101
|
Breitner J, Dodge HH, Khachaturian ZS, Khachaturian AS. "Exceptions that prove the rule"-Why have clinical trials failed to show efficacy of risk factor interventions suggested by observational studies of the dementia-Alzheimer's disease syndrome? Alzheimers Dement 2022; 18:389-392. [PMID: 35245406 PMCID: PMC8940699 DOI: 10.1002/alz.12633] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Indexed: 12/28/2022]
Affiliation(s)
- John Breitner
- Douglas Hospital Research Center and McGill University, Quebec, Canada
| | - Hiroko H. Dodge
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | | | | |
Collapse
|
102
|
Duan Y, Lv J, Zhang Z, Chen Z, Wu H, Chen J, Chen Z, Yang J, Wang D, Liu Y, Chen F, Tian Y, Cao X. Exogenous Aβ 1-42 monomers improve synaptic and cognitive function in Alzheimer's disease model mice. Neuropharmacology 2022; 209:109002. [PMID: 35196539 DOI: 10.1016/j.neuropharm.2022.109002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/28/2022] [Accepted: 02/15/2022] [Indexed: 01/16/2023]
Abstract
Growing evidence has suggested the poor correlation between brain amyloid plaque and Alzheimer's disease (AD). Presenilin1 (PS1) and presenilin2 (PS2) conditional double knockout (cDKO) mice exhibited the reduced 42-amino acid amyloid-β peptide (Aβ1-42) level and AD-like symptoms, indicating a different pathological mechanism from the amyloid cascade hypothesis for AD. Here we found that exogenous synthetic Aβ1-42 monomers could improve the impaired memory not only in cDKO mice without Aβ1-42 deposition but also in the APP/PS1/Tau triple transgenic 3 × Tg-AD mice with Aβ1-42 deposition, which were mediated by α7-nAChR. Our findings demonstrate for the first time that reduced soluble Aβ1-42 level is the main cause of cognitive dysfunction in cDKO mice, and support the opinions that low soluble Aβ1-42 level due to Aβ1-42 deposition may also cause cognitive deficits in 3 × Tg-AD mice. Therefore, "loss-of-function" of Aβ1-42 should be avoided when designing therapies aimed at reducing Aβ1-42 burden in AD.
Collapse
Affiliation(s)
- Yanhong Duan
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Junyan Lv
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Zhonghui Zhang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Zhenzhen Chen
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Hao Wu
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Jinnan Chen
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Zhidong Chen
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Jiarun Yang
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Dasheng Wang
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Yamei Liu
- School of Life Sciences, Shanghai University, No. 99 Shangda Rd., Shanghai, 200444, PR China
| | - Fuxue Chen
- School of Life Sciences, Shanghai University, No. 99 Shangda Rd., Shanghai, 200444, PR China
| | - Yang Tian
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Xiaohua Cao
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China.
| |
Collapse
|
103
|
Mahaman YAR, Embaye KS, Huang F, Li L, Zhu F, Wang JZ, Liu R, Feng J, Wang X. Biomarkers used in Alzheimer's disease diagnosis, treatment, and prevention. Ageing Res Rev 2022; 74:101544. [PMID: 34933129 DOI: 10.1016/j.arr.2021.101544] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 12/09/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD), being the number one in terms of dementia burden, is an insidious age-related neurodegenerative disease and is presently considered a global public health threat. Its main histological hallmarks are the Aβ senile plaques and the P-tau neurofibrillary tangles, while clinically it is marked by a progressive cognitive decline that reflects the underlying synaptic loss and neurodegeneration. Many of the drug therapies targeting the two pathological hallmarks namely Aβ and P-tau have been proven futile. This is probably attributed to the initiation of therapy at a stage where cognitive alterations are already obvious. In other words, the underlying neuropathological changes are at a stage where these drugs lack any therapeutic value in reversing the damage. Therefore, there is an urgent need to start treatment in the very early stage where these changes can be reversed, and hence, early diagnosis is of primordial importance. To this aim, the use of robust and informative biomarkers that could provide accurate diagnosis preferably at an earlier phase of the disease is of the essence. To date, several biomarkers have been established that, to a different extent, allow researchers and clinicians to evaluate, diagnose, and more specially exclude other related pathologies. In this study, we extensively reviewed data on the currently explored biomarkers in terms of AD pathology-specific and non-specific biomarkers and highlighted the recent developments in the diagnostic and theragnostic domains. In the end, we have presented a separate elaboration on aspects of future perspectives and concluding remarks.
Collapse
|
104
|
Yawson GK, Will MF, Huffman SE, Strandquist ET, Bothwell PJ, Oliver EB, Apuzzo CF, Platt DC, Weitzel CS, Jones MA, Ferrence GM, Hamaker CG, Webb MI. A Dual-Pronged Approach: A Ruthenium(III) Complex That Modulates Amyloid-β Aggregation and Disrupts Its Formed Aggregates. Inorg Chem 2022; 61:2733-2744. [PMID: 35102739 DOI: 10.1021/acs.inorgchem.1c01651] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a devastating neurological disorder for which soluble oligomers of the peptide amyloid-β (Aβ) are now recognized as the neurotoxic species. Metal-based therapeutics are uniquely suited to target Aβ, with ruthenium-based (Ru) complexes emerging as propitious candidates. Recently, azole-based Ru(III) complexes were observed to modulate the aggregation of Aβ in solution, where the inclusion of a primary amine proximal to the ligand coordination site improved the activity of the complexes. To advance these structure-activity relationships, a series of oxazole-based Ru complexes were prepared and evaluated for their ability to modulate Aβ aggregation. From these studies, a lead candidate, Oc, emerged that had superior activity relative to its azole predecessors in modulating the aggregation of soluble Aβ and diminishing its cytotoxicity. Further evaluation of Oc demonstrated its ability to disrupt formed Aβ aggregates, resulting in smaller amorphous species. Because altering both sides of the aggregation equilibrium for Aβ has not been previously suggested for metal-based complexes for AD, this work represents an exciting new avenue for improved therapeutic success.
Collapse
Affiliation(s)
- Gideon K Yawson
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Mark F Will
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Samantha E Huffman
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Evan T Strandquist
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Paige J Bothwell
- Core Microscope Facility, Department of Biological Sciences, Northern Illinois University, DeKalb, Illinois 60115, United States
| | - Ethan B Oliver
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - C Fiore Apuzzo
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - David C Platt
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Christopher S Weitzel
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Marjorie A Jones
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Gregory M Ferrence
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Christopher G Hamaker
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Michael I Webb
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| |
Collapse
|
105
|
Adult Hippocampal Neurogenesis in Alzheimer’s Disease: An Overview of Human and Animal Studies with Implications for Therapeutic Perspectives Aimed at Memory Recovery. Neural Plast 2022; 2022:9959044. [PMID: 35075360 PMCID: PMC8783751 DOI: 10.1155/2022/9959044] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/21/2021] [Accepted: 12/15/2021] [Indexed: 12/31/2022] Open
Abstract
The mammalian hippocampal dentate gyrus is a niche for adult neurogenesis from neural stem cells. Newborn neurons integrate into existing neuronal networks, where they play a key role in hippocampal functions, including learning and memory. In the ageing brain, neurogenic capability progressively declines while in parallel increases the risk for developing Alzheimer's disease (AD), the main neurodegenerative disorder associated with memory loss. Numerous studies have investigated whether impaired adult neurogenesis contributes to memory decline in AD. Here, we review the literature on adult hippocampal neurogenesis (AHN) and AD by focusing on both human and mouse model studies. First, we describe key steps of AHN, report recent evidence of this phenomenon in humans, and describe the specific contribution of newborn neurons to memory, as evinced by animal studies. Next, we review articles investigating AHN in AD patients and critically examine the discrepancies among different studies over the last two decades. Also, we summarize researches investigating AHN in AD mouse models, and from these studies, we extrapolate the contribution of molecular factors linking AD-related changes to impaired neurogenesis. Lastly, we examine animal studies that link impaired neurogenesis to specific memory dysfunctions in AD and review treatments that have the potential to rescue memory capacities in AD by stimulating AHN.
Collapse
|
106
|
Longhurst JK, Cummings JL, John SE, Poston B, Rider JV, Salazar AM, Mishra VR, Ritter A, Caldwell JZ, Miller JB, Kinney JW, Landers MR. Dual Task Performance Is Associated with Amyloidosis in Cognitively Healthy Adults. J Prev Alzheimers Dis 2022; 9:297-305. [PMID: 35543003 PMCID: PMC9286710 DOI: 10.14283/jpad.2022.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Preclinical Alzheimer's disease (AD) provides an opportunity for the study and implementation of interventions and strategies aimed at delaying, mitigating, and preventing AD. While this preclinical state is an ideal target, it is difficult to identify efficiently and cost-effectively. Recent findings have suggested that cognitive-motor dual task paradigms may provide additional inference. OBJECTIVES Investigate the relationship between dual task performance and amyloidosis, suggestive of preclinical Alzheimer's disease and whether dual task performance provides additional information beyond a cognitive composite, to help in the identification of amyloidosis. DESIGN Cross-sectional. SETTING Outpatient specialty brain health clinical research institution in the United States. PARTICIPANTS 52 cognitively healthy adults. MEASUREMENTS The data included demographics, amyloid standardized uptake value ratio obtained via florbetapir-PET, neuropsychological testing, apolipoprotien E genotype, and dual task performance measures. Data were analyzed via hierarchal multiple linear regression or logistic regression, controlling for age, education, and apolipoprotien E genotype. Receiver operating characteristic curves were plotted, and sensitivity and specificity calculated via 2x2 contingency tables. RESULTS There was a moderate relationship (rs>.30) between motor and cognitive dual task effects and amyloid standardized uptake value ratio (ps<.042). A strong relationship (r=.58) was found between combined dual task effect, a measure of automaticity derived from dual task performance, and amyloid standardized uptake value ratio (p<.001). Additionally, combined dual task effect showed promise in its unique contributions to amyloid standardized uptake value ratio, accounting for 7.8% of amyloid standardized uptake value ratio variance beyond cognitive composite scores (p=.018). Additionally, when incorporated into the cognitive composite, combined dual task effect resulted in improved diagnostic accuracy for determining elevated amyloid standardized uptake value ratio, and increased the sensitivity and specificity of the cognitive composite. CONCLUSSION Dual task performance using the combined dual task effect, a measure of automaticity, was a moderate predictor of cerebral amyloidosis, which suggests that it has utility in the screening and diagnosis of individuals for preclinical AD. Additionally, when combined with the cognitive composite, the combined dual task effect improves diagnostic accuracy. Further research is warranted.
Collapse
Affiliation(s)
- J K Longhurst
- Jason K. Longhurst, PT, DPT, PHD, Department of Physical Therapy and Athletic Training, Saint Louis University, Saint Louis, Missouri, USA, 63104, , tel: 314-977-8533, fax: 314-977-8513
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
107
|
Bohlke K, Zhu X, Sparto PJ, Redfern MS, Rosano C, Sejdic E, Rosso AL. The Effect of a Verbal Cognitive Task on Postural Sway Does Not Persist When the Task Is Over. SENSORS (BASEL, SWITZERLAND) 2021; 21:8428. [PMID: 34960520 PMCID: PMC8704561 DOI: 10.3390/s21248428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/01/2022]
Abstract
Dual-task balance studies explore interference between balance and cognitive tasks. This study is a descriptive analysis of accelerometry balance metrics to determine if a verbal cognitive task influences postural control after the task ends. Fifty-two healthy older adults (75 ± 6 years old, 30 female) performed standing balance and cognitive dual-tasks. An accelerometer recorded movement from before, during, and after the task (reciting every other letter of the alphabet). Thirty-six balance metrics were calculated for each task condition. The effect of the cognitive task on postural control was determined by a generalized linear model. Twelve variables, including anterior-posterior centroid frequency, peak frequency and entropy rate, medial-later entropy rate and wavelet entropy, and bandwidth in all directions, exhibited significant differences between baseline and cognitive task periods, but not between baseline and post-task periods. These results indicate that the verbal cognitive task did alter balance, but did not bring about persistent effects after the task had ended. Traditional balance measurements, i.e., root mean square and normalized path length, notably lacked significance, highlighting the potential to use other accelerometer metrics for the early detection of balance problems. These novel insights into the temporal dynamics of dual-task balance support current dual-task paradigms to reduce fall risk in older adults.
Collapse
Affiliation(s)
- Kayla Bohlke
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | - Xiaonan Zhu
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15260, USA; (X.Z.); (C.R.); (A.L.R.)
| | - Patrick J. Sparto
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | - Mark S. Redfern
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | - Caterina Rosano
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15260, USA; (X.Z.); (C.R.); (A.L.R.)
| | - Ervin Sejdic
- The Edward S. Rogers Department of Electrical and Computer Engineering, Faculty of Applied Science and Engineering, University of Toronto, Toronto, ON M5S 1A1, Canada;
- Research & Innovation Department, North York General Hospital, Toronto, ON M2K 1E1, Canada
| | - Andrea L. Rosso
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15260, USA; (X.Z.); (C.R.); (A.L.R.)
| |
Collapse
|
108
|
Soluble amyloid-beta isoforms predict downstream Alzheimer's disease pathology. Cell Biosci 2021; 11:204. [PMID: 34895338 PMCID: PMC8665586 DOI: 10.1186/s13578-021-00712-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 11/11/2021] [Indexed: 11/25/2022] Open
Abstract
Background Changes in soluble amyloid-beta (Aβ) levels in cerebrospinal fluid (CSF) are detectable at early preclinical stages of Alzheimer’s disease (AD). However, whether Aβ levels can predict downstream AD pathological features in cognitively unimpaired (CU) individuals remains unclear. With this in mind, we aimed at investigating whether a combination of soluble Aβ isoforms can predict tau pathology (T+) and neurodegeneration (N+) positivity. Methods We used CSF measurements of three soluble Aβ peptides (Aβ1–38, Aβ1–40 and Aβ1–42) in CU individuals (n = 318) as input features in machine learning (ML) models aiming at predicting T+ and N+. Input data was used for building 2046 tuned predictive ML models with a nested cross-validation technique. Additionally, proteomics data was employed to investigate the functional enrichment of biological processes altered in T+ and N+ individuals. Results Our findings indicate that Aβ isoforms can predict T+ and N+ with an area under the curve (AUC) of 0.929 and 0.936, respectively. Additionally, proteomics analysis identified 17 differentially expressed proteins (DEPs) in individuals wrongly classified by our ML model. More specifically, enrichment analysis of gene ontology biological processes revealed an upregulation in myelinization and glucose metabolism-related processes in CU individuals wrongly predicted as T+. A significant enrichment of DEPs in pathways including biosynthesis of amino acids, glycolysis/gluconeogenesis, carbon metabolism, cell adhesion molecules and prion disease was also observed. Conclusions Our results demonstrate that, by applying a refined ML analysis, a combination of Aβ isoforms can predict T+ and N+ with a high AUC. CSF proteomics analysis highlighted a promising group of proteins that can be further explored for improving T+ and N+ prediction. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00712-3.
Collapse
|
109
|
Nagase T, Tohda C. Skeletal muscle atrophy-induced hemopexin accelerates onset of cognitive impairment in Alzheimer's disease. J Cachexia Sarcopenia Muscle 2021; 12:2199-2210. [PMID: 34658156 PMCID: PMC8718090 DOI: 10.1002/jcsm.12830] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is an unmet medical need worldwide, and physical inactivity is a risk factor for AD. Performing physical exercise is difficult at old age, and thus, decline in physical movement may be a cause of age-associated lowering of the brain function. This study aimed to elucidate the molecular mechanism and onset of the skeletal muscle atrophy-induced acceleration of AD. METHODS Pre-symptomatic young 5XFAD or non-transgenic wildtype mice were used. The bilateral hindlimbs were immobilized by placing them in casts for 14 days. Cognitive function was evaluated using the object recognition and spatial memory tests. Further, the hindlimb muscles were isolated for organ culture. Conditioned media (CM) of each muscle was separated by two-dimensional polyacrylamide gel electrophoresis (2D-PAGE). Protein expressions in the CM were analysed by matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry analysis. The expression levels of candidate proteins were quantified using ELISA. After continuous intracerebroventricular (i.c.v.) infusion of recombinant hemopexin, cognitive function was evaluated. Gene microarray analysis of the hippocampus was performed to investigate the molecules involved in the accelerated memory deficit. Real-time reverse transcription polymerase chain reaction and histological analysis confirmed the expression. RESULTS Casting for 2 weeks reduced skeletal muscle weight. Object recognition memory in the cast-attached 5XFAD mice (n = 7, training vs. test, P = 0.3390) was impaired than that in age-matched wildtype (n = 7, training vs. test, P = 0.0523) and non-cast 5XFAD mice (n = 7, training vs. test, P = 0.0473). On 2D-PAGE, 88 spots were differentially expressed in muscle CM. The most increased spot in the cast-attached 5XFAD CM was hemopexin. Hemopexin levels in the skeletal muscle (n = 3, P = 0.0064), plasma (n = 3, P = 0.0386), and hippocampus (n = 3, P = 0.0164) were increased in cast-attached 5XFAD mice than those in non-cast 5XFAD mice. Continuous i.c.v. infusion of hemopexin for 2 weeks induced memory deficits in young 5XFAD mice (n = 4, training vs. test, P = 0.6764). Lipocalin-2 (Lcn2) messenger RNA (mRNA), neuroinflammation-associated factor, was increased in the hippocampus in hemopexin-infused 5XFAD mice than in control mice. LCN2 protein in the hippocampus was localized in the neurons, but not glial cells. Lcn2 mRNA levels in the hippocampus were also increased by cast-immobilization of the hindlimbs (n = 6, P = 0.0043). CONCLUSIONS These findings provide new evidence indicating that skeletal muscle atrophy has an unbeneficial impact on the occurrence of memory impairment in young 5XFAD mice, which is mediated by the muscle secreted hemopexin.
Collapse
Affiliation(s)
- Tsukasa Nagase
- Section of Neuromedical Science, Division of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Chihiro Tohda
- Section of Neuromedical Science, Division of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
110
|
Ghosh U, Yau WM, Collinge J, Tycko R. Structural differences in amyloid-β fibrils from brains of nondemented elderly individuals and Alzheimer's disease patients. Proc Natl Acad Sci U S A 2021; 118:e2111863118. [PMID: 34725161 PMCID: PMC8609303 DOI: 10.1073/pnas.2111863118] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 09/20/2021] [Indexed: 02/05/2023] Open
Abstract
Although amyloid plaques composed of fibrillar amyloid-β (Aβ) assemblies are a diagnostic hallmark of Alzheimer's disease (AD), quantities of amyloid similar to those in AD patients are observed in brain tissue of some nondemented elderly individuals. The relationship between amyloid deposition and neurodegeneration in AD has, therefore, been unclear. Here, we use solid-state NMR to investigate whether molecular structures of Aβ fibrils from brain tissue of nondemented elderly individuals with high amyloid loads differ from structures of Aβ fibrils from AD tissue. Two-dimensional solid-state NMR spectra of isotopically labeled Aβ fibrils, prepared by seeded growth from frontal lobe tissue extracts, are similar in the two cases but with statistically significant differences in intensity distributions of cross-peak signals. Differences in solid-state NMR data are greater for 42-residue amyloid-β (Aβ42) fibrils than for 40-residue amyloid-β (Aβ40) fibrils. These data suggest that similar sets of fibril polymorphs develop in nondemented elderly individuals and AD patients but with different relative populations on average.
Collapse
Affiliation(s)
- Ujjayini Ghosh
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892-0520
- Department of Chemistry, Michigan State University, East Lansing, MI 48824
| | - Wai-Ming Yau
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892-0520
| | - John Collinge
- Medical Research Council Prion Unit, University College London, London W1W 7FF, United Kingdom
- Institute of Prion Diseases, University College London, London W1W 7FF, United Kingdom
| | - Robert Tycko
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892-0520;
| |
Collapse
|
111
|
Lloret A, Esteve D, Lloret MA, Cervera-Ferri A, Lopez B, Nepomuceno M, Monllor P. When Does Alzheimer's Disease Really Start? The Role of Biomarkers. FOCUS: JOURNAL OF LIFE LONG LEARNING IN PSYCHIATRY 2021; 19:355-364. [PMID: 34690605 DOI: 10.1176/appi.focus.19305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
(Appeared originally in Int J Mol Sci 2019, 20 5536).
Collapse
Affiliation(s)
- Ana Lloret
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain; Department of Clinic Neurophysiology. University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain; Department of Human Anatomy and Embriology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; Department of Neurology. University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain
| | - Daniel Esteve
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain; Department of Clinic Neurophysiology. University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain; Department of Human Anatomy and Embriology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; Department of Neurology. University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain
| | - Maria-Angeles Lloret
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain; Department of Clinic Neurophysiology. University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain; Department of Human Anatomy and Embriology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; Department of Neurology. University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain
| | - Ana Cervera-Ferri
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain; Department of Clinic Neurophysiology. University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain; Department of Human Anatomy and Embriology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; Department of Neurology. University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain
| | - Begoña Lopez
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain; Department of Clinic Neurophysiology. University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain; Department of Human Anatomy and Embriology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; Department of Neurology. University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain
| | - Mariana Nepomuceno
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain; Department of Clinic Neurophysiology. University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain; Department of Human Anatomy and Embriology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; Department of Neurology. University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain
| | - Paloma Monllor
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Avda. Blasco Ibanez, 17, 46010 Valencia, Spain; Department of Clinic Neurophysiology. University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain; Department of Human Anatomy and Embriology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; Department of Neurology. University Clinic Hospital of Valencia, Avda. Blasco Ibanez, 19, 46010 Valencia, Spain
| |
Collapse
|
112
|
Shea TB. Improvement of cognitive performance by a nutraceutical formulation: Underlying mechanisms revealed by laboratory studies. Free Radic Biol Med 2021; 174:281-304. [PMID: 34352370 DOI: 10.1016/j.freeradbiomed.2021.07.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 12/28/2022]
Abstract
Cognitive decline, decrease in neuronal function and neuronal loss that accompany normal aging and dementia are the result of multiple mechanisms, many of which involve oxidative stress. Herein, we review these various mechanisms and identify pharmacological and non-pharmacological approaches, including modification of diet, that may reduce the risk and progression of cognitive decline. The optimal degree of neuronal protection is derived by combinations of, rather than individual, compounds. Compounds that provide antioxidant protection are particularly effective at delaying or improving cognitive performance in the early stages of Mild Cognitive Impairment and Alzheimer's disease. Laboratory studies confirm alleviation of oxidative damage in brain tissue. Lifestyle modifications show a degree of efficacy and may augment pharmacological approaches. Unfortunately, oxidative damage and resultant accumulation of biomarkers of neuronal damage can precede cognitive decline by years to decades. This underscores the importance of optimization of dietary enrichment, antioxidant supplementation and other lifestyle modifications during aging even for individuals who are cognitively intact.
Collapse
Affiliation(s)
- Thomas B Shea
- Laboratory for Neuroscience, Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA.
| |
Collapse
|
113
|
Malerba HN, Pereira AAR, Pierrobon MF, Abrao GS, Toricelli M, Akamine EH, Buck HS, Viel TA. Combined Neuroprotective Strategies Blocked Neurodegeneration and Improved Brain Function in Senescence-Accelerated Mice. Front Aging Neurosci 2021; 13:681498. [PMID: 34497504 PMCID: PMC8419356 DOI: 10.3389/fnagi.2021.681498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 07/19/2021] [Indexed: 01/22/2023] Open
Abstract
Increase in the quality of life, combined with drug strategies, has been studied as possibilities for improving memory and delaying the onset of neurodegenerative diseases. A previous study published by the group of the authors has shown that microdose lithium and enriched environment can improve memory in both mice and humans. To elucidate this relationship better, this study aimed to evaluate whether the chronic combination of these two strategies could increase healthy aging in Senescence Accelerated Mouse-Prone 8 (SAMP8). Animals were submitted to either one or both of these strategies until the age of 10 months when they were anesthetized and killed and their hippocampus was extracted. The untreated SAMP-8 group exhibited worse memory and reduced neuronal density with greater neurodegeneration and increased amyloid-β plaque density compared with the control group. Moreover, significant alterations in proteins related to long-term potentiation, such as, synaptophysin and brain-derived neurotrophic factor (BDNF), were observed in this group. The strategies used in the study maintained long-term memory, reduced anxiety, and increased neuroprotection. Both strategies were efficient in reducing neurodegeneration and increasing parameters related to memory maintenance. In many experiments, the combination of the two strategies was more effective in improving healthy aging. This study sheds light on the combination of strategies that choose to improve the quality of life and drugs with low side effects. Moreover, it opens perspectives for a new field of study for healthy aging.
Collapse
Affiliation(s)
- Helena Nascimento Malerba
- Graduate Course on Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil.,Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, São Paulo, Brazil
| | - Arthur Antonio Ruiz Pereira
- Graduate Course on Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil.,Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, São Paulo, Brazil
| | - Marcela Favoretto Pierrobon
- Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, São Paulo, Brazil
| | - Guilherme Souza Abrao
- Graduate Course on Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil.,Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, São Paulo, Brazil
| | - Mariana Toricelli
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Eliana Hiromi Akamine
- Graduate Course on Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Hudson Sousa Buck
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Tania Araujo Viel
- Graduate Course on Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil.,Laboratory of Neuropharmacology of Aging, School of Arts, Sciences and Humanities, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
114
|
Wilkins HM, Swerdlow RH. Mitochondrial links between brain aging and Alzheimer's disease. Transl Neurodegener 2021; 10:33. [PMID: 34465385 PMCID: PMC8408998 DOI: 10.1186/s40035-021-00261-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/21/2021] [Indexed: 02/08/2023] Open
Abstract
Advancing age is a major risk factor for Alzheimer's disease (AD). This raises the question of whether AD biology mechanistically diverges from aging biology or alternatively represents exaggerated aging. Correlative and modeling studies can inform this question, but without a firm grasp of what drives aging and AD it is difficult to definitively resolve this quandary. This review speculates over the relevance of a particular hallmark of aging, mitochondrial function, to AD, and further provides background information that is pertinent to and provides perspective on this speculation.
Collapse
Affiliation(s)
- Heather M Wilkins
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA
- Departments of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- Departments of Biochemistry and Molecular Biology, Medical Center, University of Kansas Medical Center, Kansas City, USA
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Research Center, Kansas City, KS, USA.
- Departments of Neurology, University of Kansas Medical Center, Kansas City, KS, USA.
- Departments of Biochemistry and Molecular Biology, Medical Center, University of Kansas Medical Center, Kansas City, USA.
- Departments of Molecular and Integrative Physiology, Medical Center, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
115
|
Rahman MM, Lendel C. Extracellular protein components of amyloid plaques and their roles in Alzheimer's disease pathology. Mol Neurodegener 2021; 16:59. [PMID: 34454574 PMCID: PMC8400902 DOI: 10.1186/s13024-021-00465-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 06/11/2021] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is pathologically defined by the presence of fibrillar amyloid β (Aβ) peptide in extracellular senile plaques and tau filaments in intracellular neurofibrillary tangles. Extensive research has focused on understanding the assembly mechanisms and neurotoxic effects of Aβ during the last decades but still we only have a brief understanding of the disease associated biological processes. This review highlights the many other constituents that, beside Aβ, are accumulated in the plaques, with the focus on extracellular proteins. All living organisms rely on a delicate network of protein functionality. Deposition of significant amounts of certain proteins in insoluble inclusions will unquestionably lead to disturbances in the network, which may contribute to AD and copathology. This paper provide a comprehensive overview of extracellular proteins that have been shown to interact with Aβ and a discussion of their potential roles in AD pathology. Methods that can expand the knowledge about how the proteins are incorporated in plaques are described. Top-down methods to analyze post-mortem tissue and bottom-up approaches with the potential to provide molecular insights on the organization of plaque-like particles are compared. Finally, a network analysis of Aβ-interacting partners with enriched functional and structural key words is presented.
Collapse
Affiliation(s)
- M Mahafuzur Rahman
- Department of Chemistry, KTH Royal Institute of Technology, SE-100 44, Stockholm, Sweden.
| | - Christofer Lendel
- Department of Chemistry, KTH Royal Institute of Technology, SE-100 44, Stockholm, Sweden.
| |
Collapse
|
116
|
Jiménez-Balado J, Eich TS. GABAergic dysfunction, neural network hyperactivity and memory impairments in human aging and Alzheimer's disease. Semin Cell Dev Biol 2021; 116:146-159. [PMID: 33573856 PMCID: PMC8292162 DOI: 10.1016/j.semcdb.2021.01.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/25/2021] [Accepted: 01/30/2021] [Indexed: 02/07/2023]
Abstract
In this review, we focus on the potential role of the γ-aminobutyric acidergic (GABAergic) system in age-related episodic memory impairments in humans, with a particular focus on Alzheimer's disease (AD). Well-established animal models have shown that GABA plays a central role in regulating and synchronizing neuronal signaling in the hippocampus, a brain area critical for episodic memory that undergoes early and significant morphologic and functional changes in the course of AD. Neuroimaging research in humans has documented hyperactivity in the hippocampus and losses of resting state functional connectivity in the Default Mode Network, a network that itself prominently includes the hippocampus-presaging episodic memory decline in individuals at-risk for AD. Apolipoprotein ε4, the highest genetic risk factor for AD, is associated with GABAergic dysfunction in animal models, and episodic memory impairments in humans. In combination, these findings suggest that GABA may be the linchpin in a complex system of factors that eventually leads to the principal clinical hallmark of AD: episodic memory loss. Here, we will review the current state of literature supporting this hypothesis. First, we will focus on the molecular and cellular basis of the GABAergic system and its role in memory and cognition. Next, we report the evidence of GABA dysregulations in AD and normal aging, both in animal models and human studies. Finally, we outline a model of GABAergic dysfunction based on the results of functional neuroimaging studies in humans, which have shown hippocampal hyperactivity to episodic memory tasks concurrent with and even preceding AD diagnosis, along with factors that may modulate this association.
Collapse
Affiliation(s)
- Joan Jiménez-Balado
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Teal S Eich
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
117
|
Wang YY, Sun YP, Luo YM, Peng DH, Li X, Yang BY, Wang QH, Kuang HX. Biomarkers for the Clinical Diagnosis of Alzheimer's Disease: Metabolomics Analysis of Brain Tissue and Blood. Front Pharmacol 2021; 12:700587. [PMID: 34366852 PMCID: PMC8333692 DOI: 10.3389/fphar.2021.700587] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/08/2021] [Indexed: 01/09/2023] Open
Abstract
With an increase in aging populations worldwide, age-related diseases such as Alzheimer's disease (AD) have become a global concern. At present, a cure for neurodegenerative disease is lacking. There is an urgent need for a biomarker that can facilitate the diagnosis, classification, prognosis, and treatment response of AD. The recent emergence of highly sensitive mass-spectrometry platforms and high-throughput technology can be employed to discover and catalog vast datasets of small metabolites, which respond to changed status in the body. Metabolomics analysis provides hope for a better understanding of AD as well as the subsequent identification and analysis of metabolites. Here, we review the state-of-the-art emerging candidate biomarkers for AD.
Collapse
Affiliation(s)
- Yang-Yang Wang
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yan-Ping Sun
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yu-Meng Luo
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Dong-Hui Peng
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiao Li
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Bing-You Yang
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qiu-Hong Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hai-Xue Kuang
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
118
|
King-Robson J, Wilson H, Politis M. Associations Between Amyloid and Tau Pathology, and Connectome Alterations, in Alzheimer's Disease and Mild Cognitive Impairment. J Alzheimers Dis 2021; 82:541-560. [PMID: 34057079 DOI: 10.3233/jad-201457] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The roles of amyloid-β and tau in the degenerative process of Alzheimer's disease (AD) remain uncertain. [18F]AV-45 and [18F]AV-1451 PET quantify amyloid-β and tau pathology, respectively, while diffusion tractography enables detection of their microstructural consequences. OBJECTIVE Examine the impact of amyloid-β and tau pathology on the structural connectome and cognition, in mild cognitive impairment (MCI) and AD. METHODS Combined [18F]AV-45 and [18F]AV-1451 PET, diffusion tractography, and cognitive assessment in 28 controls, 32 MCI, and 26 AD patients. RESULTS Hippocampal connectivity was reduced to the thalami, right lateral orbitofrontal, and right amygdala in MCI; alongside the insula, posterior cingulate, right entorhinal, and numerous cortical regions in AD (all p < 0.05). Hippocampal strength inversely correlated with [18F]AV-1451 SUVr in MCI (r = -0.55, p = 0.049) and AD (r = -0.57, p = 0.046), while reductions in hippocampal connectivity to ipsilateral brain regions correlated with increased [18F]AV-45 SUVr in those same regions in MCI (r = -0.33, p = 0.003) and AD (r = -0.31, p = 0.006). Cognitive scores correlated with connectivity of the right temporal pole in MCI (r = -0.60, p = 0.035) and left hippocampus in AD (r = 0.69, p = 0.024). Clinical Dementia Rating Scale scores correlated with [18F]AV-1451 SUVr in multiple areas reflecting Braak stages I-IV, including the right (r = 0.65, p = 0.004) entorhinal cortex in MCI; and Braak stages III-VI, including the right (r = 0.062, p = 0.009) parahippocampal gyrus in AD. CONCLUSION Reductions in hippocampal connectivity predominate in the AD connectome, correlating with hippocampal tau in MCI and AD, and with amyloid-β in the target regions of those connections. Cognitive scores correlate with microstructural changes and reflect the accumulation of tau pathology.
Collapse
Affiliation(s)
- Josh King-Robson
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Heather Wilson
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK.,Neurodegeneration Imaging Group, University of Exeter Medical School, London, UK
| | - Marios Politis
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK.,Neurodegeneration Imaging Group, University of Exeter Medical School, London, UK
| | | |
Collapse
|
119
|
Öhman F, Hassenstab J, Berron D, Schöll M, Papp KV. Current advances in digital cognitive assessment for preclinical Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12217. [PMID: 34295959 PMCID: PMC8290833 DOI: 10.1002/dad2.12217] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/30/2021] [Accepted: 06/04/2021] [Indexed: 12/24/2022]
Abstract
There is a pressing need to capture and track subtle cognitive change at the preclinical stage of Alzheimer's disease (AD) rapidly, cost-effectively, and with high sensitivity. Concurrently, the landscape of digital cognitive assessment is rapidly evolving as technology advances, older adult tech-adoption increases, and external events (i.e., COVID-19) necessitate remote digital assessment. Here, we provide a snapshot review of the current state of digital cognitive assessment for preclinical AD including different device platforms/assessment approaches, levels of validation, and implementation challenges. We focus on articles, grants, and recent conference proceedings specifically querying the relationship between digital cognitive assessments and established biomarkers for preclinical AD (e.g., amyloid beta and tau) in clinically normal (CN) individuals. Several digital assessments were identified across platforms (e.g., digital pens, smartphones). Digital assessments varied by intended setting (e.g., remote vs. in-clinic), level of supervision (e.g., self vs. supervised), and device origin (personal vs. study-provided). At least 11 publications characterize digital cognitive assessment against AD biomarkers among CN. First available data demonstrate promising validity of this approach against both conventional assessment methods (moderate to large effect sizes) and relevant biomarkers (predominantly weak to moderate effect sizes). We discuss levels of validation and issues relating to usability, data quality, data protection, and attrition. While still in its infancy, digital cognitive assessment, especially when administered remotely, will undoubtedly play a major future role in screening for and tracking preclinical AD.
Collapse
Affiliation(s)
- Fredrik Öhman
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
| | - Jason Hassenstab
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
- Department of Psychological & Brain SciencesWashington University in St. LouisSt. LouisMissouriUSA
| | - David Berron
- German Center for Neurodegenerative Diseases (DZNE)MagdeburgGermany
- Clinical Memory Research Unit, Department of Clinical Sciences MalmöLund UniversityLundSweden
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- Dementia Research Centre, Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Kathryn V. Papp
- Center for Alzheimer Research and TreatmentDepartment of Neurology, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Department of Neurology, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
120
|
Koinuma S, Shimozawa N, Yasutomi Y, Kimura N. Aging induces abnormal accumulation of Aβ in extracellular vesicle and/or intraluminal membrane vesicle-rich fractions in nonhuman primate brain. Neurobiol Aging 2021; 106:268-281. [PMID: 34329965 DOI: 10.1016/j.neurobiolaging.2021.06.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 06/10/2021] [Accepted: 06/28/2021] [Indexed: 12/30/2022]
Abstract
Aβ metabolism in the brain is mediated by endocytosis, one part of the intracellular membrane trafficking system. We previously showed that aging attenuates the interaction of dynein with dynactin, which disrupts the endosomal/lysosomal trafficking pathway involved in Aβ metabolism, resulting in intracellular accumulation of Aβ. Several studies have shown that in Alzheimer's disease (AD), intraneuronal accumulation of Aβ precedes extracellular Aβ depositions. However, it is unclear what accounts for this transition from intracellular to extracellular depositions. Accumulating evidence suggest that autophagy has an important role in AD pathology, and we observed that autophagy-related protein levels began to decrease before amyloid plaque formation in cynomolgus monkey brains. Surprisingly, experimental induction of autophagosome formation in Neuro2a cells significantly increased intracellular Aβ and decreased extracellular release of Aβ, accompanied by the prominent reduction of extracellular vesicle (EV) secretion. RNAi study confirmed that EV secretion affected intracellular and extracellular Aβ levels, and siRNA-induced downregulation of autophagosome formation enhanced EV secretion to ameliorate intracellular Aβ accumulation induced by dynein knockdown. In aged cynomolgus monkeys, Aβ levels in EV/intraluminal membrane vesicle (ILV)-rich fractions isolated from temporal lobe parenchyma were drastically increased. Moreover, EV/ILV marker proteins overlapped spatially with amyloid plaques. These findings suggest that EV would be an important carrier of Aβ in brain and abnormal accumulation of Aβ in EVs/ILVs may be involved in the transition of age-dependent Aβ pathology.
Collapse
Affiliation(s)
- Shingo Koinuma
- Section of Cell Biology and Pathology, Department of Alzheimer's Disease Research, Center for Development of Advanced Medicine for Dementia, Obu, Aichi, Japan; Division of Biosignaling, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Nobuhiro Shimozawa
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki, Japan
| | - Yasuhiro Yasutomi
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki, Japan
| | - Nobuyuki Kimura
- Section of Cell Biology and Pathology, Department of Alzheimer's Disease Research, Center for Development of Advanced Medicine for Dementia, Obu, Aichi, Japan; Laboratory of Experimental Animals, Research and Development Management Center, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan; Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
121
|
Sideris DI, Danial JSH, Emin D, Ruggeri FS, Xia Z, Zhang YP, Lobanova E, Dakin H, De S, Miller A, Sang JC, Knowles TPJ, Vendruscolo M, Fraser G, Crowther D, Klenerman D. Soluble amyloid beta-containing aggregates are present throughout the brain at early stages of Alzheimer's disease. Brain Commun 2021; 3:fcab147. [PMID: 34396107 PMCID: PMC8361392 DOI: 10.1093/braincomms/fcab147] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/08/2021] [Accepted: 05/14/2021] [Indexed: 12/02/2022] Open
Abstract
Protein aggregation likely plays a key role in the initiation and spreading of Alzheimer's disease pathology through the brain. Soluble aggregates of amyloid beta are believed to play a key role in this process. However, the aggregates present in humans are still poorly characterized due to a lack of suitable methods required for characterizing the low concentration of heterogeneous aggregates present. We have used a variety of biophysical methods to characterize the aggregates present in human Alzheimer's disease brains at Braak stage III. We find soluble amyloid beta-containing aggregates in all regions of the brain up to 200 nm in length, capable of causing an inflammatory response. Rather than aggregates spreading through the brain as disease progresses, it appears that aggregation occurs all over the brain and that different brain regions are at earlier or later stages of the same process, with the later stages causing increased inflammation.
Collapse
Affiliation(s)
- Dimitrios I Sideris
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- Neuroscience, Research and Early Development, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB21 6GH, UK
| | - John S H Danial
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Derya Emin
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Francesco S Ruggeri
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- Laboratories of Organic and Physical Chemistry, Wageningen University, Wageningen 6703 WE, Netherlands
| | - Zengjie Xia
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Yu P Zhang
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Evgeniia Lobanova
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Helen Dakin
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Suman De
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Alyssa Miller
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Jason C Sang
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Tuomas P J Knowles
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- Cavendish Laboratory, University of Cambridge, Cambridge CB3 0H3, UK
| | - Michele Vendruscolo
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Graham Fraser
- Neuroscience, Research and Early Development, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB21 6GH, UK
| | - Damian Crowther
- Neuroscience, Research and Early Development, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB21 6GH, UK
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute at Cambridge, Cambridge CB2 0XY, UK
| |
Collapse
|
122
|
Mattos MK, Chang A, Pitcher K, Whitt C, Ritterband LM, Quigg MS. A Review of Insomnia Treatments for Patients with Mild Cognitive Impairment. Aging Dis 2021; 12:1036-1042. [PMID: 34221547 PMCID: PMC8219491 DOI: 10.14336/ad.2021.0423] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/23/2021] [Indexed: 12/21/2022] Open
Abstract
Mild cognitive impairment (MCI) impacts approximately 20% of older adults, with many also experiencing sleep disorders, such as insomnia. Given the relationship between sleep and dementia, addressing sleep issues may offer an opportunity to treat reversible causes. There are two primary treatments for insomnia: behavioral-based (cognitive behavioral therapy for insomnia, CBT-I) and pharmacological interventions. Although CBT-I is recommended as first-line treatment for insomnia in older adults, sedative-hypnotics are more likely to be recommended than non-pharmacological treatments given their convenience and accessibility. However, there are significant concerns in prescribing medications to patients with MCI. To explore this disconnect, we reviewed insomnia treatments in older adults with MCI studies and current guidelines of pharmacological therapy. First, we reviewed studies presenting non-pharmacological treatment of insomnia in older adults with MCI. Although the search yielded over 4,000 non-duplicate titles, only one article presented data on non-pharmacological treatment of insomnia in MCI. The literature covering comorbid insomnia, CBT-I, and MCI is sparse. In contrast to review of non-pharmacological studies, studies on the pharmacological treatment of insomnia in older adults were ample. Finally, we reviewed international guidelines for pharmacological treatment of insomnia in cognitive disorders. More widely used pharmacological interventions show short-term effectiveness with problems of recurrence, ineffectiveness in inadvertent or purposeful chronic use, and adverse side effects. Despite evidence regarding adverse consequences, pharmacological treatment of insomnia remains the most common treatment for insomnia. Reflecting on age-related changes in older adults, particularly those with MCI, inappropriate or mismanagement of medication can lead to unnecessary complications. Further research examining effective behavioral-based sleep management options in older adults with cognitive impairment is needed with exploration of improved sleep on cognitive function.
Collapse
Affiliation(s)
- Meghan K Mattos
- University of Virginia, School of Nursing, Charlottesville, VA 22903, USA.
| | - Angela Chang
- Northwestern University Feinberg School of Medicine, Department of Medical Social Sciences, Chicago, IL 60611, USA.
| | - Katherine Pitcher
- University of Virginia Medical Center, Charlottesville, VA 22903, USA.
| | - Carley Whitt
- University of Virginia, School of Medicine, Charlottesville, VA 22903, USA.
| | - Lee M Ritterband
- University of Virginia, School of Medicine, Charlottesville, VA 22903, USA.
| | - Mark S Quigg
- University of Virginia, School of Medicine, Charlottesville, VA 22903, USA.
| |
Collapse
|
123
|
Lau HHC, Ingelsson M, Watts JC. The existence of Aβ strains and their potential for driving phenotypic heterogeneity in Alzheimer's disease. Acta Neuropathol 2021; 142:17-39. [PMID: 32743745 DOI: 10.1007/s00401-020-02201-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/17/2022]
Abstract
Reminiscent of the human prion diseases, there is considerable clinical and pathological variability in Alzheimer's disease, the most common human neurodegenerative condition. As in prion disorders, protein misfolding and aggregation is a hallmark feature of Alzheimer's disease, where the initiating event is thought to be the self-assembly of Aβ peptide into aggregates that deposit in the central nervous system. Emerging evidence suggests that Aβ, similar to the prion protein, can polymerize into a conformationally diverse spectrum of aggregate strains both in vitro and within the brain. Moreover, certain types of Aβ aggregates exhibit key hallmarks of prion strains including divergent biochemical attributes and the ability to induce distinct pathological phenotypes when intracerebrally injected into mouse models. In this review, we discuss the evidence demonstrating that Aβ can assemble into distinct strains of aggregates and how such strains may be primary drivers of the phenotypic heterogeneity in Alzheimer's disease.
Collapse
|
124
|
Sordo L, Martini AC, Houston EF, Head E, Gunn-Moore D. Neuropathology of Aging in Cats and its Similarities to Human Alzheimer’s Disease. FRONTIERS IN AGING 2021; 2:684607. [PMID: 35822024 PMCID: PMC9261448 DOI: 10.3389/fragi.2021.684607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/27/2021] [Indexed: 11/15/2022]
Abstract
Elderly cats develop age-related behavioral and neuropathological changes that ultimately lead to cognitive dysfunction syndrome (CDS). These neuropathologies share similarities to those seen in the brains of humans with Alzheimer’s disease (AD), including the extracellular accumulation of ß-amyloid (Aβ) and intraneuronal deposits of hyperphosphorylated tau, which are considered to be the two major hallmarks of AD. The present study assessed the presence and distribution of Aβ and tau hyperphosphorylation within the cat brain (n = 55 cats), and how the distribution of these proteins changes with age and the presence of CDS. For this, immunohistochemistry was performed on seven brain regions from cats of various ages, with and without CDS (n = 10 with CDS). Cats accumulate both intracytoplasmic and extracellular deposits of Aβ, as well as intranuclear and intracytoplasmic hyperphosphorylated tau deposits. Large extracellular aggregates of Aβ were found in elderly cats, mainly in the cortical brain areas, with occasional hippocampal aggregates. This may suggest that these aggregates start in cortical areas and later progress to the hippocampus. While Aβ senile plaques in people with AD have a dense core, extracellular Aβ deposits in cats exhibited a diffuse pattern, similar to the early stages of plaque pathogenesis. Intraneuronal Aβ deposits were also observed, occurring predominantly in cortical brain regions of younger cats, while older cats had few to no intraneuronal Aβ deposits, especially when extracellular aggregates were abundant. Intracytoplasmic hyperphosphorylated tau was found within neurons in the brains of elderly cats, particularly in those with CDS. Due to their ultrastructural features, these deposits are considered to be pre-tangles, which are an early stage of the neurofibrillary tangles seen in AD. The largest numbers of pre-tangles are found mainly in the cerebral cortex of elderly cats, whereas lower numbers were found in other regions (i.e., entorhinal cortex and hippocampus). For the first time, intranuclear tau was found in both phosphorylated and non-phosphorylated states within neurons in the cat brain. The highest numbers of intranuclear deposits were found in the cortex of younger cats, and this tended to decrease with age. In contrast, elderly cats with pre-tangles had only occasional or no nuclear labelling.
Collapse
Affiliation(s)
- Lorena Sordo
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
- *Correspondence: Lorena Sordo,
| | - Alessandra C. Martini
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| | - E. Fiona Houston
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| | - Danièlle Gunn-Moore
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
125
|
Mitrano DA, Houle SE, Pearce P, Quintanilla RM, Lockhart BK, Genovese BC, Schendzielos RA, Croushore EE, Dymond EM, Bogenpohl JW, Grau HJ, Webb LS. Olfactory dysfunction in the 3xTg-AD model of Alzheimer's disease. IBRO Neurosci Rep 2021; 10:51-61. [PMID: 33842910 PMCID: PMC8019944 DOI: 10.1016/j.ibneur.2020.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 12/13/2020] [Indexed: 12/01/2022] Open
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disease in which the risk of development increases with age. People with AD are plagued with deficits in their cognition, memory, and basic social skills. Many of these deficits are believed to be caused by the formation of amyloid-β plaques and neurofibrillary tangles in regions of the brain associated with memory, such as the hippocampus. However, one of the early, preclinical symptoms of AD is the loss of olfactory detection and discrimination. To determine if a mouse model of AD expresses the same olfactory dysfunction seen in human AD, 3xTg-AD mice were given a buried food test and, unlike previous studies, compared to their background and parental strains. Results showed that over 52 weeks, the 3xTg-AD mice took significantly longer to find the buried food than the control strains. The olfactory bulbs of the 3xTg-AD mice were removed, sliced, and stained using Congo red for histological analysis. Amyloid deposits were observed predominantly in the granule layer of the olfactory bulb beginning at 13 weeks of age in 3xTg-AD mice, but not in the control strains of mice. Further examination of the buried food test data revealed that 3xTg-AD females had a significantly longer latency to detect the buried food than males beginning at 26 weeks of age. Overall, this study provides further validation of the 3xTg-AD mouse model of AD and supports the idea that simple olfactory testing could be part of the diagnostic process for human AD.
Collapse
Affiliation(s)
- Darlene A. Mitrano
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
- Program in Neuroscience, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Sam E. Houle
- Program in Neuroscience, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Patrick Pearce
- Program in Neuroscience, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Ricardo M. Quintanilla
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Blakely K. Lockhart
- Program in Neuroscience, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Benjamin C. Genovese
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Rachel A. Schendzielos
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Emma E. Croushore
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Ethan M. Dymond
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - James W. Bogenpohl
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
- Program in Neuroscience, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Harold J. Grau
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
- Program in Neuroscience, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Lisa Smith Webb
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
- Program in Neuroscience, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| |
Collapse
|
126
|
Kitaigorodsky M, Crocco E, Curiel‐Cid RE, Leal G, Zheng D, Eustache MK, Greig‐Custo MT, Barker W, Duara R, Loewenstein DA. The relationship of semantic intrusions to different etiological subtypes of MCI and cognitively healthy older adults. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12192. [PMID: 34084887 PMCID: PMC8144934 DOI: 10.1002/dad2.12192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/23/2021] [Accepted: 03/30/2021] [Indexed: 01/23/2023]
Abstract
INTRODUCTION There is increasing evidence that susceptibility to proactive semantic interference (PSI) and the failure to recover from PSI (frPSI) as evidenced by intrusion errors may be early cognitive markers of both preclinical and prodromal Alzheimer's disease (AD). METHODS One hundred forty-five participants were administered extensive clinical and neuropsychological evaluations including the Loewenstein-Acevedo Scales for Semantic Interference and Learning (LASSI-L), a sensitive cognitive stress test measuring PSI and frPSI. Participants also underwent structural magnetic resonance imaging (MRI) and amyloid positron emission tomography/computed tomography (PET/CT) imaging. RESULTS PSI and frPSI errors were much more prevalent in the mild cognitive impairment (MCI)-AD (amyloid positive) group than the other diagnostic groups. The number of intrusion errors observed across the other MCI groups without amyloid pathology and those with normal cognition were comparable. DISCUSSION Semantic intrusion errors on the LASSI-L occur much less frequently in persons who have different types of non-AD-related MCI and may be used as an early cognitive marker of prodromal AD.
Collapse
Affiliation(s)
- Marcela Kitaigorodsky
- Center for Cognitive Neuroscience and Aging, Department of Psychiatry and Behavioral SciencesUniversity of Miami Miller School of MedicineMiamiUSA
| | - Elizabeth Crocco
- Center for Cognitive Neuroscience and Aging, Department of Psychiatry and Behavioral SciencesUniversity of Miami Miller School of MedicineMiamiUSA
| | - Rosie E. Curiel‐Cid
- Center for Cognitive Neuroscience and Aging, Department of Psychiatry and Behavioral SciencesUniversity of Miami Miller School of MedicineMiamiUSA
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
| | - Giselle Leal
- Center for Cognitive Neuroscience and Aging, Department of Psychiatry and Behavioral SciencesUniversity of Miami Miller School of MedicineMiamiUSA
| | - Diane Zheng
- Center for Cognitive Neuroscience and Aging, Department of Psychiatry and Behavioral SciencesUniversity of Miami Miller School of MedicineMiamiUSA
| | - Melissa K. Eustache
- Center for Cognitive Neuroscience and Aging, Department of Psychiatry and Behavioral SciencesUniversity of Miami Miller School of MedicineMiamiUSA
| | - Maria T. Greig‐Custo
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
- Wien Center for Alzheimer's Disease and Memory DisordersMount Sinai, Medical CenterNew YorkUSA
| | - William Barker
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
- Wien Center for Alzheimer's Disease and Memory DisordersMount Sinai, Medical CenterNew YorkUSA
| | - Ranjan Duara
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
- Wien Center for Alzheimer's Disease and Memory DisordersMount Sinai, Medical CenterNew YorkUSA
| | - David A. Loewenstein
- Center for Cognitive Neuroscience and Aging, Department of Psychiatry and Behavioral SciencesUniversity of Miami Miller School of MedicineMiamiUSA
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
| |
Collapse
|
127
|
Tahmi M, Rippon B, Palta P, Sherwood G, Hernandez G, Soto L, Ceballos F, Pardo M, Laing K, Igwe K, He H, Teresi JA, Moreno H, Razlighi Q, Brickman AM, Luchsinger JA. In Vivo Amyloid, Neurodegeneration, and Verbal Learning in Late Middle-Aged Hispanics. J Alzheimers Dis 2021; 82:317-325. [PMID: 34024821 DOI: 10.3233/jad-201304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The National Institute on Aging (NIA)/Alzheimer's Association (AA) 2018 framework conceptualizes Alzheimer's disease (AD) biologically. Evidence of brain amyloid by biomarkers defines AD pathologic change and the Alzheimer's continuum. The presence of tau or neurodegeneration in the absence of amyloid defines non-AD pathologic change. OBJECTIVE To examine the relation of in vivo amyloid and neurodegeneration with verbal learning, one of the cognitive abilities affected early in AD, in late middle age. METHODS This was a cross-sectional study of amyloid and neurodegeneration biomarkers in a community-based cohort of 350 late-middle aged Hispanics without dementia (mean age: 64.15±3.34; 72.0%women). Amyloid (A) was measured as global standardized uptake value ratio (SUVR) with 18F-Florbetaben positron emission tomography (PET). Neurodegeneration (N) was ascertained as cortical thickness (CT) in AD signature areas using brain magnetic resonance imaging. We examined A/N continuously, categorically, by A/N profiles, and profile categories. The amyloid threshold for positivity was defined using the K means method. The CT threshold was defined as 2 standard deviations below the mean CT. Verbal learning was ascertained using total recall and delayed recall in the Buschke Selective Reminding test (SRT). RESULTS Higher cortical thickness was associated with higher performance in SRT delayed recall. Amyloid SUVR was not related to SRT performance. The low CT category was associated with lower performance in SRT delayed recall, while Amyloid categories were not related to any SRT score. The non-AD pathologic change group (A-N+) performed worse in SRT delayed recall compared to the Normal A/N profile group (A-N-). CONCLUSION In late middle-aged Hispanics without dementia, non-AD pathologic change, but not the Alzheimer's continuum, was related to verbal learning.
Collapse
Affiliation(s)
- Mouna Tahmi
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Brady Rippon
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Priya Palta
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.,Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| | - Greysi Sherwood
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Gabriela Hernandez
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Luisa Soto
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Fernando Ceballos
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Michelle Pardo
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Krystal Laing
- Department of Neurology, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Kay Igwe
- Department of Neurology, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Hengda He
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | | - Herman Moreno
- Department of Neurology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | | | - Adam M Brickman
- Department of Neurology, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA.,Gertrude H. Sergievsky Center, Columbia University Irving Medical Center, New York, NY, USA
| | - José A Luchsinger
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.,Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
128
|
Alves SS, Silva-Junior RMPD, Servilha-Menezes G, Homolak J, Šalković-Petrišić M, Garcia-Cairasco N. Insulin Resistance as a Common Link Between Current Alzheimer's Disease Hypotheses. J Alzheimers Dis 2021; 82:71-105. [PMID: 34024838 DOI: 10.3233/jad-210234] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Almost 115 years ago, Alois Alzheimer described Alzheimer's disease (AD) for the first time. Since then, many hypotheses have been proposed. However, AD remains a severe health public problem. The current medical approaches for AD are limited to symptomatic interventions and the complexity of this disease has led to a failure rate of approximately 99.6%in AD clinical trials. In fact, no new drug has been approved for AD treatment since 2003. These failures indicate that we are failing in mimicking this disease in experimental models. Although most studies have focused on the amyloid cascade hypothesis of AD, the literature has made clear that AD is rather a multifactorial disorder. Therefore, the persistence in a single theory has resulted in lost opportunities. In this review, we aim to present the striking points of the long scientific path followed since the description of the first AD case and the main AD hypotheses discussed over the last decades. We also propose insulin resistance as a common link between many other hypotheses.
Collapse
Affiliation(s)
- Suélen Santos Alves
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Rui Milton Patrício da Silva-Junior
- Department of Internal Medicine, Ribeirão Preto Medical School -University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Gabriel Servilha-Menezes
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Jan Homolak
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Melita Šalković-Petrišić
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Norberto Garcia-Cairasco
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
129
|
Wang YJ, Hu H, Yang YX, Zuo CT, Tan L, Yu JT. Regional Amyloid Accumulation and White Matter Integrity in Cognitively Normal Individuals. J Alzheimers Dis 2021; 74:1261-1270. [PMID: 32176644 DOI: 10.3233/jad-191350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Recent studies have shown that amyloid-β (Aβ) burden influenced white matter (WM) integrity before the onset of dementia. OBJECTIVE To assess whether the effects of Aβ burden on WM integrity in cognitively normal (CN) individuals were regionally specific. METHODS Our cohort consisted of 71 CNs from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database who underwent both AV45 amyloid-PET and diffusion tensor imaging. Standardized uptake value ratio (SUVR) was computed across four bilateral regions of interest (ROIs) corresponding to four stages of in vivo amyloid staging model (Amyloid stages I-IV). Linear regression models were conducted in entire CN group and between APOEɛ4 carriers and non-carriers. RESULTS Our results indicated that higher global Aβ-SUVR was associated with higher mean diffusivity (MD) in the entire CN group (p = 0.023), and with both higher MD (p = 0.015) and lower fractional anisotropy (FA) (p = 0.026) in APOEɛ4 carriers. Subregion analysis showed that higher Amyloid stage I-II Aβ-SUVRs were associated with higher MD (Stage-1: p = 0.030; Stage-2: p = 0.016) in the entire CN group, and with both higher MD (Stage-1: p = 0.004; Stage-2: p = 0.010) and lower FA (Stage-1: p = 0.022; Stage-2: p = 0.014) in APOEɛ4 carriers. No associations were found in APOEɛ4 non-carriers and in Amyloid stage III-IV ROIs. CONCLUSIONS Our results indicated that the effects of Aβ burden on WM integrity in CNs might be regionally specific, particularly in Amyloid stage I-II ROIs, and modulated by APOEɛ4 status.
Collapse
Affiliation(s)
- Ya-Juan Wang
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, China
| | - Yu-Xiang Yang
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chuan-Tao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, China.,Department of Neurology, Qingdao Municipal Hospital, Qingdao University, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | | |
Collapse
|
130
|
Li LL, Ma YH, Bi YL, Sun FR, Hu H, Hou XH, Xu W, Shen XN, Dong Q, Tan L, Yang JL, Yu JT. Serum Uric Acid May Aggravate Alzheimer's Disease Risk by Affecting Amyloidosis in Cognitively Intact Older Adults: The CABLE Study. J Alzheimers Dis 2021; 81:389-401. [PMID: 33814427 DOI: 10.3233/jad-201192] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Serum uric acid (SUA) affects the reaction of oxidative stress and free radicals in the neurodegenerative processes. However, whether SUA impacts Alzheimer's disease (AD) pathology remains unclear. OBJECTIVE We aimed to explore whether high SUA levels can aggravate the neurobiological changes of AD in preclinical AD. METHODS We analyzed cognitively intact participants (n = 839, age 62.16 years) who received SUA and cerebrospinal fluid (CSF) biomarkers (amyloid-β [Aβ], total tau [t-Tau], and phosphorylated tau [p-Tau]) measurements from the Chinese Alzheimer's Biomarker and LifestylE (CABLE) database using multivariable-adjusted linear models. RESULTS Levels of SUA in the preclinical AD elevated compared with the healthy controls (p = 0.007) and subjects with amyloid pathology had higher concentration of SUA than controls (p = 0.017). Roughly, equivalent levels of SUA displayed among cognitively intact individuals with or without tau pathology and neurodegeneration. CSF Aβ1 - 42 (p = 0.019) and Aβ1 - 42/Aβ1 - 40 (p = 0.027) were decreased and CSF p-Tau/Aβ1 - 42 (p = 0.009) and t-Tau/Aβ1 - 42 (p = 0.043) were increased with the highest (> 75th percentile) SUA when compared to lowest SUA, implying a high burden of cerebral amyloidosis in individuals with high SUA. Sensitivity analyses using the usual threshold to define hyperuricemia and precluding drug effects yielded robust associations. Nevertheless, the quadratic model did not show any U-shaped relationships between them. CONCLUSION SUA may aggravate brain amyloid deposition in preclinical AD, which corroborated the detrimental role of SUA.
Collapse
Affiliation(s)
- Lin-Lin Li
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan-Lin Bi
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Fu-Rong Sun
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xiao-He Hou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei Xu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xue-Ning Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian, China.,Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jiu-Long Yang
- Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
131
|
Oldan JD, Jewells VL, Pieper B, Wong TZ. Complete Evaluation of Dementia: PET and MRI Correlation and Diagnosis for the Neuroradiologist. AJNR Am J Neuroradiol 2021; 42:998-1007. [PMID: 33926896 DOI: 10.3174/ajnr.a7079] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 11/14/2020] [Indexed: 12/12/2022]
Abstract
This article will familiarize neuroradiologists with the pathophysiology, clinical findings, and standard MR imaging and PET imaging features of multiple forms of dementia as well as new emerging techniques. Cases were compiled from multiple institutions with the goal of improved diagnostic accuracy and improved patient care as well as information about biomarkers on the horizon. Dementia topics addressed include the following: Alzheimer disease, frontotemporal dementia, cerebral amyloid angiopathy, Lewy body dementia, Parkinson disease and Parkinson disease variants, amyotrophic lateral sclerosis, multisystem atrophy, Huntington disease vascular dementia, and Creutzfeldt-Jakob disease.
Collapse
Affiliation(s)
- J D Oldan
- From the Department of Radiology (J.D.O., V.L.J), University of North Carolina, Chapel Hill, North Carolina
| | - V L Jewells
- From the Department of Radiology (J.D.O., V.L.J), University of North Carolina, Chapel Hill, North Carolina
| | - B Pieper
- Department of Radiology (B.P.), Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - T Z Wong
- Department of Radiology (T.Z.W.), Duke University Hospital, Durham, North Carolina
| |
Collapse
|
132
|
Plasminogen Activators in Neurovascular and Neurodegenerative Disorders. Int J Mol Sci 2021; 22:ijms22094380. [PMID: 33922229 PMCID: PMC8122722 DOI: 10.3390/ijms22094380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022] Open
Abstract
The neurovascular unit (NVU) is a dynamic structure assembled by endothelial cells surrounded by a basement membrane, pericytes, astrocytes, microglia and neurons. A carefully coordinated interplay between these cellular and non-cellular components is required to maintain normal neuronal function, and in line with these observations, a growing body of evidence has linked NVU dysfunction to neurodegeneration. Plasminogen activators catalyze the conversion of the zymogen plasminogen into the two-chain protease plasmin, which in turn triggers a plethora of physiological events including wound healing, angiogenesis, cell migration and inflammation. The last four decades of research have revealed that the two mammalian plasminogen activators, tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA), are pivotal regulators of NVU function during physiological and pathological conditions. Here, we will review the most relevant data on their expression and function in the NVU and their role in neurovascular and neurodegenerative disorders.
Collapse
|
133
|
Carli G, Tondo G, Boccalini C, Perani D. Brain Molecular Connectivity in Neurodegenerative Conditions. Brain Sci 2021; 11:brainsci11040433. [PMID: 33800680 PMCID: PMC8067093 DOI: 10.3390/brainsci11040433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/15/2021] [Accepted: 03/23/2021] [Indexed: 12/28/2022] Open
Abstract
Positron emission tomography (PET) allows for the in vivo assessment of early brain functional and molecular changes in neurodegenerative conditions, representing a unique tool in the diagnostic workup. The increased use of multivariate PET imaging analysis approaches has provided the chance to investigate regional molecular processes and long-distance brain circuit functional interactions in the last decade. PET metabolic and neurotransmission connectome can reveal brain region interactions. This review is an overview of concepts and methods for PET molecular and metabolic covariance assessment with evidence in neurodegenerative conditions, including Alzheimer’s disease and Lewy bodies disease spectrum. We highlight the effects of environmental and biological factors on brain network organization. All of the above might contribute to innovative diagnostic tools and potential disease-modifying interventions.
Collapse
Affiliation(s)
- Giulia Carli
- School of Psychology, Vita-Salute San Raffaele University, 20121 Milan, Italy; (G.C.); (G.T.); (C.B.)
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20121 Milan, Italy
| | - Giacomo Tondo
- School of Psychology, Vita-Salute San Raffaele University, 20121 Milan, Italy; (G.C.); (G.T.); (C.B.)
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20121 Milan, Italy
| | - Cecilia Boccalini
- School of Psychology, Vita-Salute San Raffaele University, 20121 Milan, Italy; (G.C.); (G.T.); (C.B.)
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20121 Milan, Italy
| | - Daniela Perani
- School of Psychology, Vita-Salute San Raffaele University, 20121 Milan, Italy; (G.C.); (G.T.); (C.B.)
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20121 Milan, Italy
- Nuclear Medicine Unit, San Raffaele Hospital, 20121 Milan, Italy
- Correspondence: ; Tel.: +39-02-26432224
| |
Collapse
|
134
|
Scheffer S, Hermkens DMA, van der Weerd L, de Vries HE, Daemen MJAP. Vascular Hypothesis of Alzheimer Disease: Topical Review of Mouse Models. Arterioscler Thromb Vasc Biol 2021; 41:1265-1283. [PMID: 33626911 DOI: 10.1161/atvbaha.120.311911] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Sanny Scheffer
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands (S.S., D.M.A.H., M.J.A.P.D.)
| | - Dorien M A Hermkens
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands (S.S., D.M.A.H., M.J.A.P.D.)
| | - Louise van der Weerd
- Departments of Radiology & Human Genetics, Leiden University Medical Center, the Netherlands (L.v.d.W.)
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije University of Amsterdam, the Netherlands (H.E.d.V.)
| | - Mat J A P Daemen
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands (S.S., D.M.A.H., M.J.A.P.D.)
| |
Collapse
|
135
|
Ficiarà E, Boschi S, Ansari S, D'Agata F, Abollino O, Caroppo P, Di Fede G, Indaco A, Rainero I, Guiot C. Machine Learning Profiling of Alzheimer's Disease Patients Based on Current Cerebrospinal Fluid Markers and Iron Content in Biofluids. Front Aging Neurosci 2021; 13:607858. [PMID: 33692679 PMCID: PMC7937894 DOI: 10.3389/fnagi.2021.607858] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by a complex etiology that makes therapeutic strategies still not effective. A true understanding of key pathological mechanisms and new biomarkers are needed, to identify alternative disease-modifying therapies counteracting the disease progression. Iron is an essential element for brain metabolism and its imbalance is implicated in neurodegeneration, due to its potential neurotoxic effect. However, the role of iron in different stages of dementia is not clearly established. This study aimed to investigate the potential impact of iron both in cerebrospinal fluid (CSF) and in serum to improve early diagnosis and the related therapeutic possibility. In addition to standard clinical method to detect iron in serum, a precise quantification of total iron in CSF was performed using graphite-furnace atomic absorption spectrometry in patients affected by AD, mild cognitive impairment, frontotemporal dementia, and non-demented neurological controls. The application of machine learning techniques, such as clustering analysis and multiclassification algorithms, showed a new potential stratification of patients exploiting iron-related data. The results support the involvement of iron dysregulation and its potential interaction with biomarkers (Tau protein and Amyloid-beta) in the pathophysiology and progression of dementia.
Collapse
Affiliation(s)
- Eleonora Ficiarà
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Torino, Italy
| | - Silvia Boschi
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Torino, Italy.,Department NEUROFARBA, University of Firenze, Firenze, Italy
| | - Shoeb Ansari
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Torino, Italy
| | - Federico D'Agata
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Torino, Italy
| | - Ornella Abollino
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Paola Caroppo
- Unit of Neurology 5 and Neuropathology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuseppe Di Fede
- Unit of Neurology 5 and Neuropathology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologico Carlo Besta, Milan, Italy
| | - Antonio Indaco
- Unit of Neurology 5 and Neuropathology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologico Carlo Besta, Milan, Italy
| | - Innocenzo Rainero
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Torino, Italy
| | - Caterina Guiot
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Torino, Italy
| |
Collapse
|
136
|
The Role of Butyrylcholinesterase and Iron in the Regulation of Cholinergic Network and Cognitive Dysfunction in Alzheimer's Disease Pathogenesis. Int J Mol Sci 2021; 22:ijms22042033. [PMID: 33670778 PMCID: PMC7922581 DOI: 10.3390/ijms22042033] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD), the most common form of dementia in elderly individuals, is marked by progressive neuron loss. Despite more than 100 years of research on AD, there is still no treatment to cure or prevent the disease. High levels of amyloid-β (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain are neuropathological hallmarks of AD. However, based on postmortem analyses, up to 44% of individuals have been shown to have high Aβ deposits with no clinical signs, due to having a “cognitive reserve”. The biochemical mechanism explaining the prevention of cognitive impairment in the presence of Aβ plaques is still unknown. It seems that in addition to protein aggregation, neuroinflammatory changes associated with aging are present in AD brains that are correlated with a higher level of brain iron and oxidative stress. It has been shown that iron accumulates around amyloid plaques in AD mouse models and postmortem brain tissues of AD patients. Iron is required for essential brain functions, including oxidative metabolism, myelination, and neurotransmitter synthesis. However, an imbalance in brain iron homeostasis caused by aging underlies many neurodegenerative diseases. It has been proposed that high iron levels trigger an avalanche of events that push the progress of the disease, accelerating cognitive decline. Patients with increased amyloid plaques and iron are highly likely to develop dementia. Our observations indicate that the butyrylcholinesterase (BChE) level seems to be iron-dependent, and reports show that BChE produced by reactive astrocytes can make cognitive functions worse by accelerating the decay of acetylcholine in aging brains. Why, even when there is a genetic risk, do symptoms of the disease appear after many years? Here, we discuss the relationship between genetic factors, age-dependent iron tissue accumulation, and inflammation, focusing on AD.
Collapse
|
137
|
Husain MA, Laurent B, Plourde M. APOE and Alzheimer's Disease: From Lipid Transport to Physiopathology and Therapeutics. Front Neurosci 2021; 15:630502. [PMID: 33679311 PMCID: PMC7925634 DOI: 10.3389/fnins.2021.630502] [Citation(s) in RCA: 141] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/20/2021] [Indexed: 12/23/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder characterized by extracellular amyloid β (Aβ) and intraneuronal tau protein aggregations. One risk factor for developing AD is the APOE gene coding for the apolipoprotein E protein (apoE). Humans have three versions of APOE gene: ε2, ε3, and ε4 allele. Carrying the ε4 allele is an AD risk factor while carrying the ε2 allele is protective. ApoE is a component of lipoprotein particles in the plasma at the periphery, as well as in the cerebrospinal fluid (CSF) and in the interstitial fluid (ISF) of brain parenchyma in the central nervous system (CNS). ApoE is a major lipid transporter that plays a pivotal role in the development, maintenance, and repair of the CNS, and that regulates multiple important signaling pathways. This review will focus on the critical role of apoE in AD pathogenesis and some of the currently apoE-based therapeutics developed in the treatment of AD.
Collapse
Affiliation(s)
- Mohammed Amir Husain
- Centre de Recherche Sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada.,Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Benoit Laurent
- Centre de Recherche Sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada.,Département de Biochimie et Génomique Fonctionnelle, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mélanie Plourde
- Centre de Recherche Sur le Vieillissement, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada.,Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
138
|
Bacterial sepsis increases hippocampal fibrillar amyloid plaque load and neuroinflammation in a mouse model of Alzheimer's disease. Neurobiol Dis 2021; 152:105292. [PMID: 33556539 DOI: 10.1016/j.nbd.2021.105292] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/06/2020] [Accepted: 02/03/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Sepsis, a leading cause for intensive care unit admissions, causes both an acute encephalopathy and chronic brain dysfunction in survivors. A history of sepsis is also a risk factor for future development of dementia symptoms. Similar neuropathologic changes are associated with the cognitive decline of sepsis and Alzheimer's disease (AD), including neuroinflammation, neuronal death, and synaptic loss. Amyloid plaque pathology is the earliest pathological hallmark of AD, appearing 10 to 20 years prior to cognitive decline, and is present in 30% of people over 65. As sepsis is also more common in older adults, we hypothesized that sepsis might exacerbate amyloid plaque deposition and plaque-related injury, promoting the progression of AD-related pathology. METHODS We evaluated whether the brain's response to sepsis modulates AD-related neurodegenerative changes by driving amyloid deposition and neuroinflammation in mice. We induced polymicrobial sepsis by cecal ligation and puncture (CLP) in APP/PS1-21 mice, a model of AD-related β-amyloidosis. We performed CLP or sham surgery at plaque onset (2 months of age) and examined pathology 2 months after CLP in surviving mice. RESULTS Sepsis significantly increased fibrillar amyloid plaque formation in the hippocampus of APP/PS1-21 mice. Sepsis enhanced plaque-related astrocyte activation and complement C4b gene expression in the brain, both of which may play a role in modulating amyloid formation. CLP also caused large scale changes in the gut microbiome of APP/PS1 mice, which have been associated with a pro-amyloidogenic and neuroinflammatory state. CONCLUSIONS Our results suggest that experimental sepsis can exacerbate amyloid plaque deposition and plaque-related inflammation, providing a potential mechanism for increased dementia in older sepsis survivors.
Collapse
|
139
|
Kabir MT, Uddin MS, Mathew B, Das PK, Perveen A, Ashraf GM. Emerging Promise of Immunotherapy for Alzheimer's Disease: A New Hope for the Development of Alzheimer's Vaccine. Curr Top Med Chem 2021; 20:1214-1234. [PMID: 32321405 DOI: 10.2174/1568026620666200422105156] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurodegenerative disorder and the characteristics of this devastating disorder include the progressive and disabling deficits in the cognitive functions including reasoning, attention, judgment, comprehension, memory, and language. OBJECTIVE In this article, we have focused on the recent progress that has been achieved in the development of an effective AD vaccine. SUMMARY Currently, available treatment options of AD are limited to deliver short-term symptomatic relief only. A number of strategies targeting amyloid-beta (Aβ) have been developed in order to treat or prevent AD. In order to exert an effective immune response, an AD vaccine should contain adjuvants that can induce an effective anti-inflammatory T helper 2 (Th2) immune response. AD vaccines should also possess the immunogens which have the capacity to stimulate a protective immune response against various cytotoxic Aβ conformers. The induction of an effective vaccine's immune response would necessitate the parallel delivery of immunogen to dendritic cells (DCs) and their priming to stimulate a Th2-polarized response. The aforesaid immune response is likely to mediate the generation of neutralizing antibodies against the neurotoxic Aβ oligomers (AβOs) and also anti-inflammatory cytokines, thus preventing the AD-related inflammation. CONCLUSION Since there is an age-related decline in the immune functions, therefore vaccines are more likely to prevent AD instead of providing treatment. AD vaccines might be an effective and convenient approach to avoid the treatment-related huge expense.
Collapse
Affiliation(s)
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | | | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
140
|
Leblhuber F, Ehrlich D, Steiner K, Geisler S, Fuchs D, Lanser L, Kurz K. The Immunopathogenesis of Alzheimer's Disease Is Related to the Composition of Gut Microbiota. Nutrients 2021; 13:361. [PMID: 33504065 PMCID: PMC7912578 DOI: 10.3390/nu13020361] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 12/11/2022] Open
Abstract
The microbiota-gut-brain axis plays an important role in the development of neurodegenerative diseases. Commensal and pathogenic enteric bacteria can influence brain and immune system function by the production of lipopolysaccharides and amyloid. Dysbiosis of the intestinal microbiome induces local and consecutively systemic immune-mediated inflammation. Proinflammatory cytokines then trigger neuroinflammation and finally neurodegeneration. Immune-mediated oxidative stress can lead to a deficiency of vitamins and essential micronutrients. Furthermore, the wrong composition of gut microbiota might impair the intake and metabolization of nutrients. In patients with Alzheimer's disease (AD) significant alterations of the gut microbiota have been demonstrated. Standard Western diet, infections, decreased physical activity and chronic stress impact the composition and diversity of gut microbiota. A higher abundancy of "pro-inflammatory" gut microbiota goes along with enhanced systemic inflammation and neuroinflammatory processes. Thus, AD beginning in the gut is closely related to the imbalance of gut microbiota. Modulation of gut microbiota by Mediterranean diet, probiotics and curcumin can slow down cognitive decline and alter the gut microbiome significantly. A multi-domain intervention approach addressing underlying causes of AD (inflammation, infections, metabolic alterations like insulin resistance and nutrient deficiency, stress) appears very promising to reduce or even reverse cognitive decline by exerting positive effects on the gut microbiota.
Collapse
Affiliation(s)
- Friedrich Leblhuber
- Department of Gerontology, Neuromed Campus, Kepler University Clinic, Linz A-4020, Austria; (F.L.); (D.E.); (K.S.)
| | - Daniela Ehrlich
- Department of Gerontology, Neuromed Campus, Kepler University Clinic, Linz A-4020, Austria; (F.L.); (D.E.); (K.S.)
| | - Kostja Steiner
- Department of Gerontology, Neuromed Campus, Kepler University Clinic, Linz A-4020, Austria; (F.L.); (D.E.); (K.S.)
| | - Simon Geisler
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck A-6020, Austria; (S.G.); (D.F.)
| | - Dietmar Fuchs
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck A-6020, Austria; (S.G.); (D.F.)
| | - Lukas Lanser
- Department of Internal Medicine, Medical University of Innsbruck, Innsbruck A-6020, Austria;
| | - Katharina Kurz
- Department of Internal Medicine, Medical University of Innsbruck, Innsbruck A-6020, Austria;
| |
Collapse
|
141
|
Abstract
This article presents an overview of imaging agents for PET that have been applied for research and diagnostic purposes in patients affected by dementia. Classified by the target which the agents visualize, seven groups of tracers can be distinguished, namely radiopharmaceuticals for: (1) Misfolded proteins (ß-amyloid, tau, α-synuclein), (2) Neuroinflammation (overexpression of translocator protein), (3) Elements of the cholinergic system, (4) Elements of monoamine neurotransmitter systems, (5) Synaptic density, (6) Cerebral energy metabolism (glucose transport/ hexokinase), and (7) Various other proteins. This last category contains proteins involved in mechanisms underlying neuroinflammation or cognitive impairment, which may also be potential therapeutic targets. Many receptors belong to this category: AMPA, cannabinoid, colony stimulating factor 1, metabotropic glutamate receptor 1 and 5 (mGluR1, mGluR5), opioid (kappa, mu), purinergic (P2X7, P2Y12), sigma-1, sigma-2, receptor for advanced glycation endproducts, and triggering receptor expressed on myeloid cells-1, besides several enzymes: cyclooxygenase-1 and 2 (COX-1, COX-2), phosphodiesterase-5 and 10 (PDE5, PDE10), and tropomyosin receptor kinase. Significant advances in neuroimaging have been made in the last 15 years. The use of 2-[18F]-fluoro-2-deoxy-D-glucose (FDG) for quantification of regional cerebral glucose metabolism is well-established. Three tracers for ß-amyloid plaques have been approved by the Food and Drug Administration and European Medicines Agency. Several tracers for tau neurofibrillary tangles are already applied in clinical research. Since many novel agents are in the preclinical or experimental stage of development, further advances in nuclear medicine imaging can be expected in the near future. PET studies with established tracers and tracers for novel targets may result in early diagnosis and better classification of neurodegenerative disorders and in accurate monitoring of therapy trials which involve these targets. PET data have prognostic value and may be used to assess the response of the human brain to interventions, or to select the appropriate treatment strategy for an individual patient.
Collapse
Affiliation(s)
- Aren van Waarde
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands.
| | - Sofia Marcolini
- University of Groningen, University Medical Center Groningen, Department of Neurology, Groningen, the Netherlands
| | - Peter Paul de Deyn
- University of Groningen, University Medical Center Groningen, Department of Neurology, Groningen, the Netherlands; University of Antwerp, Born-Bunge Institute, Neurochemistry and Behavior, Campus Drie Eiken, Wilrijk, Belgium
| | - Rudi A J O Dierckx
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands; Ghent University, Ghent, Belgium
| |
Collapse
|
142
|
Mifflin MA, Winslow W, Surendra L, Tallino S, Vural A, Velazquez R. Sex differences in the IntelliCage and the Morris water maze in the APP/PS1 mouse model of amyloidosis. Neurobiol Aging 2021; 101:130-140. [PMID: 33610962 PMCID: PMC8122060 DOI: 10.1016/j.neurobiolaging.2021.01.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 11/11/2020] [Accepted: 01/18/2021] [Indexed: 12/05/2022]
Abstract
Transgenic rodent models were created to decipher pathogenic mechanisms associated with Alzheimer’s disease (AD), and behavioral apparatuses such as the Morris water maze (MWM) are used to assess cognition in mice. The IntelliCage was designed to circumvent issues of traditional behavioral tests, such as frequent human handling. The motivation to complete IntelliCage tasks is water consumption, which is less stressful than escaping from a pool in the MWM. Here, we examined behavioral performances of mice in the IntelliCage and MWM tasks. Twelve-month-old male and female APP/PS1 and non-transgenic mice first underwent 42 days of IntelliCage testing to assess prefrontal cortical and hippocampal function followed by MWM testing for six days. We found that females performed better in the IntelliCage while males performed superiorly in the MWM. Mechanistically, female APP/PS1 mice had a higher Amyloid-β plaque load throughout the brain, which is inconsistent with their performance in the IntelliCage. Collectively, these results inform scientists about the sex-based differences when testing animals in different behavioral paradigms that tap similar cognitive functions.
Collapse
Affiliation(s)
- Marc A Mifflin
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Wendy Winslow
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Likith Surendra
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Savannah Tallino
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Austin Vural
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Ramon Velazquez
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
143
|
Zhou K, Yang F, Li Y, Chen Y, Zhang X, Zhang J, Wang J, Dai J, Cai L, Cui M. Synthesis and Evaluation of Fluorine-18 Labeled 2-Phenylquinoxaline Derivatives as Potential Tau Imaging Agents. Mol Pharm 2021; 18:1176-1195. [PMID: 33475377 DOI: 10.1021/acs.molpharmaceut.0c01078] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this study, three pairs of optically pure 18F-labeled 2-phenylquinoxaline derivatives were evaluated as Tau imaging agents for the diagnosis of Alzheimer's disease (AD). The chiral 2-fluoromethyl-1,2-ethylenediol side chain was attached to the 2-phenylquinoxaline backbone to increase hydrophilicity, thereby improving the binding affinity of the probe to tangles and their selectivity toward Tau tangles over β-amyloid plaques (Aβ). These probes displayed excellent fluorescent properties and high selectivity for tangles on brain sections from transgenic mice (rTg4510) and AD patients. Quantitative binding assays with AD homogenates showed that the probes (R)-5 and (S)-16 have a high affinity (Ki = 4.1 and 10.3 nM, respectively) and high selectivity (30.5-fold and 34.6-fold, respectively) for tangles over Aβ. The high affinity and selectivity of (R)-[18F]5 and (S)-[18F]16 for tangles were further confirmed with autoradiography on AD brain tissue in vitro. In addition, they displayed sufficient blood-brain barrier penetration (7.06% and 10.95% ID/g, respectively) and suitable brain kinetics (brain2 min/brain60 min = 10.1, 6.5 respectively) in normal mice. Ex vivo metabolism studies and micro-positron emission computed tomography (PET) revealed high brain biostability, good brain kinetic properties, and low nonspecific binding for (S)-[18F]16. Together, these results demonstrate that (R)-[18F]5 and (S)-[18F]16 are promising PET probes for Tau tangles imaging.
Collapse
Affiliation(s)
- Kaixiang Zhou
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing 100875, China
| | - Fan Yang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing 100875, China
| | - Yuying Li
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing 100875, China
| | - Yimin Chen
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing 100875, China
| | - Xiaojun Zhang
- Department of Nuclear Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Jinming Zhang
- Department of Nuclear Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Junfeng Wang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Charlestown 02129, Massachusetts, United States
| | - Jiapei Dai
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan 430074, China
| | - Lisheng Cai
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda 20892, Maryland, United States
| | - Mengchao Cui
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
144
|
Abstract
Amyloid-β (Aβ) PET imaging has now been available for over 15 years. The ability to detect Aβ in vivo has greatly improved the clinical and research landscape of Alzheimer's disease (AD) and other neurodegenerative conditions. Aβ imaging provides very reliable, accurate, and reproducible measurements of regional and global Aβ burden in the brain. It has proved invaluable in anti-Aβ therapy trials, and is now recognized as a powerful diagnostic tool. The appropriate use of Aβ PET, when combined with comprehensive clinical evaluation by a dementia-trained specialist, can improve the accuracy of a clinical diagnosis of AD and substantially alter management. It can assist in differentiating AD from other neurodegenerative conditions, often by its ability to rule out the presence of Aβ. When combined with tau imaging, further increase in specificity for the diagnosis of AD can be achieved. The integration of Aβ PET, in conjunction with biomarkers of tau, neurodegeneration and neuroinflammation, into large, longitudinal, observational cohort studies continues to increase our understanding of the development of AD. Its incorporation into clinical trials has been pivotal in defining the most effective anti-Aβ biological therapies and optimal dosing so that effective disease modifying therapy now appears imminent. Aβ deposition is a gradual and protracted process, permitting a wide treatment window for anti-Aβ therapies and Aβ PET has made trials in this preclinical AD period feasible. Continuing improvement in Aβ tracer target to background ratio is allowing trials in earlier AD that tailor drug dosage to Aβ level. The quest to standardize quantification and define universally applicable thresholds for all Aβ tracers has produced the Centiloid method. Centiloid values that correlate well with neuropathologic findings and prognosis have been identified. Rapid cloud-based automated individual scan analysis is now possible and does not require MRI. Challenges remain, particularly around cross camera standardized uptake value ratio variation that need to be addressed. This review will compare available Aβ radiotracers, discuss approaches to quantification, as well as the clinical and research applications of Aβ PET.
Collapse
Affiliation(s)
- Natasha Krishnadas
- Florey Department of Neurosciences and Mental Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Victoria, Australia; Department of Molecular Imaging & Therapy, Austin Health, Victoria, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Victoria, Australia
| | - Vincent Doré
- Department of Molecular Imaging & Therapy, Austin Health, Victoria, Australia; Health and Biosecurity Flagship, The Australian eHealth Research Centre, CSIRO, Victoria, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Victoria, Australia; The Australian Dementia Network (ADNeT), Melbourne, Australia; The University of Melbourne, Victoria, Australia.
| |
Collapse
|
145
|
Xie Y, Yang Q, Liu C, Zhang Q, Jiang J, Han Y. Exploring the Pattern Associated With Longitudinal Changes of β-Amyloid Deposition During Cognitively Normal Healthy Aging. Front Med (Lausanne) 2021; 7:617173. [PMID: 33585514 PMCID: PMC7874155 DOI: 10.3389/fmed.2020.617173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022] Open
Abstract
The aim of this study was to determine a pattern associated with longitudinal changes of β-amyloid (Aβ) deposition during cognitively normal(CN) healthy aging. We used 18F-florbetapir (AV-45) PET images of the brains of 207 cognitively normal subjects (CN1), obtained through the Alzheimer's Disease Neuroimaging Initiative (ADNI), to identify the healthy aging pattern and 76 cognitively normal healthy subjects (CN2), obtained through the Xuanwu Hospital of Capital Medical University, Beijing, China, to verify it. A voxel-based correlation analysis of standardized uptake value ratio (SUVR) map image and age was conducted using the DPABI (Data Processing & Analysis of Brain Imaging) software to identify the pattern. The sum of squares due to errors (SSE), R-square (R2) and the root-mean-square error (RMSE) were calculated to assess the quality of curve fitting. Among them, R2 was proposed as the coherence coefficient, which was as an index to assess the correlation between SUVR value of the pattern and subjects' age. The pattern characterized by age-associated longitudinal changes of Aβ deposition was mainly distributed in the right middle and inferior temporal gyrus, the right temporal pole: middle temporal gyrus, the right inferior occipital gyrus, the right inferior frontal gyrus (triangular portion), and the right precentral gyrus. There were a significant positive correlation between the SUVR value of the pattern and age for each CN group (CN1: R2 = 0.120, p < 0.001 for quadratic model; CN2: R2 = 0.152, p = 0.002 for quadratic model). These findings suggest a pattern of changes in Aβ deposition that can be used to distinguish physiological changes from pathophysiological changes, constituting a new method for elucidating the neuropathological mechanism of Alzheimer's disease.
Collapse
Affiliation(s)
- Yunyan Xie
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Qin Yang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Chunhua Liu
- Key Laboratory of Specialty Fiber Optics and Optical Access Networks, Joint International Research Laboratory of Specialty Fiber Optics and Advanced Communication, School of Communication and Information Technology, Shanghai University, Shanghai, China
| | - Qi Zhang
- Key Laboratory of Specialty Fiber Optics and Optical Access Networks, Joint International Research Laboratory of Specialty Fiber Optics and Advanced Communication, School of Communication and Information Technology, Shanghai University, Shanghai, China
| | - Jiehui Jiang
- Key Laboratory of Specialty Fiber Optics and Optical Access Networks, Joint International Research Laboratory of Specialty Fiber Optics and Advanced Communication, School of Communication and Information Technology, Shanghai University, Shanghai, China.,Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China.,Institute of Biomedical Engineering, Shanghai University, Shanghai, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| | | |
Collapse
|
146
|
Rådestig MA, Skoog J, Zetterberg H, Kern J, Zettergren A, Sacuiu S, Waern M, Wetterberg H, Blennow K, Skoog I, Kern S. Cognitive Performance and Cerebrospinal Fluid Markers in Preclinical Alzheimer's Disease: Results from the Gothenburg H70 Birth Cohort Studies. J Alzheimers Dis 2021; 79:225-235. [PMID: 33216028 DOI: 10.3233/jad-200751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND We have previously shown that older adults with preclinical Alzheimer's disease (AD) pathology in cerebrospinal fluid (CSF) had slightly worse performance in Mini-Mental State Examination (MMSE) than participants without preclinical AD pathology. OBJECTIVE We therefore aimed to compare performance on neurocognitive tests in a population-based sample of 70-year-olds with and without CSF AD pathology. METHODS The sample was derived from the population-based Gothenburg H70 Birth Cohort Studies in Sweden. Participants (n = 316, 70 years old) underwent comprehensive cognitive examinations, and CSF Aβ-42, Aβ-40, T-tau, and P-tau concentrations were measured. Participants were classified according to the ATN system, and according to their Clinical Dementia Rating (CDR) score. Cognitive performance was examined in the CSF amyloid, tau, and neurodegeneration (ATN) categories. RESULTS Among participants with CDR 0 (n = 259), those with amyloid (A+) and/or tau pathology (T+, N+) showed similar performance on most cognitive tests compared to participants with A-T-N-. Participants with A-T-N+ performed worse in memory (Supra span (p = 0.003), object Delayed (p = 0.042) and Immediate recall (p = 0.033)). Among participants with CDR 0.5 (n = 57), those with amyloid pathology (A+) scored worse in category fluency (p = 0.003). CONCLUSION Cognitively normal participants with amyloid and/or tau pathology performed similarly to those without any biomarker evidence of preclinical AD in most cognitive domains, with the exception of slightly poorer memory performance in A-T-N+. Our study suggests that preclinical AD biomarkers are altered before cognitive decline.
Collapse
Affiliation(s)
- Maya Arvidsson Rådestig
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Johan Skoog
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychology, University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, 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.,UCL Institute of Neurology (H.Z.), Queen Square, London, United Kingdom.,The UK Dementia Research Institute at UCL, London, United Kingdom
| | - Jürgen Kern
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Anna Zettergren
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Simona Sacuiu
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Margda Waern
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Hanna Wetterberg
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - 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
| | - Ingmar Skoog
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Silke Kern
- Center for Ageing and Health (AgeCap) at the University of Gothenburg, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| |
Collapse
|
147
|
Shaffer RM, Li G, Adar SD, Dirk Keene C, Latimer CS, Crane PK, Larson EB, Kaufman JD, Carone M, Sheppard L. Fine Particulate Matter and Markers of Alzheimer's Disease Neuropathology at Autopsy in a Community-Based Cohort. J Alzheimers Dis 2021; 79:1761-1773. [PMID: 33459717 PMCID: PMC8061707 DOI: 10.3233/jad-201005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Evidence links fine particulate matter (PM2.5) to Alzheimer's disease (AD), but no community-based prospective cohort studies in older adults have evaluated the association between long-term exposure to PM2.5 and markers of AD neuropathology at autopsy. OBJECTIVE Using a well-established autopsy cohort and new spatiotemporal predictions of air pollution, we evaluated associations of 10-year PM2.5 exposure prior to death with Braak stage, Consortium to Establish a Registry for AD (CERAD) score, and combined AD neuropathologic change (ABC score). METHODS We used autopsy specimens (N = 832) from the Adult Changes in Thought (ACT) study, with enrollment ongoing since 1994. We assigned long-term exposure at residential address based on two-week average concentrations from a newly developed spatiotemporal model. To account for potential selection bias, we conducted inverse probability weighting. Adjusting for covariates with tiered models, we performed ordinal regression for Braak and CERAD and logistic regression for dichotomized ABC score. RESULTS 10-year average (SD) PM2.5 from death across the autopsy cohort was 8.2 (1.9) μg/m3. Average age (SD) at death was 89 (7) years. Each 1μg/m3 increase in 10-year average PM2.5 prior to death was associated with a suggestive increase in the odds of worse neuropathology as indicated by CERAD score (OR: 1.35 (0.90, 1.90)) but a suggestive decreased odds of neuropathology as defined by the ABC score (OR: 0.79 (0.49, 1.19)). There was no association with Braak stage (OR: 0.99 (0.64, 1.47)). CONCLUSION We report inconclusive associations between PM2.5 and AD neuropathology at autopsy among a cohort where 94% of individuals experienced 10-year exposures below the current EPA standard. Prior studies of AD risk factors and AD neuropathology are similarly inconclusive, suggesting alternative mechanistic pathways for disease or residual confounding.
Collapse
Affiliation(s)
- Rachel M. Shaffer
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, USA
| | - Ge Li
- VA Northwest Network Mental Illness Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Sara D. Adar
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - C. Dirk Keene
- Division of Neuropathology, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Caitlin S. Latimer
- Division of Neuropathology, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Paul K. Crane
- School of Medicine, University of Washington, Seattle, WA, USA
| | - Eric B. Larson
- School of Medicine, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Joel D. Kaufman
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, USA
- Departments of Medicine and Epidemiology, University of Washington School of Public Health, Seattle, WA, USA
| | - Marco Carone
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| | - Lianne Sheppard
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, USA
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| |
Collapse
|
148
|
Xing Z, Zuo Z, Hu D, Zheng X, Wang X, Yuan L, Zhou L, Qi F, Yao Z. Influenza vaccine combined with moderate-dose PD1 blockade reduces amyloid-β accumulation and improves cognition in APP/PS1 mice. Brain Behav Immun 2021; 91:128-141. [PMID: 32956831 DOI: 10.1016/j.bbi.2020.09.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022] Open
Abstract
Immune dysfunction is implicated in Alzheimer's disease (AD), whereas systemic immune modulation may be neuroprotective. Our previous results have indicated immune challenge with Bacillus Calmette-Guerin attenuates AD pathology in animal models by boosting the systemic immune system. Similarly, independent studies have shown that boosting systemic immune system, by blocking PD-1 checkpoint pathway, modifies AD. Here we hypothesized that influenza vaccine would potentiate function of moderate dose anti-PD-1 and therefore combining them might allow reducing the dose of PD-1 antibody needed to modify the disease. We found that moderate-dose PD-1 in combination with influenza vaccine effectively attenuated cognitive deficit and prevented amyloid-β pathology build-up in APP/PS1 mice in a mechanism dependent on recruitment of peripheral monocyte-derived macrophages into the brain. Eliminating peripheral macrophages abrogated the beneficial effect. Moreover, by comparing CD11b+ compartments in the mouse parenchyma, we observed an elevated subset of Ly6C+ microglia-like cells, which are reportedly derived from peripheral monocytes. In addition, myeloid-derived suppressor cells are strongly elevated in the transgenic model used and normalized by combination treatment, indicating restoration of brain immune homeostasis. Overall, our results suggest that revitalizing brain immunity by combining IV with moderate-dose PD-1 inhibition may represent a therapeutic immunotherapy for AD.
Collapse
Affiliation(s)
- Zhiwei Xing
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Zejie Zuo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Dandan Hu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, PR China
| | - Xiaona Zheng
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Xiao Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Lifang Yuan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Lihua Zhou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Department of Anatomy, Sun Yat-sen University, School of Medicine, Guangzhou 510089, PR China.
| | - Fangfang Qi
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China.
| | - Zhibin Yao
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China.
| |
Collapse
|
149
|
de Vidania S, Palomares-Perez I, Frank-García A, Saito T, Saido TC, Draffin J, Szaruga M, Chávez-Gutierrez L, Calero M, Medina M, Guix FX, Dotti CG. Prodromal Alzheimer's Disease: Constitutive Upregulation of Neuroglobin Prevents the Initiation of Alzheimer's Pathology. Front Neurosci 2020; 14:562581. [PMID: 33343276 PMCID: PMC7744294 DOI: 10.3389/fnins.2020.562581] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/28/2020] [Indexed: 12/17/2022] Open
Abstract
In humans, a considerable number of the autopsy samples of cognitively normal individuals aged between 57 and 102 years have revealed the presence of amyloid plaques, one of the typical signs of AD, indicating that many of us use mechanisms that defend ourselves from the toxic consequences of Aß. The human APP NL/F (hAPP NL/F) knockin mouse appears as the ideal mouse model to identify these mechanisms, since they have high Aß42 levels at an early age and moderate signs of disease when old. Here we show that in these mice, the brain levels of the hemoprotein Neuroglobin (Ngb) increase with age, in parallel with the increase in Aß42. In vitro, in wild type neurons, exogenous Aß increases the expression of Ngb and Ngb over-expression prevents Aß toxicity. In vivo, in old hAPP NL/F mice, Ngb knockdown leads to dendritic tree simplification, an early sign of Alzheimer’s disease. These results could indicate that Alzheimer’s symptoms may start developing at the time when defense mechanisms start wearing out. In agreement, analysis of plasma Ngb levels in aged individuals revealed decreased levels in those whose cognitive abilities worsened during a 5-year longitudinal follow-up period.
Collapse
Affiliation(s)
- Silvia de Vidania
- Molecular Neuropathology, Physiological and Pathological Processes, Centro de Biología Molecular Severo Ochoa, CSIC/UAM, Madrid, Spain
| | - Irene Palomares-Perez
- Molecular Neuropathology, Physiological and Pathological Processes, Centro de Biología Molecular Severo Ochoa, CSIC/UAM, Madrid, Spain
| | - Ana Frank-García
- Department of Neurology, Instituto de Salud Carlos III (ISCIII), Division Neurodegenerative Disease, University Hospital La Paz, Madrid, Spain
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi, Japan
| | - Takaomi C Saido
- Department of Neurocognitive Science, Nagoya City University Graduate School of Medical Science, Nagoya, Japan
| | - Jonathan Draffin
- Molecular Neuropathology, Physiological and Pathological Processes, Centro de Biología Molecular Severo Ochoa, CSIC/UAM, Madrid, Spain
| | - María Szaruga
- KU Leuven Department for Neurosciences, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| | - Lucía Chávez-Gutierrez
- KU Leuven Department for Neurosciences, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| | - Miguel Calero
- CIBERNED, Queen Sofia Foundation Alzheimer Center, CIEN Foundation, Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel Medina
- CIBERNED, Queen Sofia Foundation Alzheimer Center, CIEN Foundation, Instituto de Salud Carlos III, Madrid, Spain
| | - Francesc X Guix
- Molecular Neuropathology, Physiological and Pathological Processes, Centro de Biología Molecular Severo Ochoa, CSIC/UAM, Madrid, Spain
| | - Carlos G Dotti
- Molecular Neuropathology, Physiological and Pathological Processes, Centro de Biología Molecular Severo Ochoa, CSIC/UAM, Madrid, Spain
| |
Collapse
|
150
|
Novel PET Biomarkers to Disentangle Molecular Pathways across Age-Related Neurodegenerative Diseases. Cells 2020; 9:cells9122581. [PMID: 33276490 PMCID: PMC7761606 DOI: 10.3390/cells9122581] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/25/2020] [Accepted: 11/28/2020] [Indexed: 12/11/2022] Open
Abstract
There is a need to disentangle the etiological puzzle of age-related neurodegenerative diseases, whose clinical phenotypes arise from known, and as yet unknown, pathways that can act distinctly or in concert. Enhanced sub-phenotyping and the identification of in vivo biomarker-driven signature profiles could improve the stratification of patients into clinical trials and, potentially, help to drive the treatment landscape towards the precision medicine paradigm. The rapidly growing field of neuroimaging offers valuable tools to investigate disease pathophysiology and molecular pathways in humans, with the potential to capture the whole disease course starting from preclinical stages. Positron emission tomography (PET) combines the advantages of a versatile imaging technique with the ability to quantify, to nanomolar sensitivity, molecular targets in vivo. This review will discuss current research and available imaging biomarkers evaluating dysregulation of the main molecular pathways across age-related neurodegenerative diseases. The molecular pathways focused on in this review involve mitochondrial dysfunction and energy dysregulation; neuroinflammation; protein misfolding; aggregation and the concepts of pathobiology, synaptic dysfunction, neurotransmitter dysregulation and dysfunction of the glymphatic system. The use of PET imaging to dissect these molecular pathways and the potential to aid sub-phenotyping will be discussed, with a focus on novel PET biomarkers.
Collapse
|