PET(-) using a cut-off of >1.11. The accuracies of single clinical markers [apolipoprotein E4 (ApoE4) genotype, demographics, cognitive measures and cerebrospinal fluid analysis] in predicting amyloid-PET status were evaluated using receiver operating characteristic curve analysis. A logistic regression model was then used to determine the optimal model with combined clinical markers to predict amyloid-PET status. RESULTS Cerebrospinal fluid amyloid-β (Aβ) showed the best predictive accuracy of amyloid-PET status [area under the curve (AUC) = 0.927]. Whilst ApoE4 genotype (AUC = 0.737) and Alzheimer's Disease Assessment Scale - Cognitive Subscale (ADAS-Cog) 13 (AUC = 0.724) independently discriminated amyloid-PET(+) and amyloid-PET(-) MCI individuals, the combination of clinical markers (ApoE4 carrier, age >60 years and ADAS-Cog 13 > 13.5) improved the predictive accuracy of amyloid-PET status (AUC = 0.827, P < 0.001). CONCLUSIONS Cerebrospinal fluid Aβ, which is an invasive procedure, is most accurate in predicting amyloid-PET status in MCI individuals. The combination of ApoE4, age and ADAS-Cog 13 also accurately predicts amyloid-PET status. As this combination of clinical markers is cheap, non-invasive and readily available, it offers an attractive surrogate assessment for amyloid status amongst MCI individuals in resource-limited settings.
Collapse
Affiliation(s)
- M Ba
- Department of Neurology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - K P Ng
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
| | - X Gao
- Department of Neurology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - M Kong
- Department of Neurology, Yantaishan Hospital, Yantai City, China
| | - L Guan
- Department of Neurology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - L Yu
- Department of Neurology, Yantaishan Hospital, Yantai City, China
| | | |
Collapse
|
198
|
Abu Hamdeh S, Virhammar J, Sehlin D, Alafuzoff I, Cesarini KG, Marklund N. Brain tissue Aβ42 levels are linked to shunt response in idiopathic normal pressure hydrocephalus. J Neurosurg 2019; 130:121-129. [PMID: 29350601 DOI: 10.3171/2017.7.jns171005] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/24/2017] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The authors conducted a study to test if the cortical brain tissue levels of soluble amyloid beta (Aβ) reflect the propensity of cortical Aβ aggregate formation and may be an additional factor predicting surgical outcome following idiopathic normal pressure hydrocephalus (iNPH) treatment. METHODS Highly selective ELISAs (enzyme-linked immunosorbent assays) were used to quantify soluble Aβ40, Aβ42, and neurotoxic Aβ oligomers/protofibrils, associated with Aβ aggregation, in cortical biopsy samples obtained in patients with iNPH (n = 20), sampled during ventriculoperitoneal (VP) shunt surgery. Patients underwent pre- and postoperative (3-month) clinical assessment with a modified iNPH scale. The preoperative CSF biomarkers and the levels of soluble and insoluble Aβ species in cortical biopsy samples were analyzed for their association with a favorable outcome following the VP shunt procedure, defined as a ≥ 5-point increase in the iNPH scale. RESULTS The brain tissue levels of Aβ42 were negatively correlated with CSF Aβ42 (Spearman’s r = -0.53, p < 0.05). The Aβ40, Aβ42, and Aβ oligomer/protofibril levels in cortical biopsy samples were higher in patients with insoluble cortical Aβ aggregates (p < 0.05). The preoperative CSF Aβ42 levels were similar in patients responding (n = 11) and not responding (n = 9) to VP shunt treatment at 3 months postsurgery. In contrast, the presence of cortical Aβ aggregates and high brain tissue Aβ42 levels were associated with a poor outcome following VP shunt treatment (p < 0.05). CONCLUSIONS Brain tissue measurements of soluble Aβ species are feasible. Since high Aβ42 levels in cortical biopsy samples obtained in patients with iNPH indicated a poor surgical outcome, tissue levels of Aβ species may be associated with the clinical response to shunt treatment.
Collapse
Affiliation(s)
- Sami Abu Hamdeh
- 1Department of Neuroscience, Section of Neurosurgery, Uppsala University
| | - Johan Virhammar
- 2Department of Neuroscience, Neurology, Uppsala University Hospital, Uppsala University
| | - Dag Sehlin
- 3Department of Public Health and Caring Sciences/Geriatrics, Uppsala University; and
| | - Irina Alafuzoff
- 4Department of Pathology, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | | | - Niklas Marklund
- 1Department of Neuroscience, Section of Neurosurgery, Uppsala University
| |
Collapse
|
199
|
Risacher SL, Saykin AJ. Neuroimaging in aging and neurologic diseases. HANDBOOK OF CLINICAL NEUROLOGY 2019; 167:191-227. [PMID: 31753134 DOI: 10.1016/b978-0-12-804766-8.00012-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuroimaging biomarkers for neurologic diseases are important tools, both for understanding pathology associated with cognitive and clinical symptoms and for differential diagnosis. This chapter explores neuroimaging measures, including structural and functional measures from magnetic resonance imaging (MRI) and molecular measures primarily from positron emission tomography (PET), in healthy aging adults and in a number of neurologic diseases. The spectrum covers neuroimaging measures from normal aging to a variety of dementias: late-onset Alzheimer's disease [AD; including mild cognitive impairment (MCI)], familial and nonfamilial early-onset AD, atypical AD syndromes, posterior cortical atrophy (PCA), logopenic aphasia (lvPPA), cerebral amyloid angiopathy (CAA), vascular dementia (VaD), sporadic and familial behavioral-variant frontotemporal dementia (bvFTD), semantic dementia (SD), progressive nonfluent aphasia (PNFA), frontotemporal dementia with motor neuron disease (FTD-MND), frontotemporal dementia with amyotrophic lateral sclerosis (FTD-ALS), corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), dementia with Lewy bodies (DLB), Parkinson's disease (PD) with and without dementia, and multiple systems atrophy (MSA). We also include a discussion of the appropriate use criteria (AUC) for amyloid imaging and conclude with a discussion of differential diagnosis of neurologic dementia disorders in the context of neuroimaging.
Collapse
Affiliation(s)
- Shannon L Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
200
|
Cohen AD, Landau SM, Snitz BE, Klunk WE, Blennow K, Zetterberg H. Fluid and PET biomarkers for amyloid pathology in Alzheimer's disease. Mol Cell Neurosci 2018; 97:3-17. [PMID: 30537535 DOI: 10.1016/j.mcn.2018.12.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 12/05/2018] [Indexed: 02/04/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by amyloid plaques and tau pathology (neurofibrillary tangles and neuropil threads). Amyloid plaques are primarily composed of aggregated and oligomeric β-amyloid (Aβ) peptides ending at position 42 (Aβ42). The development of fluid and PET biomarkers for Alzheimer's disease (AD), has allowed for detection of Aβ pathology in vivo and marks a major advancement in understanding the role of Aβ in Alzheimer's disease (AD). In the recent National Institute on Aging and Alzheimer's Association (NIA-AA) Research Framework, AD is defined by the underlying pathology as measured in patients during life by biomarkers (Jack et al., 2018), while clinical symptoms are used for staging of the disease. Therefore, sensitive, specific and robust biomarkers to identify brain amyloidosis are central in AD research. Here, we discuss fluid and PET biomarkers for Aβ and their application.
Collapse
Affiliation(s)
- Ann D Cohen
- Department of Psychiatry, University of Pittsburgh School of Medicine, United States of America.
| | - Susan M Landau
- Neurology Helen Wills Neuroscience Institute, University of California, Berkeley, United States of America; Lawrence Berkeley National Laboratory, Molecular Biophysics and Integrated Bioimaging Functional Imaging Department, Life Sciences Division, United States of America
| | - Beth E Snitz
- Department of Neurology, University of Pittsburgh School of Medicine, United States of America
| | - William E Klunk
- Department of Psychiatry, University of Pittsburgh School of Medicine, United States of America
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Molndal, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, University College, London, United Kingdom of Great Britain and Northern Ireland
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Molndal, Sweden; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, University College, London, United Kingdom of Great Britain and Northern Ireland; Department of Molecular Neuroscience, UCL Institute of Neurology, United Kingdom of Great Britain and Northern Ireland; UK Dementia Research Institute at UCL, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|